Title of Invention

2-METHYLMORPHOLINE PYRIDO-, PYRAZO-AND PYRIMIDO-PYRIMIDINE DERIVATIVES AS MTOR INHIBITORS

Abstract There is provided a compound of Formula (I), or a pharmaceutically acceptable salt thereof. There are also provided processes for the manufacture of a compound of Formula (1), and the use of a compound of Formula (1) as a medicament and in the treatment of cancer.
Full Text FORM 2
THE PATENTS ACT, 1970
(39 of 1970)
&
THE PATENTS RULES, 2003
COMPLETE SPECIFICATION
(See section 10, rule 13)
"2-METHYLMORPHOLINE PYRIDO-, PYRAZO-AND PYRIMIDO-PYRIMIDINE DERIVATIVES AS
mTOR INHIBITORS"

KuDOS PHARMACEUTICALS LIMITED, of 15 Stanhope Gate London, W1K 1LN, United Kingdom.
The following specification particularly describes the invention and the manner in which it is to be performed.


WO 2008/023161

PCT/GB2n(l 7/003179

2-METHYLMORPHOLINE PYRIDO-,PYRAZO- AND PYRIMIDO-PYRIMIDINE DERIVATIVES AS MTOR INHIBITORS
The present invention relates to compounds which act as mTOR inhibitors, their use
and their synthesis.
Background
5 Growth factor/mitogenic activation of the phosphatidylinositol 3-kinase (PI3K)/AKT
signalling pathway ultimately leads to the key cell cycle and growth control regulator mTOR, the mammalian target of rapamycin (alternatively referred to as FRAP (FKBP12 and rapamycin associated protein), RAFT1 (rapamycin and FKBP12 target 1), RAPT1 (rapamycin target 1) - all derived from the interaction with the FK-506-binding protein FKBP12, and SEP
10 (sirolimus effector protein)). mTOR is a mammalian serine/threonine kinase of approximately 289 kDa in size and a member of the evolutionary conserved eukaryotic TOR kinases (refs. 1-4). The mTOR protein is a member of the PB-kinase like kinase (PUCK) family of proteins due to its C-terminal homology (catalytic domain) with PI3-kinase and the other family members, e.g. DNA-PKcs (DNA dependent protein kinase), ATM (Ataxia-
15 telangiectasia mutated). In addition to a catalytic domain in the C-terminus, mTOR contains a FKBP12/rapamycin complex binding domain (FRB). At the N-terminus up to 20 HEAT (Huntingtin, EF3, alpha regulatory subunit of PP2A and TOR.) motifs are found whilst more C-terminal is a FAT (FRAP-ATM-TRRAP) domain, and at the extreme C-terminus of the protein an additional FAT domain is found (FAT-C) (refs. 5,6).
20 TOR has been identified as a central regulator of both cell growth (size) and
proliferation, which is in part governed by translation initiation. TOR dependant phosphorylation of S6-kinase (S6K1) allows translation of ribosomal proteins involved in cell cycle progression (refs. 7-9). Cap-dependant translation is regulated by the phosphorylation of the eukaryotic translation initiation factor 4E (eIF4E)-binding protein 1 (4E-BP1 (PHAS-
25 1)). This modification prevents PHAS-1 binding eIF4E, thereby permitting formation of an active eIF4F translation complex (reviewed in refs. 10,11,12). Activation of these signalling elements is dependant on insulin, other growth factors and nutrients suggesting a gatekeeper role for mTOR in the control of cell cycle progression only under favourable environmental conditions. The PI3K/AKT signalling cascade lies upstream of mTOR and this has been
30 shown to be deregulated in certain cancers and results in growth factor independent activation in, for example, PTEN deficient cells. mTOR lies at the axis of control for this pathway and inhibitors of this kinase (e.g. sirolimus (rapamycin or Rapamune™) and everolimus (RAD001

WO 2008/023161

PCT/GB2OO7/003179

or Certican ' )) are already approved for immunosuppression and drug eluting stents (reviewed in refs. 13f 14), and are now receiving particular interest as novel agents for cancer treatment.
Tumour cell growth arises from the deregulation of normal growth control 5 mechanisms such as the loss of tumour suppressor function(s). One such tumour suppressor is the phosphatase and tensin homologue deleted from chromosome ten (PTEN). This gene, also known as mutated in multiple advanced cancers (MMAC), has been shown to play a significant role in cell cycle arrest and is the most highly mutated tumour suppressor after p53. Up to 30% of glioblastoma, endometrial and prostate cancers have somatic mutations or
10 deletions of this locus (refs. 15,16).
PI3K converts phosphatidylinositol 4,5, bisphosphate (PIP2) to phosphatidylinositol 3,4,5, triphosphate (PIP3) whilst PTEN is responsible for removing the 33 phosphate from PEP3 producing PTP2. PI3-K and PTEN act to maintain an appropriate level of PIP3 which recruits and thus activates AKT (also known as PKB) and the downstream signalling cascade
15 that is then initiated. In the absence of PTEN, there is inappropriate regulation of this cascade, AKT becomes effectively constitutively activated and cell growth is deregulated. An alternative mechanism for the deregulation of this cell signalling process is the recent identification of a mutant form of the PI3K isoform, pllOalpha (ref. 17). The apparent increased activity of this mutant is thought to result in increased PIP3 production, presumably
20 in excess of that which the function of PTEN can counteract. Increased signalling from PI3K, thus results in increased signalling to mTOR and consequently, its downstream activators.
In addition to the evidence linking mTOR with cell cycle regulation (from Gl to S-phase) and that inhibition of mTOR results in inhibition of these regulatory events it has been shown that down regulation of mTOR activity results in cell growth inhibition (Reviewed in
25 refs. 7,18,19). The known inhibitor of mTOR, rapamycin, potently inhibits proliferation or growth of cells derived from a range of tissue types such as smooth muscle, T-cells as well as cells derived from a diverse range of tumour types including rhabdomyosarcoma, neuroblastoma, glioblastoma and meduUoblastoma, small cell lung cancer, osteosarcoma, pancreatic carcinoma and breast and prostate carcinoma (reviewed in ref. 20). Rapamycin has
30 been approved and is in clinical use as an immunosuppressant, its prevention of organ rejection being successful and with fewer side effects than previous therapies (refs. 20, 21). Inhibition of mTOR by rapamycin and its analogues (RAD001, CCI-779) is brought about by

WO 2008/023161

PCT/GB2007/003I79

the prior interaction of the drug with the FK506 binding protein, FKBP12. Subsequently, the complex of FKBP12/rapamycin then binds to the FRB domain of mTOR and inhibits the downstream signalling from mTOR.
The potent but non-specific inhibitors of PI3K, LY294002 and wortmannin, also have 5 been shown to inhibit the kinase function of mTOR but act through targeting the catalytic domain of the protein (ref 21). Further to the inhibition of mTOR function by small molecules targeted to the kinase domain, it has been demonstrated that kinase dead mTOR cannot transmit the upstream activating signals to the downstream effectors of mTOR, PHAS-1 or p70S6 kinase (ref. 22). It is also shown that not all functions of mTOR are rapamycin
10 sensitive and this may be related to the observation that rapamycin alters the substrate profile of mTOR rather than inhibiting its activity per se (ref. 23). Analysis of the interactions of mTOR with other cellular factors has revealed that in addition to the mTOR-Raptor complex, there is also an mTOR-Rictor complex representing a rapamycin insensitive activity of mTOR (B) (Sarbassov et al Current Biology, 2004, 14, 1296-1302). This activity likely accounts for
15 the discrepancy between kinase dead mTOR and the alteration of mTOR signalling by rapamycin and its derivatives. The discrepancy also identifies the possibility of a therapeutic advantage in inhibiting directly the catalytic activity of mTOR. It has been suggested that a catalytic inhibitor of mTOR may be a more effective antagonist of cancer cell proliferation and survival and that rapamycin may be more useful in combination with agents that can
20 compensate for its failure to completely disrupt pathway signalling (Choo and Blenis, Cancer Cell, 2006, 9, 77-79; Hay, Cancer Cell, 2005, 8, 179-183). Therefore, it is proposed that a kinase domain directed inhibitor of mTOR may be a more effective inhibitor of mTOR.
In addition to rapamycin's ability to induce growth inhibition (cytostasis) in its own right, rapamycin and its derivatives have been shown to potentiate the cytotoxicity of a
25 number of chemotherapies including cisplatin, camptothecin and doxorubicin (reviewed in ref. 20). Potentiation of ionising radiation induced cell killing has also been observed following inhibition of mTOR (ref. 24). Experimental and clinical evidence has shown that rapamycin analogues are showing evidence of efficacy in treating cancer, either alone or in combination with other therapies (see refs. 10, 18, 20). These findings suggest that
30 pharmacological inhibitors of mTOR kinase should be of therapeutic value for treatment of the various forms of cancer comprising solid tumours such as carcinomas and sarcomas and the leukaemias and lymphoid malignancies. In particular, inhibitors of mTOR kinase should

WO 2008/023161

PCT/GB2007/003179

be of therapeutic value for treatment of, for example, cancer of the breast, colorectum, lung (including small cell lung cancer, non-small cell lung cancer and bronchioalveolar cancer) and prostate, and of cancer of the bile duct, bone, bladder, head and neck, kidney, liver, gastrointestinal tissue, oesophagus, ovary, pancreas, skin, testes, thyroid, uterus, cervix and 5 vulva, and of leukaemias (including ALL and CML), multiple myeloma and lymphomas.
Renal cell carcinoma in particular, has been identified as sensitive to the rapamycin derivative CCI-779, resulting from a loss of VHL expression (Thomas et al Nature Medicine, 2006, 12, 122-127). Tumours that have lost the promyelocytic leukaemia (PML) tumour suppressor, have also been shown to be sensitive to inhibition of mTOR by rapamycin as a
10 consequence of disruption of the regulation of the mTOR signalling pathway (Bernadi, Nature, 2006,442, 779-785) and the use of an mTOR kinase inhibitor in these diseases should be of therapeutic value. These latter examples in addition to those of PTEN deficiency or PI3K mutation indicate where a targeted approach to the use of mTOR inihibitors may prove particularly effective due to an underlying genetic profile, but are not considered to be
15 exclusive targets.
Recent studies have revealed a role for mTOR kinase in other diseases (Easton & Houghton, Expert Opinion on Therapeutic Targets, 2004, 8, 551-564). Rapamycin has been demonstrated to be a potent immunosuppressant by inhibiting antigen-induced proliferation of T cells, B cells and antibody production (Sehgal, Transplantation Proceedings, 2003, 35, 7S-
20 14S) and thus mTOR kinase inhibitors may also be useful immunosuppressives. Inhibition of the kinase activity of mTOR may also be useful in the prevention of restenosis, that is the control of undesired proliferation of normal cells in the vasculature in response to the introduction of stents in the treatment of vasculature disease (Morice et al., New England Journal of Medicine, 2002, 346, 1773-1780). Furthermore, the Rapamycin analogue,
25 everolimus, can reduce the severity and incidence of cardiac allograft vasculopathy (Eisen el a;., New England Journal of Medicine, 2003, 349, 847-858). Elevated mTOR kinase activity has been associated with cardiac hypertrophy, which is of clinical importance as a major risk factor for heart failure and is a consequence of increased cellular size of cardiomyocytcs (Tee & Blenis, Seminars in Cell and Developmental Biology, 2005, 16, 29-37). Thus mTOR
30 kinase inhibitors are expected to be of value in the prevention and treatment of a wide variety of diseases in addition to cancer.

WO 2008/023161

PCT/GB2007/003171)

The vast majority of mTOR pharmacology to date has focused on inhibition of mTOR via rapamycin or its analogues. However, as noted above, the only non-rapamycin agents that have been reported to inhibit mTOR's activity via a kinase domain targetted mechanism are the small molecule LY294002 and the natural product wortmannin (ref. 21). 5 Summary of the Invention
The present inventors have identified compounds which are ATP-competitive inhibitors of mTOR, and hence are non-rapamycin like in their mechanism of action.
Accordingly, the first aspect of the present invention provides a compound of formula T:

, or a pharmaceutically acceptable salt thereof, wherein:
one or two of X5, X6 and X8 is N, and the others are CH;
R7 is selected from halo, OR01, SRS1, NRN1RN2, NRN7aC(0)Rcl, NRN7bS02RS2a, an optionally
substituted 5- to 20-memberedheteroaryl group, or an optionally substituted C5.2o aryl group,
15 where R01 and Rsl are selected from H, an optionally substituted C5.2o aryl group, an optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted C1-7 alky] group; RN1 and RN2 are independently selected from H, an optionally substituted C1-7 alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, an optionally substituted C5-20 aryl group or RN1 and RN2 together with the nitrogen to which they are bound
20 form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms;
RC1 is selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5-to 20-membered heteroaryl group, an optionally substituted C1-7 alkyl group or NRN8RN9, where RNS and RN9 are independently selected from H, an optionally substituted C1.7 alkyl group, an optionally substituted 5- to 20-memberedheteroaryl group, an optionally substituted
25 C5.20 a*yl group or RN8 and RN9 together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms; RS2a is selected from H, an optionally substituted C5-20 aryl group, an optionally substituted 5-to 20-memberedheteroaryl group, or an optionally substituted C1.7 alkyl group; RN7a and RN7b are selected from H and a CM alkyl group;

WO 2008/023161

PCT/GB2007/003179

R2 is selected from H, halo, OR02, SRS2b, NRN5RN6, an optionally substituted 5- to 20-
memberedheteroaryl group, and an optionally substituted C5-20 aryl group,
wherein R02 and RS2b are selected from H5 an optionally substituted C5.20 aryl group, an
optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted C1.7
5 alkyl group; RN5 and RN6 are independently selected from H, an optionally substituted C1.7
alkyl group, an optionally substituted 5- to 20-membered heteroaiyl group, and an optionally
substituted C5.20 aryl group, or RN5 and RN6 together with the nitrogen to which they are
bound form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms.
According to a second aspect of the present invention there is provided a compound of
10 formula la or lb:

, or a pharmaceutically acceptable salt thereof, wherein:
one or two of X5, X6 and X8 is N, and the others are CH;
R7 is selected from halo, OR01, SRS1, NRN,RNZ, NRN7aC(0)Rcl, NRN7bS02RS2a, an optionally
15 substituted 5- to 20-membered heteroaryl group, or an optionally substituted C5.20 aryl group, where R01 and Rsl are selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted C1.7 alkyl group; RN1 and RN2 are independently selected from H, an optionally substituted C1.7 alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, an optionally
20 substituted C5,2o aryl group or RN1 and RN2 together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms; RC! is selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5-to 20-membered heteroaryl group, an optionally substituted C1.7 alkyl group or NRmRw, where RN8 and RN9 are independently selected from H, an optionally substituted C1.7 alkyl
25 group, an optionally substituted 5- to 20-membered heteroaryl group, an optionally substituted C5-20 aryl group or R and R together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms; RS2a is selected from H, an optionally substituted C3-20 aryl group, an optionally substituted 5-to 20-membered heteroaryl group, or an optionally substituted Cf_7 alkyl group;

WO 2008/023161

PCT/GB2007/003179

RN7a and RN7b are selected from H and a CM alkyl group;
R2 is selected from H, halo, OR02, SRS2b, NRN5RN6, an optionally substituted 5- to 20-
memberedheteroaryl group, and an optionally substituted C5.20 aryl groups
wherein R02 and RS2b are selected from H, an optionally substituted C5.2o aryl group, an
5 optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted C1.7
allcyl group; RN5 and RN6 are independently selected from H, an optionally substituted C1.7
alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, and an optionally
substituted C5.20 aryl group, or RN5 and RN6 together with the nitrogen to which they are
bound form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms.
10 According to a third aspect of the present invention there is provided a compound of

, or a pharmaceutically acceptable salt thereof, wherein: one or two of X5, X6 and X8 is N, and the others are CH;
15 R7 is selected from halo, OR01, SRS1, NRNlRN2, NRN7°C(0)RC1, NRN7bS02RS2a, an optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted C5.20 aryl group, where R01 and Rsl are selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted C1-7 alkyl group; RN1 and RN2 are independently selected from H, an optionally substituted C1-7
20 alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, an optionally substituted C5.20 aryl group or RNl and RN2 together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms; RC1 is selected from H, an optionally substituted C5-20 aryl group, an optionally substituted 5-to 20-membered heteroaryl group, an optionally substituted C1.7 alkyl group or NRN8RN9S
25 where RN8 and RN9 are independently selected from H, an optionally substituted C1-7 alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, an optionally substituted C5.20 aryl group or RN8 and RN9 together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms;

WO 2008/023161

PCT/GB2007/003179

Rs2a is selected from H, an optionally substituted C5.2o aryl group, an optionally substituted 5-to 20-membered heteroaryl group, or an optionally substituted C\.j alkyl group; RN7a and RN7b are selected from H and a CM alkyl group;
R2 is selected froni H, halo, OR02, SRS2\ NRN5RN6S an optionally substituted 5- to 20-5 membered heteroaryl group, and an optionally substituted C5-20 aryl group,
wherein R02 and R are selected from H, an optionally substituted Cs.2o aryl group, an optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted C1-7 alkyl group; RN5 arid RN6 are independently selected from H, an optionally substituted C1.7 alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, and an optionally 10 substituted C5.20 aryl group, or RN5 and RN6 together with the nitrogen to which they are bound form an optimally substituted heterocyclic ring containing between 3 and 8 ring atoms. According XO a further aspect of the present invention there is provided a compound of formula I:

15 , or a pharmaceuticlly acceptable salt thereof, wherein: one or two of X5, X6 and X8 is N, and the others are CH;
R7 is selected from halo, OR01, SRSI, NRNlRN2, NRN7aC(0)Rcl, NRN7bS02RS2a, an optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted C5.2o aryl group, where R01 and Rsl are selected from H, an optionally substituted C5.20 aryl group, an
20 optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted C1.7 alkyl group; RNI atfd R are independently selected from H, an optionally substituted C1-7 alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, an optionally substituted C5.20 aryl group or RN1 and RN2 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms;
25 RC1 is selected fron H, an optionally substituted C5-20 aryl group, an optionally substituted 5-to 20-membered heteroaryl group, an optionally substituted C1-7 alkyl group or NRN8RN9, where RN8 and RN9 are independently selected from H, an optionally substituted C1-7 alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, an optionally substituted

2008/023161

PCT/GB2007/003179

C5.2o aryl group or RN8 and RN9 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms;
RS2a is selected from H, an optionally substituted C5.2o aryl group, an optionally substituted 5-to 20-memberedheteroaryl group, or an optionally substitute^ Ci.7 alkyl group; 5 RN7a and RN7b are selected from H and a CM alkyl group;
R2 is selected from H, halo, OR02, SRS2b, NRN5RN6, an optionally substituted 5- to 20-memberedheteroaryl group, and an optionally substituted C$.iQ aryl group, wherein R02 and RS2b are selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5- to 20-memberedheteroaryl group, 0r an optionally substituted C1.7
10 alkyl group; RN5 and RN6 are independently selected from ^ an optionally substituted C1.7 alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, and an optionally substituted C5-20 aryl group, or RN5 and RN6 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms.
According to &tfxrt£m'aspertofifeprGS^wvtsn&mtbeTQ is provided a compound1 of
15 formula la or lb:

or a pharmaceutically acceptable salt thereof,wherein:
one or two of Xs, X6 and X8 is N, and the others are CH;
R7 is selected from halo, OR01, SRS\ NRNIRN25 NRN7aC(0)Itcl, NRN7bS02RS2a, an optionally
20 substituted 5- to 20-memberedheteroaryl group, or an optionally substituted C5.20 aryl group, where R01 and Rsl are selected from H, an optionally Substituted C5_2o aryl group, an optionally substituted 5- to 20-membered heteroaryl group, 0r an optionally substituted d.7 alkyl group; RN1 and RN2 are independently selected from H, an optionally substituted C1.7 alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, an optionally
25 substituted C5-20 aryl group or RN1 and RN2 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atornS
RCI is selected from H, an optionally substituted C5-20 aryl ghoup, an optionally substituted 5-to 20-membered heteroaryl group, an optionally substitute C1-7 aIkyl group or NRN8RN9, where RNB and RN9 are independently selected from H, an optionally substituted C1.7 alkyl

WO 2008/023161

PCT/GB2007/003179

group, an optionally substituted 5- to 20-membered heteroaryl group, an optionally substituted C5.20 aryi group or RN8 and RN9 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms;
RS2a is selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5-5 to 20-membered heteroaryl group, or an optionally substituted C1.7 alkyl group; RN7a and RN7b are selected from H and a CM alkyl group;
R2 is selected from H, halo, OR02, SRS2b, NRN5RN6, an optionally substituted 5- to 20-membered heteroaryl group, and an optionally substituted C5-20 aryl group, wherein R02 and RS2b are selected from H, an optionally substituted C5.2o aryl group, an
10 optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted C1.7 alkyl group; RN5 and RN6 are independently selected from H, an optionally substituted C1.7 alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, and an optionally substituted C5.20 aryl group, or RN5 and RN6 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms.
15 According to a further aspect of the present invention there is provided a compound of
formula la:

. or a pharmaceutical acceptable salt thereof,wherein: one or two of Xs, X6 and X8 is N, and the others are CH;
20 R7 is selected from halo, OR01, SRSI, NRN,RN2, NRN7aC(0)Rc\ NRWbS02RS2a, an optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted C5.20 aryl group, where R01 and Rsl are selected from H, an optionally substituted C5-20 aryl group, an optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted C1.7 alkyl group; RNI and RN2 are independently selected from H, an optionally substituted C1.7
25 alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, an optionally substituted C5.20 aryl group or RN] and RN2 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms;
RCI is selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5-to 20-membered heteroaryl group, an optionally substituted C1-7 alkyl group or NRN8RN9,

WO 2008/023161

PCT/GB2007/003179

where RN8 and RN9 are independently selected from H, an optionally substituted C1.7 alkyl group, an optionally substituted 5- to 20-memberedheteroaryl group, an optionally substituted C5-20 aryl group or RN8 and RN9 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms; 5 RS2a is selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5-to 20-memberedheteroaryl group, or an optionally substituted C1.7 alkyl group; RN7a and RN7b are selected from H and a CM alkyl group;
R2 is selected from H, halo, OR02, SRS2b, NRN5RN6, an optionally substituted 5- to 20-memberedheteroaryl group, and an optionally substituted C5.20 aryl group,
10 wherein R02 and RS2b are selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5- to 20-memberedheteroaryl group, or an optionally substituted C1.7 alkyl group; RN5 and RNS are independently selected from H, an optionally substituted C1.7 alkyl group, an optionally substituted 5- to 20-memberedheteroaryl group, and an optionally substituted C5.2o aryl group, or RN5 and RN6 together with the nitrogen to which they are
15 bound form a heterocyclic ring containing between 3 and 8 ring atoms.
According to a further aspect of the present invention there is provided a pharmaceutical composition comprising a compound of of formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or diluent. According to a further aspect of the present invention there is provided a compound of
20 formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof,for use in a method of treatment of the human or animal body.
According to a further aspect of the present invention there is provided the use of a compound of formula (I). (la) or (lb), or a pharmaceutically acceptable salt thereof,in the preparation of a medicament for treating a disease ameliorated by the inhibition of mTOR.
25 The compounds of formula (I), (la) or (lb), or a pharmaceutically acceptable salt
thereof, have activity as pharmaceuticals, in particular as modulators or inhibitors of mTOR activity, and may be used in the treatment of proliferative and hyperproliferative diseases/conditions, examples of which include the following cancers:
(1) carcinoma, including that of the bladder, brain, breast, colon, kidney, liver,
30 lung, ovary, pancreas, prostate, stomach, cervix, colon, endometrium, thyroid and skin;
(2) hematopoietic tumors of lymphoid lineage, including acute lymphocytic
leukaemia, B-cell lymphoma and Burketts lymphoma;

WO 2008/023161

PCI7GB2007/003179

(3) hematopoietic tumours of myeloid lineage, including acute and chronic myelogenous leukaemias and promyelocytic leukaemia;
(4) tumours of mesenchymal origin, including fibrosarcoma and rhabdomyosarcoma; and
5 (5) other tumours, including melanoma, seminoma, tetratocarcinoma, neuroblastoma and glioma.
Further aspects of the invention provide the use of a compound of formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for the treatment of: cancer, immuno-suppression, immune tolerance, autoimmune disease,
10 inflammation, bone loss, bowel disorders, hepatic fibrosis, hepatic necrosis, rheumatoid arthritis, restenosis, cardiac allograft vasculopathy, psoriasis, beta-thalassaemia, and ocular conditions such as dry eye. mTOR inhibitors may also be effective as antifungal agents.
Another further aspect of the invention provides for the use of a compound of formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, in the preparation of a
15 medicament for use as an adjunct in cancer therapy or for potentiating tumour cells for treatment with ionizing radiation or chemotherapeutic agents.
Thus the compounds of the present invention provide a method for treating cancer characterised by inhibition of mTOR, i.e. the compounds may be used to produce an anti¬cancer effect mediated alone or in part by the inhibition of mTOR.
20 Such a compound of the invention is expected to possess a wide range of anti-cancer
properties as activating mutations in mTOR have been observed in many human cancers, including but not limited to, melanoma, papillary thyroid tumours, cholangio carcinomas, colon, ovarian and lung cancers. Thus it is expected that a compound of the invention will possess anti-cancer activity against these cancers. It is in addition expected that a compound
25 of the present invention will possess activity against a range of leukaemias, lymphoid malignancies and solid tumours such as carcinomas and sarcomas in tissues such as the liver, kidney, bladder, prostate, endometrium, breast and pancreas. In particular such compounds of the invention are expected to slow advantageously the growth of primary and recurrent solid tumours of, for example, the skin, colon, thyroid, lungs, endometrium and ovaries. More
30 particularly such compounds of the invention, or a pharmaceutically acceptable salt thereof, are expected to inhibit the growth of those primary and recurrent solid tumours which are associated with mTOR, especially those tumours which are significantly dependent on mTOR

WO 201)8/023161

PCT/GB2007/003179

for their growth and spread, including for example, certain tumours of the skin, colon, thyroid, endometrium, lungs and ovaries. Particularly the compounds of the present invention are useful in the treatment of melanomas and gliomas.
Thus according to this aspect of the invention there is provided a compound of the 5 formula I or 1(A), or a pharmaceutically acceptable salt thereof, as defined herein for use as a medicament.
According to a further aspect of the invention there is provided the use of a compound of the formula (I), (fe) or (lb), or a pharmaceutically acceptable salt thereof, as defined herein in the manufacture of a medicament for use in the production of a mTOR inhibitory effect in a
10 warm-blooded animal such as man.
According to this aspect of the invention there is provided the use of a compound of the formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, as defined herein in the manufacture of a medicament for use in the production of an anti-cancer effect in a warm-blooded animal such as man.
15 According to a further feature of the invention, there is provided the use of a
compound of the formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, as defined herein in tfce manufacture of a medicament for use in the treatment of melanoma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder,
20 prostate, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries.
According to a further feature of the invention, there is provided the use of a compound of ths formula (L) (La} or (Ib) phacmaceutically acceptable salt thereof, as defined herein in the manufacture of a medicament for use in the treatment of melanoma.
25 glioma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, endometrium, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries.
According to a further aspect of the invention there is provided the use of a compound
30 of the formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, as defined herein in the production of a mTOR inhibitory effect in a warm-blooded animal such as man.

WO 2008/023161

PCT/GB20U7/00317!)

According to this aspect of the invention there is provided the use of a compound of the formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, as defined herein in the production of an anti-cancer effect in a warm-blooded animal such as man.
According to a further feature of the invention, there is provided the use of a 5 compound of the formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, as defined herein in the treatment of melanoma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries.
10 According to a further feature of the invention, there is provided the use of a
compound of the formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, as defined herein in the treatment of melanoma, glioma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, endometrium.
15 breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries.
According to a further feature of this aspect of the invention there is provided a method for producing a mTOR inhibitory effect in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a
20 compound of formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, as defined herein.
According to a further feature of this aspect of the invention there is provided a method for producing an anti-cancer effect in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a
25 compound of formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, as defined
herein.
According to an additional feature of this aspect of the invention there is provided a method of treating melanoma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in 30 the liver, kidney, bladder, prostate, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries, in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective

WO 2008/023161

PCT/GB2007/003179

amount of a compound of formula (I), (la) or (lb) or a pharmaceutically acceptable salt thereof as defined herein.
According to an additional feature of this aspect of the invention there is provided a method of treating melanoma, glioma, papillary thyroid tumours, cholangiocarcinomas, colon 5 cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, endometrium, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries, in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of formula (I), (la) or (lb) or a
10 pharmaceutically acceptable salt thereof as defined herein.
In a further aspect of the invention there is provided a pharmaceutical composition which comprises a compound of the formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, as defined herein in association with a pharmaceutically-acceptable diluent or carrier for use in the production of a mTOR inhibitory effect in a warm-blooded animal such
15 as man.
In a further aspect of the invention there is provided a pharmaceutical composition which comprises a compound of the formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, as defined herein in association with a pharmaceutically-acceptable diluent or carrier for use in the production of an anti-cancer effect in a warm-blooded animal such as
20 man.
In a further aspect of the invention there is provided a pharmaceutical composition which comprises a compound of the formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, as defined herein in association with a pharmaceutically-acceptable diluent or carrier for use in the treatment of melanoma, papillary thyroid tumours, cholangiocarcinomas,
25 colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries in a warm-blooded animal such as man.
In a further aspect of the invention there is provided a pharmaceutical composition
30 which comprises a compound of the formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, as defined herein in association with a pharmaceutically-acceptable diluent or carrier for use in the treatment of melanoma, glioma, papillary thyroid tumours,

WO 2008/023161

PCT/GB2IW7/003I79

cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid
malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, endometrium,
breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs
and ovaries in a warm-blooded animal such as man.
5 Other further aspects of the invention provide for the treatment of disease ameliorated
by the inhibition of mTOR, comprising administering to a subject in need of treatment a
therapeutically-effective amount of a compound of formula (I), (la) or (lb), or a
phannaceutically acceptable salt thereof, preferably in the form of a pharmaceutical
composition and the treatment of cancer, comprising administering to a subject in need of
10 treatment a therapeutically-effective amount of a compound of formula (I), (la) or (lb), or a
pharmaceutically acceptable salt thereof, in combination, preferably in the form of a
pharmaceutical composition, simultaneously or sequentially with ionizing radiation or
chemotherapeutic agents.
Definitions
15 The term "aromatic ring" is used herein in the conventional sense to refer to a cyclic
aromatic structure, that is, a structure having delocalised Jt-electron orbitals.
Nitrogen-containing heterocyclic ring having from 3 to 8 ring atoms: The term "Nitrogen-containing heterocyclic ring having from 3 to 8 ring atoms" as used herein refers to a 3 to 8 membered heterocylic ring containing at least one nitrogen ring atom. The term 20 "together with the nitrogen to which they are bound, form a heterocyclic ring containing between 3 and 8 ring atoms" as used herein refers to a 3 to 8 membered heterocylic ring containing at least one nitrogen ring atom. Examples of these groups include, but are not limited to:
Ni: aziridine (C3 ie 3 membered), azetidine (C4 ie 4 membered). pyrrolidine
25 (tetrahydropyrrole) (C5 ie 5 membered), pyrroline (e.g., 3-pyrroline,
2,5-dihydropyrrole) (C5 ie 5 membered), 2H-pyrrole or 3H-pyrrole (isopyrrolc,
isoazole) (C5 ie 5 membered), piperidine (Q ie 6 membered), dihydropyridine (C6 ie 6
membered), tetrahydropyridine (C6 ie 6 membered), azepine (C7 ie 7 membered);
N2: imidazolidine (C5 ie 5 membered), pyrazolidine (diazolidine) (C5 ie 5 membered),
30 imidazoline (C5 ie 5 membered), pyrazolone (dihydropyrazole) (C5 ie 5 membered),
piperazine (C6 ie 6 membered);

WO 2008/023161

PCT/GB2007/003179

N1O1: tetrahydrooxazole (C5 ie 5 membered), dihydrooxazole (C5 ie 5 membered),
tetrahydroisoxazole (C5 ie 5 membered), dihydroisoxazole (C5 ie 5 membered),
morpholine (C6 ie 6 membered), tetrahydrooxazine (C6, ie 6 membered).
dihydrooxazine (C6 ie 6 membered), oxazine (C6 ie 6 membered);
5 N1S1: thiazoline (C5 ie 5 membered), thiazolidine (C5 ie 5 membered),
thiomorpholine (C6 ie 6 membered);
N2O]: oxadiazine (C6 ie 6 membered);
N1O1S1: oxathiazine (C6 ie 6 membered).
Alkyl: The term "alkyl" as used herein, pertains to a monovalent moiety obtained by 10 removing a hydrogen atom from a carbon atom of a hydrocarbon compound having from 1 to 20 carbon atoms (unless otherwise specified), which may be aliphatic or alicyclic, and which may be saturated or unsaturated (e.g. partially unsaturated, fully unsaturated). Thus, the term "alkyl" includes the sub-classes saturated alkyl, alkenyl, alkynyl, saturated cycloalkyl, cycloalkyenyl, cylcoalkynyl, etc., discussed below. Unless otherwise specified, preferable 15 "alkyl" groups are saturated alkyl or saturated cycloalkyl groups, more preferably saturated alkyl groups.
In the context of alkyl groups, the prefixes (e.g. CM, C1-7, C1.20, C2-7, C3.7, etc.) denote the number of carbon atoms, or range of number of carbon atoms. For example, the term "CM alkyl", as used herein, pertains to an alkyl group having from 1 to 4 carbon atoms. 20 Examples of groups of alkyl groups include CM alkyl ("lower alkyl"), C1.7 alkyl, and C1.20 alkyl. Note that the first prefix may vary according to other limitations; for example, for unsaturated alkyl groups, the first prefix must be at least 2; for cyclic alkyl groups, the first prefix must be at least 3; etc.
The term saturated alkyl group includes saturated linear alkyl and saturated branched 25 alkyl.
Examples of (unsubstituted) saturated alkyl groups include, but are not limited to,
methyl (Ci), ethyl (C2), propyl (C3), butyl (C4), pentyl (C5), hexyl (Ce), heptyl (C7), octyl (Cg),
nonyl (C9), decyl (CJO), undecyl (Cn), dodecyl (C)2), tridecyl (C13), tetradecyl (CM),
pentadecyl (Cl5), and eicodecyl (C20).
30 Examples of (unsubstituted) saturated linear alkyl groups include, but are not limited
to, methyl (Ci), ethyl (C2), n-propyl (C3), n-butyl (C4), n-pentyl (amyl) (C5), n-hexyl (C6), and n-heptyl (C7).

WO 2008/023161

PCT/GB2007/n0317'J

Examples of (unsubstituted) saturated branched alkyl groups include iso-propyl (C3), iso-butyl (C4), sec-butyl (C4), tert-butyl (C4), iso-pentyl (C5), and neo-pentyl (C5).
Alkenyl: The term "alkenyl", as used herein, pertains to an alkyl group having one or more carbon-carbon double bonds. Examples of groups of alkenyl groups include C2-4 5 alkenyl, C2-7 alkenyl, C2-20 alkenyl.
Examples of (unsubstituted) unsaturated alkenyl groups include, but are not limited to, ethenyl (vinyl, -CH=CH2), 1-propenyl (-CH=CH-CH3), 2-propenyl (allyl, -CH-CH=CH2), isopropenyl (1-methylvinyl, -C(CH3):=CH2), butenyl (C4), pentenyl (C5), and hexenyl (C$).
Alkynyl: The term "alkynyl", as used herein, pertains to an alkyl group having one or 10 more carbon-carbon triple bonds. Examples of groups of alkynyl groups mclude C2-4 alkynyl, C2-7 alkynyl, C2-20 alkynyl.
Examples of (unsubstituted) unsaturated alkynyl groups include, but are not limited to. ethynyl (ethinyl, -C=CH) and 2-propynyl (propargyl, -CH2-OCH).
Cycloalkyl: The term "cycloalkyl", as used herein, pertains to an alkyl group which is 15 also a cyclyl group; that is, a monovalent moiety obtained by removing a hydrogen atom from an alicyclic ring atom of a carbocyclic ring of a carbocyclic compound, which carbocyclic ring may be saturated or unsaturated (e.g. partially unsaturated, fully unsaturated), which moiety has from 3 to 20 carbon atoms (unless otherwise specified), including from 3 to 20 ring atoms. Thus, the term "cycloalkyl" includes the sub-classes saturated cycloalkyl, 20 cycloalkenyl and cycloalkynyl. Preferably, each ring has from 3 to 7 ring atoms. Examples of groups of cycloalkyl groups include C3.20 cycloalkyl, C3.15 cycloalkyl, C3.10 cycloalkyl, C3.7 cycloalkyl.
Examples of cycloalkyl groups include, but are not limited to, those derived from:
saturated monocyclic hydrocarbon compounds: cyclopropane (C3), cyclobutane (C4),
25 cyclopentane (C5), cyclohexane (Ce), cycloheptane (C7), methylcyclopropane (C4),
dimethy[cyclopropane (C5), methylcyclobutane (C5), dimethylcyclobutane (C6),
methylcyclopentane (Ce), dimethylcyclopentane (C7), methylcyclohexane (C7),
dimethylcyclohexane (Cg), menthane (Cio);
unsaturated monocyclic hydrocarbon compounds: cyclopropene (C4),
30 cyclobutene (C4), cyclopentene (C5), cyclohexene (Ce), methylcyclopropene (C4),
dimethylcyclopropene (C5), methylcyclobutene (C5), dimethylcyclobutene (Cg),

WO 2008/023161

PCT/CB2007/003179

methylcyclopentene (C6), dimethylcyclopentene (C7), methylcyclohexene (C7),
dimethylcyclohexene (C6);
saturated polycyclic hydrocarbon compounds: thujane (Cio), carane (Cio), pinane
(C10), bornane (Cio), norcarane (C7), norpinane (C7), norbomane (C7), adamantane
5 (Cio), decalin (decahydronaphthalene) (Cio);
unsaturated polycyclic -hydrocarbon compounds: camphene (Cio), limonene (Cio),
pinene (Ct0);
polycyclic hydrocarbon compounds having an aromatic ring: indene (C9), indane
(e.g., 2,3-dihydro-lH-indene) (C9), tetraline (1^,3,4-tetrahydronaphthalene) (Cio),
10 acenaphthene (C12X fluorene (Co), phenalene (C13), acephenanthrene (C15),
aceanthrene (C16), cholanthrene (C2o).
Heterocyclyl: The term "heterocyclyl", as used herein, pertains to a monovalent
moiety obtained by removing a hydrogen atom from a ring atom of a heterocyclic compound,
which moiety has from 3 to 20 ring atoms (unless otherwise specified), of which from 1 to 10
15 are ring heteroatoms. Preferably, each ring has from 3 to 7 ring atoms, of which from 1 to 4
are ring heteroatoms. Preferably the ring heteroatoms are selected from O, N and S. The
heterocyclic ring may, unless otherwise specified, be carbon or nitrogen linked, and wherein a
-CH2- group can optionally be replaced by a -C(O)-, and a ring sulphur atom may be
optionally oxidised to form the S-oxides.
20 In this context, the prefixes (e.g. C3.20, C3.7, Cs-6, etc.) denote the number of ring
atoms, or range of number of ring atoms, whether carbon atoms or heteroatoms. For example, the term "Cs-^heterocyclyl" or "5 to 6 membered heterocyclyl", as used herein, pertains to a heterocyclyl group having 5 or 6 ring atoms. Examples of groups of heterocyclyl groups include C3.20 heterocyclyl (ie 3 to 20 membered heterocyclyl), C5.20 heterocyclyl (ie 5 to 20 25 membered heterocyclyl), C3-15 heterocyclyl (ie 3 to 15 membered heterocyclyl), C5.15 heterocyclyl (ie 5 to 15 membered heterocyclyl), C3.12 heterocyclyl (ie 3 to 12 membered heterocyclyl), C5.12 heterocyclyl (ie 5 to 12 membered heterocyclyl), C3_l0 heterocyclyl (ie 3 to 10 membered heterocyclyl), Cs-io heterocyclyl (ie 5 to 10 membered heterocyclyl), C3.7 heterocyclyl (ie 3 to 7 membered heterocyclyl), C5-7 heterocyclyl (ie 5 to 7 membered 30 heterocyclyl), and C5-6 heterocyclyl (ie 5 to 6 membered heterocyclyl).
Examples of monocyclic heterocyclyl groups include, but are not limited to, those derived from:

WO 2008/023161

PCT/GB2007/003179

N1: aziridine (C3 ie 3 memberd), azetidine (C4 ie 4 membered), pyrrolidine
(tetrahydropyrrole) (C5 ie 5 membered), pyrroline (e.g., 3-pyrroline,
2,5-dihydropyrrole) (C5 ie 5 membered), 2H-pyrrole or 3H-pyrrole (isopyrrole,
isoazole) (C5 ie 5 membered), piperidine (C6 is 6 membered), dihydropyridine (C6 is 6
5 membered), tetrahydropyridine (C$ ie 6 membered), azepine (C7 ie 7 membered);
01: oxirane (C3 ie 3 membered), oxetane (C4 ie 4 membered), oxolane
(tetrahydrofuran) (C5 ie 5 membered), oxole (dihydrofuran) (C5 ie 5 membered),
oxane (tetrahydropyran) (Cs ie 6 membered), dihydropyran (Cg ie 6 membered), pyran
(Ce ie 6 membered), oxepin (C7 ie 7membered);
10 S[: thiirane (C3 ie 3 membered), thietane (C4 ie 4 membered), thiolane
(tetrahydrothiophene) (C5 ie 5 membered), thiane (tetrahydrothiopyran) (Ce ie
6membered), thiepane (C7 ie 7 membered);
O2: dioxolane (C5 ie 5 membered), dioxane (C$ ie 6 membered). and dioxepane (C7 ie
7 membered);
15 O3: trioxane (C6 ie 6 membered);
N2: imidazolidine (C5 ie 5 membered), pyrazolidine (diazolidine) (C5 ie 5 membered),
imidazoline (Cs ie 5 membered), pyrazoline (dihydropyrazole) (C5 ie 5 membered),
piperazine (C6 ie 6membered);
N1O1: tetrahydrooxazole (C5 ie 5 membered), dihydrooxazole (C5 ie 5 membered),
20 tetrahydroisoxazole (C5 ie 5 membered), dihydroisoxazole (C5 ie 5 membered),
morpholine (Cg ie 6 membered), tetrahydrooxazine (C6 ie 6 membered),
dihydrooxazine (C& ie 6 membered), oxazine (Ce ie 6 membered);
N] S1: thiazol ine (C5 ie 5 membered), thiazolidine (C5 ie 5 membered),
thiomorpholine (C6 ie 6 membered);
25 N2O1: oxadiazine (C5 ie 6 membered);
0[Si: oxathiole (Cs ie 5 membered) and oxathiane (thioxane) (C5 ie 6 membered); and,
N1O1S1: oxathiazine(C6 is ie 6 membered).
Examples of substituted (non-aromatic) monocyclic heterocyclyl groups include those
30 derived from saccharides, in cyclic form, for example, mranoses (C5 ie 5 membered), such as
arabinofuranose, lyxofiiranose, ribofuranose, and xylofuranse, and pyranoses (C6 ie 6

WO 2008/023161

PCT/GB2007/003179

membered), such as allopyranose, altropyranose, glucopyranose, mannopyranose, gulopyranose, idopyranose, galactopyranose, and talopyranose.
Spiro-C3_7 cycloalkyl or heterocyclyl: The term "spiro C3.7 cycloaikyl or heterocyclyl" as used herein, refers to a C3-7 cycloalkyl or C3.7 heterocyclyl ring (3 to 7 membered) joined to 5 another ring by a single atom common to both rings.
C5.20 aryl: The term "C5.20 aryl" as used herein, pertains to a monovalent moiety
obtained by removing a hydrogen atom from an aromatic ring atom of a C5.20 aromatic
compound, said compound having one ring, or two or more rings (e.g., fused), and having
from 5 to 20 ring atoms, and wherein at least one of said ring(s) is an aromatic ring.
10 Preferably, each ring has from 5 to 7 ring atoms.
The ring atoms may be all carbon atoms, as in "carboaryl groups" in which case the group may conveniently be referred to as a "C5-20 carboaryl" group.
Examples of C5.20 aryl groups which do not have ring heteroatoms (i.e. C5-20 carboaryl groups) include, but are not limited to, those derived from benzene (i.e. phenyl) (C6), 15 naphthalene (Cio), anthracene (C14), phenanthrene (CH), and pyrene (C\G).
Alternatively, the ring atoms may include one or more heteroatoms, including but not limited to oxygen, nitrogen, and sulfur, as in "heteroaryl groups". In this case, the group may conveniently be referred to as a "C5.20 heteroaryl" group, wherein "C5-20" denotes ring atoms, whether carbon atoms or heteroatoms (or otherwise referred to as a 5 to membered heteroaryl 20 group). Preferably, each ring has from 5 to 7 ring atoms, of which from 1 to 4 are ring heteroatoms. Commonly, heteroatoms are selected from oxygen, nitrogen or sulphur.
Examples of C5.20 heteroaryl groups include, but are not limited to, C5 heteroaryl groups (5 membered heteroaryl groups) derived from furan (oxole), thiophene (thiole), pyrrole (azole), imidazole (1,3-diazole), pyrazole (1,2-diazole), triazole, oxazole, isoxazole, 25 thiazole, isothiazole, oxadiazole, tetrazole and oxatriazole; and C6 heteroaryl groups (6 membered heteroaryl groups) derived from isoxazine, pyridine (azine), pyridazine (1,2-diazine), pyrimidine (1,3-diazine; e.g., cytosine, thymine, uracil), pyrazine (1,4-diazine) and triazine.
The heteroaryl group may be bonded via a carbon or hetero ring atom.
30 Examples of C5-20 heteroaryl groups which comprise fused rings, include, but are not
limited to, C9 heteroaryl groups (9 membered heteroaryl groups) derived from benzofaran, isobenzofuran, benzothiophene, indole, isoindole: C1o heteroaryl groups (10 membered

VO 2008/023161

PCT/GB20II7/003179

heteroaiyl groups) derived from quinoline, isoquinoline, benzodiazine, pyridopyridme; CM heteroaiyl groups (14 membered heteroaryl groups) derived from acridine and xanthenc.
The above alkyl, heterocyclyl, and aryl groups, whether alone or part of another substituent, may themselves optionally be substituted with one or more groups selected from 5 themselves and the additional substituents listed below. Halo: -F, -CI, -Br, and -I. Hydroxy: -OH.
Ether: -OR, wherein R is an ether substituent, for example, a C1.7 alkyl group (also referred to as a C1.7 alkoxy group), a C3.20 heterocyclyl group (also referred to as a C3.20 10 heterocyclyloxy group), or a C5.20 aryl group (also refen-ed to as a C5.2o aryloxy group), preferably a C1-7 alkyl group. Nitro: -N02.
Cyano (nitrile, carbonitrile): -CN.
Acyl (keto): -C(=0)R, wherein R is an acyl substituent, for example, H, a C1.7 alkyl
15 group (also referred to as C1.7 alkylacyl or C1.7 alkanoyl), a C3.20 heterocyclyl group (also
referred to as C3..20 heterocyclylacyl), or a C5.20 aryl group (also referred to as C5.20 arylacyl),
preferably a C1.7 alkyl group. Examples of acyl groups include, but are not limited to,
-C(=0)CH3 (acetyl), -C(=0)CH2CH3 (propionyl), -C(=0)C(CH3)3 (butyryl), and -C(0)Ph
(benzoyl, phenone).
20 Carboxy (carboxylic acid): -COOH.
Ester (carboxylate, carboxylic acid ester, oxycarbonyl): -C(=0)OR, wherein R is an
ester substituent, for example, a C1-7 alkyl group, a C3-20 heterocyclyl group, or a C5.20 aryl
group, preferably a C 1.7 alkyl group. Examples of ester groups include, but are not limited to,
-C(=0)OCH3, -C(=0)OCH2CH3, -C(=0)OC(CH3)3, and -C(=0)OPh.
25 Amido (carbamoyl, carbamyl, aminocarbonyl, carboxamide): -C=NR1R'R2, wherein
R1 and R2 are independently amino substituents, as defined for amino groups. Examples of amido groups include, but are not limited to, -C(=0)NH2, -C(=0)NHCH3, -C(=0)N(CH3)2? -C(=0)NHCH2CH3, and -C(=0)N(CH2CH3)2, as well as amido groups in which R1 and R2, together with the nitrogen atom to which they are attached, form a heterocyclic structure as in, 30 for example, piperidinocarbonyl, morpholinocarbonyl, thiomorpholinocarbonyl, and piperazinylcarbonyl.

WO 2008/023161

PCT/GB2O07/O03179

Amino: -NR!R2, wherein R1 and R2 are independently amino substituents, for example, hydrogen, a C1-7 alkyl group (also referred to as C1-7 alkylamino or di-C1-7 alkylamino), a C3.20 heterocyclyl group, or a C5.20 aryl group, preferably H or a C1-7 alkyl group, or, in the case of a "cyclic" amino group, Rl and R2, taken together with the nitrogen 5 atom to which they are attached, form a heterocyclic ring having from 4 to 8 ring atoms. Examples of amino groups include, but are not limited to, -NH2, -NHCH3, -NHCH(CH3)2, -N(CHa)2, -N(CH2CH3)2, and -NHPh. Examples of cyclic amino groups include, but are not limited to, aziridinyl, azetidinyl. pyrrolidinyl, piperidino, piperazinyl, perhydrodiazepinyl, morpholino, and thiomorpholino. The cylic amino groups may be substituted on their ring by
10 any of the substituents defined here, for example carboxy, carboxylate and amido.
Aminosulfonyl -S(=0)2NR1R2, wherein Rl and R2 each independently is an amino substituent, as defined for amino groups. Examples of aminosulfony groups include, but are not limited to, -S(K))2NH2,-S(=0)2NHCH3, -S(0)2NHCH2CPI3 and - S(=0)2N(CH3)2.
Acylamido (acylamino): -NR'C(=0)R2, wherein R1 is an amide substituent, for
15 example, hydrogen, a C1.7 alkyl group, a C3.20 heterocyclyl group, or a C5.2o aryl group, preferably H or a C1-7 alkyl group, most preferably H, and R2 is an acyl substituent, for example, a C1.7 alkyl group, a C3-2o heterocyclyl group, or a C5-20 aryl group, preferably a C1-7 alkyl group. Examples of acylamide groups include, but are not limited to. -NHC(=0)CH3, -NHC(=0)CH2CH3, and -NHC(=0)Ph. R1 and R2 may together form a cyclic structure, as in,
20 for example, succinimidyl, maleimidyl, and phthalimidyl:

Urcido: -N(Rl)CONR2R3 wherein R2 and R3 are independently amino substituents, as defined for amino groups, and Rl is a ureido substituent, for example, hydrogen, a C1.7alk.yl group, a C3.20heterocyclyl group, or a C5.2oaryl group, preferably hydrogen or a C1-7alkyl 25 group. Examples of ureido groups include, but are not limited to, -NHCONH2, -NHCONHMe, -NHCONHEt, -NHCONMe2, -NHCONEt2, -NMeCONH2, -NMeCONHMe, -NMeCONHEt, -NMeCONMe2, -NMeCONEt2 and -NHC(=0)NHPh.
Acyloxy (reverse ester): -OC(=0)R, wherein R is an acyloxy substituent, for example, a C1-7 alkyl group, a C3.20 heterocyclyl group, or a C5.2o aryl group, preferably a C1.7 alkyl

WO 2008/023161

PCT/GB20(I7/(M3179

group. Examples of acyloxy groups include, but are not limited to, -OC(=0)CH3 (acetoxy), -OC0=O)CH2CH3, -OC(=0)C(CH3)3, -0C(=O)Ph, -OC(=0)C6H4F, and -OC(=0)CH2Ph.
Thiol: -SH.
Thioether (sulfide); -SR, wherein R is a thioether substituent, for example, a C1.7 alkyl 5 group (also referred to as a C1.7 alkylthio group), a C3.20 heterocyclyl group, or a C5-20 aryl group, preferably a C1.7 allcyl group. Examples of C1-7 alkylthio groups include, but are not limited to, -SCH3 and -SCH2CH3.
Sulfoxide (sulfinyl): -S(=0)R, wherein R is a sulfoxide substituent, for example, a C1-7
alkyl group, a C3.20 heterocyclyl group, or a C5.20 aryl group, preferably a Cj_7 alkyl group.
10 Examples of sulfoxide groups include, but are not limited to, -S(=0)CH3 and -S(=0)CH2CH3.
Sulfonyl (sulfone): -S(=0)2R, wherein R is a sulfone substituent, for example, a C1-7
alkyl group, a C3.20 heterocyclyl group, or a C5-20 aryl group, preferably a C^ alkyl group.
Examples of sulfone groups include, but are not limited to, -S(=0)2CH3 (methanesulfonyl,
mesyl), -S(=0)2CF3, -S(=0)2CH2CH3, and 4-methylphenylsulfonyl (tosyl).
15 Thioamido (thiocarbamyl): -C^S^R'R2, wherein Rl and R2 are independently amino
substituents, as defined for amino groups. Examples of amido groups include, but are not limited to, -C(=S)NH2, -C(=S)NHCH3, -C(=S)N(CH3)2, and -C(=S)NHCH2CH3.
Sulfonamino: -NR1S(=0)2R, wherein R1 is an amino substituent, as defined for amino groups, and R is a sulfonamino substituent, for example, a C1.7aUt.yl group, a C3.2oheterocyclyl 20 group, or a C5.2oaryl group, preferably a Ci^alkyl group. Examples of sulfonamino groups include, but are not limited to, -NHS(=0)2CH3, -NHS(=0)2Ph and -N(CH3)S(-0)2C6H5.
In addition, two or more adjacent substituents may be linked such that together with
the atoms to which they are attached from a C3.7 cycloalkyl, C3_2o heterocyclyl or C5.20 aryl
ring.
25 As mentioned above, the groups that form the above listed substituent groups, e.g. C1.7
alkyl, C3.2o heterocyclyl and C5-20 aryl may themselves be substituted. Thus, the above definitions cover substituent groups which are substituted.
According to a farther aspect of the present invention there is provided a compound of formula I:

WO 2008/(123161

PCT/GB2

, or a pharmaceutically acceptable salt thereof, wherein: one or two of X5, X6 and X8 is N, and the others are CH;
R7 is halo, OR01, SRS1, NRNIRN2, NRN7aC(=0)Rcl, NRN7bS02RS2a, a C5.20 heteroaryl group 5 optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or Ci-7alkyl, C2-7alkenyl, C2-7alkynyl, C3-7cycloaIkyl, C3-7cycloalkenyl, C3.2oheterocyclyl, Csoaiyl, Cs^oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol,
10 C1.7alkyl, Calkenyl, C2-7alkynyl, C3-7cycloalkyl, C3.7Cycloalkenyl3 C3.2oheterocyclyl. C5.2oaiyl, C5-20heteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino), or a C5.20 aryl group optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or Ci-7alkyl, C2-7alkenyl, C2-7alkynyl, C3.7cycloalkyl, C3-7cycloalkenyI,
15 C3_2oheterocyclyl, Cs^oaryl, Cs^oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, Ci.7alkyl, C2.7alkenyl, C2-7alkynyI, C3-7cycloalkyl, C3-7Cycloalkenyl, C3.2oheterocyclyl3 Cs.2oaryl, C5.2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy.
20 thioether, sulfoxide, sulfonyl, thioamido and sulfonamino),
where R01 and RS1 are H, a C5.20 aryl group, a C5-20 heteroaryl group, or a C1.7 alkyl group where each Ci.7alkyl, Cs^oheteroaryl, or C5-20aryl is optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or C1-7alkyL C2-7alkenyl, C2.7alkynyl, C3.7cycloalkyl, C3_7cycloalkenyl, C3.2oheterocyclyl; C5-2oaryl,
25 Cs^oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, Cj.7alkyl, C2-7alkenyl, C2-7alkynyl, C3_7cycloalkylJ C3-7cycloalkenyl, C3.2oheterocyclyl, C5.2oaryl, C5-2oheteroaryl.




ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfbnamino);
R l and RN2 are independently H, a C1-7alkyl group, a Cs-2oheteroaryl group, a C3.20 aryl group or RN1 and RN2 together with the nitrogen to which they are bound form a heterocyclic 5 ring containing between 3 and 8 ring atoms, where each Ci-7alkyl, C5-2oheteroaryl, Cs-ioaiy or heterocyclic is optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or C1.7BSk.yl, C2-7alkenyl, C2-7alkynyl, C3-7cycloalkyl, C3.7cycloalkenyl, C3-2oheterocyclyl, Cs_2oaryl, Cs^oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each
10 optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, C^alkyl, C2-7alkenyl, C2-7alkynyl, C3_7cycloalkyl, C3.7cycloalkenyl, C3_2oheterocyclyl, C5-20aryl, C5_2oheteroaryl, ether, acyl, ester, amido, amino, acylamido. ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino); Ra is H, a C5.20 aryf group, a C5.20 heteroaryl group, a C1.7 alkyl group or NRN5RN!) where
15 Rm and RN9 are independently selected from H, a C1.7 alkyl group, a C5-20 heteroaryl group, a C5.20 aryl group or RN8 and RN9 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms, where each Cj^alkyl, C5-2oheteroaryl, C5-2oaryl or heterocyclic ring is optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or C^alkyl, C2-7alkenyl,
20 C2-7alkynyl, C3.7cycloalkyl, C3_7cycloalkenyl, C3-2oheterocyclyl, C5-2oaryl, Cs^oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, ChalkyL, C2-7alkenyl, C2-7alkynyl, C3_7cycloalkyl, C3.7cycloalkenyl, C3_2oheterocyclyl, C5-20aryl, C5.2oheteroaryl, ether, acyl,
25 ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino);
R 2n is H, a C5-20 aryl group, a C5.20 heteroaryl group, or a C1-7 alkyl group where each Ci. 7alkyl, C5-2oheteroaryl or Csoaiyl is optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or Chalky 1, C2-7alkenyl, C2-7alkynyl,
30 C3.7cycloalkyl, C3_7cycloalkenyl, C3-2oheterocyclyl, Cs-aoaryl, Csoheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo.

WO 2008/023161

PCT/GB2007/003179

hydroxyl, nitro, cyano, carboxy, thiol, C|.7alkyl, C2-7aIkenyl, C2.7alk.ynyl, C^cycloalkyl, C3-7cycloalkenyl, C3-2 R2 is H, halo, OR02, SRS2b, NRN5RN6, a C5-20 heteroaryl group optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or Ci.7alkyl, C2-7alkenyl, C2-7alkynyl, C3-7cycloalkyl, C3_7cycloalkenyl, C3.2oheterocyclyl, C5.2oaryl, C5.2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether,
10 sulfoxide, sulfonyl., thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, Ci.7alkyl, C2-7aIkenyl, C2.7alkynyl, C3-7cycloalkyl, C3-7cycloalkenyl, C3_2oheterocyclyl, C5-2oaryl, C5_2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy. thioether, sulfoxide, sulfonyl, thioamido and sulfonamino), or a Cs.2o aryl group optionally substituted by one or more
15 groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or Ci_7alkyl, C2.7alkenyl, C2-7alkynyl, C3-7cycloalkyl, C3_7cycloalkenyl, C3_2oheterocyclyl, C5.2oaryl, Cs^oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, C^alkyl, C2.7alkenyl,
20 C2-7alkynyl, C3-7cycloalkyl, C3-7cycloalkenyl, C3_2oheterocyclyl, C5-2oaryl, Cs-2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino),
wherein R02 and RS2b are H, a C5.20 aryl group, a C5.20 heteroaryl group, or a C1.7 alkyl group where each Ci^alkyl, Cs^oheteroaryl or C5.2oaryl is optionally substituted by one or more
25 groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or Ci-jalkyl, C2-7alkenyl, C2-7alkynyl, C3_7cycloalkyl, C3-7cycloalkenyl, C3.2oheterocyclyl, C5-2oaryl, C5-2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, Ci.7alkyl, C2-7alkenyl,
30 C2-7alkynyl, C3-7cycloalkyl, C3_7cycloalkenyl, C3_2oheterocyclyl, CMoaryl, C5.2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino);

WO 2008/023161

PCT/GB2007/003179

RN5 and RNC are independently H, a C1.7 alkyl group, a C5.20 heteroaiyl group, a C5.20 aryl group, or RN5 and RN6 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms where each C^alkyl, Cs^oheteroaryl, C^-zoaryl or heterocyclic ring is optionally substituted by one or more groups selected from halo, 5 hydroxyl, nitro, cyano, carboxy, and thiol, or Ci^alkyl, C^alkenyl, C2-7alkynyl, Cs^cycloalkyl, C3-7cycloalkenyl, C3.2oheterocyclyl, Cs^oaryl, Cs_2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, Ci_7alkyl, C2-7alkenyl, C2-7alkynyl, C3-7cycloalkyl, 10 C3.7cycloalkenyl5 C3-2oheterocyclyl, Cs-2Qaiyls Cs-2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino).
According to a further aspect of the present invention there is provided a compound of formula la or lb:

15
, or a pharmaceutical^ acceptable salt thereof, wherein:
one or two of X5, X6 and X8 is N, and the others are CH;
R7 is halo, OR01, SRSI, NRN,RN2, NRN7aC(=0)Rcl, NRN7bS02RS2a, a C5.20 neteroaryl group
optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano.
20 carboxy, and thiol, or C1-7alkyl, C2-7alkenyl, C2-7alkynyI, C3.7cycloalkyl, C3.7cycIoalkenyl. C3-2oheterocyclyl, Csoaryl, C5.20heteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, C|.7alkyl, C2-7ahcenyl, C2.7alkynyl, C3.7cycIoalkyl, C3.7cycloalkenyI, C3_2oheterocycfyl,
25 Csjoaryl, C5-20oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino), or a C5.20 aryl group optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or C|.7alkyl, C2-7alkenyl, C2-7alkynyl, C3-7cycloaIkyl, C3-7cycloalkenyl, C3-2oheterocyclyl, C5-2oaryl C5.2oheteroaryl, ether, acyl, ester, amido, amino, acylamido,

WO 2008/023161

PCT/GB2007/003179

ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxy!, nitro, cyano, carboxy, thiol, C1-7alkyl, C2-7alkenyl, C2-7aikynyl, C3-7cyclo alkyl, C3.7cycloalkenyl, C3_2oheterocyclyl, Cs.2oaryI, C5-2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, 5 thioether, sulfoxide, sulfonyl, thioamido and sulfonamino),
where R01 and RS1 are H, a C5.20 aryl group, a C5.20 heteroaryl group, or a C1-7 alkyl group where each Ci^alkyl, Cs^oheteroaryl, or C5_2oaryl is optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or C1.7aikyl, C2-7alkenyl, C2_7alkynyl, C^cycloalkyl, C3.7cycloalkenyl, C3.2oheterocyclyl, Cs^oaryl,
10 C5.2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, Ci.7alkyl, C2-7alkenyl, C2-7alkynyl, C3,7cycloalkyl, C3-7cycloalkenyl, C3.2oheterocyclyl, Csoaryl, C5-2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl,
15 thioamido and sulfonamino);
RN1 and RN2 are independently H, a C[.7alkyl group, a C5-20heteroaryl group, a C5.20 aryl group or RN1 and RN2 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms, where each d^alkyl, C5-2oheteroaryl, Cs.soaryl or heterocyclic is optionally substituted by one or more groups selected from halo, hydroxy!,
20 nitro, cyano, carboxy, and thiol, or C1-7alkyl, C2-7alkenyl, C2.7alkynyl, C3~7cycloalkyl, C3.7cycloalkenyl, C3_2oheterocyclyl, C5-2oaryl, C5_2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, C1.7alkyl, C2-7alkenyl, C2-7alkynyl, C3.7cycloalkyl, C3.7cycloalkenyl,
25 C3-2oheterocyclyl, C5-2oaryl, C5-2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino); RC1 is H, a C5-20 aryl group, a C5.2o heteroaryl group, a C1-7 alkyl group or NRNR9 where Rm and RN9 are independently selected from H, a C1.7 alkyl group, a C5.20 heteroaryl group, a C5-20 aryl group or RN8 and RN9 together with the nitrogen to which they are bound form a
30 heterocyclic ring containing between 3 and 8 ring atoms, where each C1-7alkyl, C5. 2oheteroaryl, C5.2oaryl or heterocyclic ring is optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or C1-7alkyl, C2.7alkcnyl,

WO 2008/023161

PCI7GB2007/003179

C2.7alkynyi, C3.7cycloalkyl, C3.7cycloalkeny], C3-2heterocyclyl, C5.2oaryl, C^oheteroary], ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, C].7alkyl, C2.7alkenyl, C2-7alkynyl, 5 C3-7cycloalkyl, C3-7cycIoalkenyl, C3.20heterocyclyl3 C5-2oaryl, C5.2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino);
RS2n is H, a C5-20 aiyl group, a C5-20 heteroaryl group, or a C1.7 alkyl group where each Q. 7alkyl, Cs^oheteroaryl or C5_2oaryl is optionally substituted by one or more groups selected
10 from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or C^alkyl, C^alkenyl, C^alkynyl, C3-7cycloalkyl, C3.7cycloalkenyl, C3.2oheterocyclyl, Cs.2oaryl, C5-2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, Chalky 1, Cz^alkenyl, C2.7alkynyl, C3.7cycloalkyl,
15 Cs^cycloalkenyl, C3.2oheterocyclyl, Csjoaryl, C5-2oheteroaryI, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino);
RN7a and RN7b are H or a CM alkyl group; R2 is H, halo, OR02, SRS2b, NRN5RN6, a C5.20 heteroaryl group optionally substituted by one or
20 more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or Ci_7alkyl, C2-7alkenyl, C2-7alkynyl, C3.7cycloalkyl, C3.7cycloalkenyl, C3.2oheterocyclyl, C5.2oaryl, C5-2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, Ci^alkyl, C2-7alkenyl,
25 C2-7alkynyl, C3.7cycloalkyl, C3-7cycloalkenyl) C3_2oheterocyclyl, C5_2oaryl, Cs^oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino), or a Cg.20 aryl group optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or C^alkyl, C2-7alkenyl, C2-7alkynyl, C3.7cycloalkyl, C3-7cycloalkenyl, C3-2oheterocyclyl, Cs.20aryl,
30 C^oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, Chalky!, C2-7aIkenyl,

WO 2008/023161

PCT/GB2007/003179

C2.7alkynyl, C3-7cycloalkyl, C3-7cycloalkenyl, C3_20heterocyctyl C5-20aryl, C5.20heteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino),
wherein R02 and RS2b are H, a C5_20 aryl group, a C5-20 heteroaryl group, or a d.7 alkyl group 5 where each Cj.7alkyl, C5-2oheteroaryl or C5.20aryl is optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or Ci.7alkyl, C2.7alkenyl, C2-7alkynyl, C3.7cycloalkyl, C3_7cycloalkenyl, C3_20heterocyclyl, C5-20aryl, C5.20heteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more
10 groups selected from halo, hydroxyl, nitro, cyano, carbox/, thiol, C(_7alkyl, C2-?alkenyl, C2.7alkynyl, C3_7cycloalkyl, C3.7cycloalkenyl, C3_20heterocyclyl, C5-20aryl, C5.2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy. thioether, sulfoxide, sulfonyl, thioamido and sulfonamino); UNS and ftN6 are "mdependenfty H, a C1.7 d&y\ group, a C5-z taerUHrj group, a Z5.2Q ary'j
15 group, or RN5 and RN6 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms where each C^alkyl, C5.2oheteroaryl, C5.20aryl or heterocyclic ring is optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or Ci.7alKyl, C2-7alkenyl, C2.7alkynyl, C3_7cycloalkylJ C3.7cycloalkenyl, C3.20heterocyclyl, C5.20aryl, C5.20heteroaryl, ether, acyl,
20 ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or mofe groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, Ci.7alkyl, C2.7alkenyl, C^alkynyl, C3_7cycloalkyl, C3.7cycloalkenyl, C3.2oheterocyclyl, C5.20aryl, C5-2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide- sulfonyl, thioamido and
25 sulfonamino).

, or a pharmaceutically acceptable salt thereof, wherein:
According to a further aspect of the present invention there is provided a compound of formula la:

WO 2008/023161

PCT/GB2007/003179

one or two of X5, X6 and X8 is N, and the others are CH;
R7 is halo, OR01, SRS1, NRNlRN2, NRN7aC(0)Rcl, NRN7bS02RS2a, a C5.20 heteroaryl group optionally substituted by one or more groups selected from halo, hydroxy 1, nitro, cyano, carboxy, and thiol, or Chalky 1, C2_7alkenyl, C2-7alkynyl3 C3.7cycloalkyl, C3-7cycloalkenyl, 5 C3-2oheterocyclyl, C.2oaryl, C^oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, Ci-7alkyl, C2.7alkenyl, C2-7alkynyl, C3_7cycloalkyl, C3-7cycloalkenyl, C3-2oheterocyclyl, C5-2oaryl, Cgoheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy,
10 thioether, sulfoxide, sulfonyl, thioamido and sulfonamino), or a C5.20 aryl group optionally substituted by one or more groups selected from halo, hydroxy], nitro, cyano, carboxy, and thiol, or Ci^alkyl, C2_7alkenyl, C2-7alkynyl, Cs^cycloalkyl, C3.7cycloalkenyl, C3_2oheterocyclyl, Csoaryl, C5-2oheteroaryI, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally
15 substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, C1.7alky 1, C2.7alken.yl, C2-7alkyny 1, C3.7cycloalkyl, C3.7cycloalkeny 1, C3.2oheterocyclyl, C5-2oaryl, C5_2ohcteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino), where R01 and Rsl are H, a C5.20 aryl group, a Cs.2o heteroaryl group, or a C1-7 alkyl group
20 where each Chalky 1, C5-2oheteroaryl, or Cs-2oaryl is optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or Ci^alkyl, C2-7alkenyl, C2-7alkynyl, C3.7cycloalkyl, C3-7cycloalkenyl, C3-2oheterocyclyl, Cs-2oaryl, Cs-zoheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more
25 groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, C1.7alkyl, C2-7alkenyI, C2-7alkynyl, C3-7cycloalkyl, C3.7cycloalkenyl, C3.2oheterocyclyl, Csoaryl, C5.2(]heteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino); RN1 and Rm are independently H, a Ci^alkyl group, a C5-2oheteroaryl group, a C5.20 aryl
30 group or RNI and RN2 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms, where each C1_7alkyl3 C5_2oheteroaryl, C5.2oaryl or heterocyclic is optionally substituted by one or more groups selected from halo, hydroxyl,

WO 2008/023161

PCT/GB2007/003179

nitro, cyano, carboxy, and thiol, or Ci-7alkyl, C2-7alkenyl, C2-7alkynyl, C3.7cycloalkyl, C3.7cycloalkenyl, C3„2oheterocyclyl, Csoaryl, C5.2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, 5 carboxy, thiol, Ci.7alkyl, C2-7alkenyl, C2-7alkynyl, C3_7cycIoalkyl, C3.7cycloalkenyl, Cwoheterocyclyl, Cs.2oaryl, Cs-2oheteroaryl, etlier, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino); RC1 is H, a C5.20 aryl group, a C5.20 heteroaryl group, a C1.7 alkyl group or NRN8RN9 where RN8 and RN9 are independently selected from H, a C1.7 alkyl group, a C5.20 heteroaryl group, a
10 C5.20 aryl group or RN8 and RN9 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms- where each Ci.7alkyl, C5. 2oheteroaryl, Csoaryl or heterocyclic ring is optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or Csalkyl, C2-7alkenyl. Chalky svyl, C^cycloalkyl, C3.7Gycloalk.eayl, C3_2orieterocyclyl, Cs.oaryl, C5_2tkete*oaryl,
15 ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, C^alkyl, C^alkenyl, C2-7alkynyl, C3.7cycloalkyl, Ca^cycloalkenyl, C3.2oheterocyclyl, Cs^oaryl, Cs^oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and
20 sulfonamino);
RS2a is H, a C5.20 aryl group, a C5-20 heteroaryl group, or a C1-7 alkyl group where each Ci_ 7alkyl, Cs-2oheteroaryl or C5.2oaryl is optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or Ci^alkyl, C2-7aIkenyl, C^&lkynyl, C3.7cycloalkyl, C3.7cycloalkenyl, C3.2oheterocyclyl, C5.2oaryl, Cs^oheteroaryl, ether, acyl,
25 ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, Ci^alkyl, Cwalkenyl, C^alkynyl, C3-7cycloalkyls C3.7cycloalkenyl, C3.2oheterocyclyl, Cs^oaryl, Cs^oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and
30 sulfonamino);
RN7a and RN7b are H or a CM alkyl group;

WO 2008/023161

PCT/GB201I7/003179

R2 is H, halo, OR02, SRS2b, NRN5RN6, a C5_2o heteroaryl group optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or C1-7aJkyl, C2.7alk.enyl, C2-7alkynyl, C3-7Tcycloalkyl, Cj^cycloalkenyl, C3-2oheterocyclyl, Cwoaryl, Cs^oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, 5 sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, C1-7alkyl, C2-7alkenyl, C2-7alkynyl, Cscycloalkyl, C3-7cycloalkenyl, C3.2oheterocyclyl, CsoaryL Csoheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino), or a C5.20 aryl group optionally substituted by one or more
10 groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or Ci^alkyl, C2.7alkenyl, C2-7alkynyl, C3.7cycloalkyl, C3.7cycloalkenyl, C3_2oheterocyclyl, Csoaiyl, C5.2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, Chalky!, C2.7alkenyl,
15 C2-7alkynyl, C3-7cycloalkyl, C3-7cycloalkenyl, C3-2oheterocyclyl, C5-2oaryl, Cs^oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino),
wherein R02 and RS2b are H, a C5-20 aryl group, a C5-20 heteroaryl group, or a C1-7 alkyl group where each Ci-7alkyl, Cs^oheteroaryl or Cs^oaryl is optionally substituted by one or more
20 groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or C1-7alkyl, C2.7alkenyl, C2-7 alkynyl, C3-7cycloalkyl, C3.7cycloalkenyI, C3_2oheterocyclyl, Csoaryl, C5.2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, Ci^alkyl, C2-7alkenyl,
25 C2-7alkynylJ C3-7cycloalkyl, C3_7cycloalkenyl, C3-2oheterocyclyl, Cs-2oaryI, Cs^oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino);
RNS and RN6 are independently H, a d.7 alkyl group, a C5.20 heteroaryl group, a C5.20 aryl group, or RN5 and RN5 together with the nitrogen to which they are bound form a heterocyclic
30 ring containing between 3 and 8 ring atoms where each C]_7alkyl, Csoheteroaryl, C5.2oaryl or heterocyclic ring is optionally substituted by one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, and thiol, or Ci-7alkyl, C2-7alkenyl, C2-7alkynyl,

WO 2008/023161

PCT/GB2007/003179

C3.7cycloalkyls Cscycloalkenyl, C3.2oheterocyclyl, C5.2oaryl, C5_2oheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido3 acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino (each optionally substituted with one or more groups selected from halo, hydroxyl, nitro, cyano, carboxy, thiol, Ci_7alkyl, C2-7alkenyl, C2..7alkynyl, C3.7cycloalkyl; 5 C3.7cycloalkenyl, C3.2oheterocyclyl, Csoaryl, Csoheteroaryl, ether, acyl, ester, amido, amino, acylamido, ureido, acyloxy, thioether, sulfoxide, sulfonyl, thioamido and sulfonamino). Further Preferences
The following preferences can apply to each aspect of the present invention, where 10 applicable. The preferences for each group may be combined with those for any or all of the other groups, as appropriate. X5, X6, and*8
When two of X5, X6 and Xs are N, preferably X5 and X8 are N. It is preferred that only one of X\ X6 and X8 is "N. More preferably one of X5 and Xs 15 is N, and most preferably X8 is N. R7
R7 is preferably selected from an optionally substituted C5-20 aryl group, OR01, SRS1,
NRmRN2, NRN7aC(0)Rcl and NRN7bS02RS2a, where R01, RS1, RN1, RN2, RN7a, RN7\ RC1 and
RS2a are as previously defined. It is further preferred that R7 is preferably selected from an
20 optionally substituted C5-20 aryl group, OR01, NRNIRN2, NRN7[1C(0)RCI and NRN7bS02Rs2a.
If R7 is ORot, then preferably R01 is a C1.7 alkyl group, which may be substituted.
If R7 is NRN,RN2, then preferably RN2 is selected from H and CM alkyl (e.g. methyl)
and more preferably is H. If RNl is C1.7 alkyl, it is preferably selected from C3.7 cycloalkyl. If
RN1 is C5.20 aryl, it is preferably selected from C5.10 aryl (e.g. phenyl, pyrrolyl, pyridyl,
25 pyrazolyl, furanyl, thiophenyl, pyrazinyl, pyrimidinyl, tetrazolyi, thiazoiyi, indazolyl,
imidazolyl, triazolyl, oxadiazolyl) and more preferably C5-6 aryl (e.g. phenyl, pyrrolyl,
pyridyl, pyrazolyl, furanyl, thiophenyl, pyrazinyl, pyrimidinyl. tetrazolyi, thiazoiyi,
imidazolyl, triazolyl, oxadiazolyl). Particularly preferred groups include furyl, phenyl,
pyridyl, pyrrolyl, pyrazolyl and thiophenyl. The aforementioned groups are optionally
30 substituted, and in some embodiments are preferably substituted. Substituent groups may
include, but are not limited to, C4.7 alkyl, C3.20 heterocyclyl, C5.20 aryl, carboxy, ester, ether

WO 2008/023161

PCT/GB2007/003179

(eg C1-7alkoxy), hydroxy, aryloxy, cyano, halo, nitro, araido, sulfonyl, sulfonylamino, amino sulfonyl and amino.
If R7 is NRN7BC(0)RC1, then RN7a is preferably H. RC1 may be an optionally substituted C5.2o aryl group (e.g. phenyl, imadazolyl, quinoxalinyl), C3.20 heterocyclyl, C1-7 5 alkyl (e.g. propenyl, methyl (substituted with thiophenyl)) or NRN8RN9. RN8 is preferably hydrogen, and RN9 is preferably C1-7 alkyl (e.g. ethyl).
If R7 is NRN7bS02RS2a, then RN7b is preferably H. RS2a is preferably CU7 alkyl (e.g. methyl).
If R7 is a C5.20 aryl group, it is preferably an optionally substituted C5.10 aryl and more 10 preferably an optionally substituted C5-6 aryl group. Most preferably it is an optionally substituted phenyl group, wherein the optional substituents are preferably selected from halo, hydroxyl, C1.7 alkyl, C1.7 alkoxy, Csarylamino and Calkylamino and wherein the susbtitutent alkyl, alkoxy, or aryl groups may be further optionally substituted by one or more groups selected from halo, hydroxyl, C1.7 alkyl, C1-7 alkoxy, Caryl, Cs-earylamino and 15 Ci^alkylamino.
If R7 is a 5 to 20 membered heteroaryl group, it is preferably an optionally substituted 5 to 10 membered heteroaryl and more preferably an optionally substituted 5 or 6 memebered heteroaryl group.
In one embodiment, R7 is an optionally substituted 05,20 aryl group or an optionally 20 substituted 5 to 20 membered heteroaryl group, wherein the optional substituents are preferably selected from halo, hydroxyl, cyano, C1.7 alkyl, C^alkoxy, sulfonamino (for example -NHS(=0)2Ci-7alkyl) amino (for example -NH2, Cs-garylamino, Ci^alkylamino, and di-(C1_7allqrl)armno), and amido (for example -CONH2, -CONHCt_7alkyl, -CON(C,.7alkyl)2 and -CONHheterocycyl) and wherein the susbtitutent alkyl. alkoxy, or aryl groups may be 25 further optionally substituted by one or more groups selected from halo, hydroxyl, C1.7 alkyl, C1.7 alkoxy, C5.6aryl, —NHS(=0)2.C1.7alkyl, C5-6arylamino, di-(Ci.7alkyl)amino and Ci.7alkylamino.
In one embodiment, R7 is an optionally substituted phenyl group, wherein the optional
substituents are preferably selected from halo, hydroxyl. cyano, CM alkyl, C1.7alkoxy.
30 sulfonamino (for example -NHS(=0)2Ci.7alkyl) amino (for example -NH2, Cs_6arylamino,
Ci.7alkylamino, and di-(C1-7alkyl)amino), and amido (for example -CONH2, -CONHC1alkyl,
-CON(Ci.7alkyl)2 and -CONHheterocycyl) and wherein the susbtitutent alkyl, alkoxy, or aryl

WO 2008/023161

PCT/GB2007/003179

groups may be further optionally substituted by one or more groups selected from halo, hydroxyl, C1.7 alkyl, C1.7 alkoxy, Cs-garyl, -NHS(=0)2C1-7alkyl, C5.6arylaminC5-6, di-(Ci.7alkyl)amino and Ci-7alkylamino.
In one embodiment, R7 is an optionally substituted phenyl group, wherein the optional 5 substituents are preferably selected from halo, hydroxyl, cyano, C1-7 alkyl, C1-7alkoxy, amino (for example -NH2, C5.6arylamino, C1-7alkylamino, and di-(C1.7alkyl)amino), and amido (for example -CONH2, -CONHCi.7alkyl, -CON(C1-7alkyl)2 and -CONHheterocycyl) and wherein the susbtitutent alkyl, alkoxy, or ary! groups may be further optionally substituted by one or more groups selected from halo, hydroxyl, C1.7 alkyl, C1_7 alkoxy, Csaryl, C5-6aryIamino, di-
10 (Ci-7alkyl)amino and Ci-7alkylamino.
In one embodiment, R7 is an optionally substituted phenyl group, wherein the optional substituents are preferably selected from fluoro, hydroxyl, cyano, nitro, methyl, methoxy, -OCH2CH3, -NHa, -NHS02CH3, -CH2NHS02CH3, -OCHF2, -CH2OH, -C02H-CONH2, -CONHMe, -CONHEt, -CONHCH(CH3)2, -CONHCH2CH2F, -CONHCH2CHF2,
15 -C0NHCH2CH20H, -CONMeEt, -CONMe2, N-methylpiperazinylcarbonyl and 4-hydroxypiperidinylcarbonyl.
In one embodiment, R7 is an optionally substituted phenyl group, wherein the optional substituents are preferably selected from fluoro, hydroxyl, cyano, nitro, methyl, methoxy, -CH2OH, -C02H, -CONH2j -CONHMe, -CONHEt, -C0NHCH2CH2F, -CONHCH2CHF2,
20 -CONHCHzCHzOH, -CONMeEt, -CONMe2j N-methylpiperazinylcarbonyl and 4-hy droxy piper idinylcarbonyl.
In one embodiment, R7 is an optionally substituted phenyl group, wherein the optional substituents are preferably selected from methoxy, -OCH2CH3, -NH2, -NHSO2CH3, -CH2NHS02CH3, -OCHF2, -CH2OH, -CONH2, -CONHMe and -C0NHCH(CH3)2.
25 In one embodiment R7 is an optionally substituted 5 or 6 memebered nitrogen
containing heteroaryl group such as a pyridine group, wherein the optional substituents are selected from halo, hydroxyl, cyano, C1.7 alkyl, Cu7alkoxy, amino (for example -NH2, C5.6arylamino, Ci_7alkylamino, and di-(Ci.7alkyl)amino), and amido (for example -C02NH2j -C02NHCi.7alkyl, -C02N(Ci-7alkyl)2 and -CONHheterocycyl) and wherein the susbtitutent
30 alkyl, alkoxy, or. aryl groups may be further optionally substituted by one or more groups selected from halo, hydroxyl, C1-7 alkyl, C1.7 alkoxy, C5-6aryl, C5.6arylammo, di-(C].7alkyl)amino and C1-7alkylamino.

WO 2008/023161

PCT/GB2007/003179

In one embodiment, R7 is a pyridinyl group optionally substituted halo, hydroxyl, cyano, C1.7 alkyl, C1-7alkoxy, amino (for example -NH2, Csarylamino, C1-7alkylamino; and di-(Ci_7alkyi)amino), and amido (for example -C02NH2, -C02NHCi.7alkyl, -C02N(C,.7alkyl)2 and -CONHheterocycyl) and wherein the susbtitutent alkyl, alkoxy, or aryl groups may be 5 further optionally substituted by one or more groups selected from halo, hydroxyl, C1.7 alkyl, C1-7 alkoxy, Cs-earyl, Csarylamino, di-(C1-7alkyl)ammo and C1-7-valkylamino.
In one embodiment, R7 is a pyridinyl group optionally substituted with NH2.
Tn one embodiment, R7 is an optionally substituted phenyl group selected from

wherein
Z is H, For OR03;
R03 is selected from hydrogen or an optionally substituted C1-6 alkyl group;
RNl° is selected from hydrogen, C(0)RC23 C(S)RC3, S02RS3, an optionally substituted C5.20
15 heterocyclyl group, an optionally substituted C3.20 aryl group, or an optionally substituted Ct. 10 alkyl group where R and R are selected from H, an optionally substituted C5-20 aryl group, an optionally substituted C5.20 heterocyclyl group, an optionally substituted C1.7 alkyl group or NRNliRNI2, where RNIi and RNI2 are independently selected from H, an optionally substituted C1-7 alkyl group, an optionally substituted C5-20 heterocyclyl group, an optionally
20 substituted C5.2o aryl group or RNn and RN12 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms; and RS3 is selected from H, an optionally substituted C5.20 aryl group, an optionally substituted C5.20 heteroaryl group, or an optionally substituted C1.7 alkyl group; RNl0a is selected from hydrogen or an optionally substituted CMO alkyl group; or

WO 2008/023161

PCT/GB2007/003179

,N10a
iNH)
R and R together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms.
In one embodiment, R7 is an optionally substituted phenyl group selected from


5 wherein
R03 is selected from hydrogen or an optionally substituted Cw alky' group; and Rmo is selected from C(0)RC2, C(S)RC3, S02Rs\ an optionally substituted C5-20 heteroaryl group, an optionally substituted C5.20 aryl group, or an optionally substituted CMO alley 1 group where R02 and RC3 are selected from H, an optionally substituted Cg-20 aryl group, an
10 optionally substituted C5.20 heteroaryl group, an optionally substituted C(.7 alkyl group or NRNURN12, where RN1 * and RNia are independently selected from H, an optionally substituted C1.7 alkyl group, an optionally substituted C5.20 heteroaryl group, an optionally substituted C5-20 aryl group or RNn and RN12 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms; and RS3 is selected from H, an

15 optionally substituted C5.20 aryl group, an optionally substituted C5.20 heteroaryl group, or an optionally substituted C1-7 alkyl group. In one embodiment, R7 is
03.
wherein 20 ZisH.ForOR'
,C2
iNIO
R is selected from hydrogen, C(0)R , an optionally substituted C5-20 heteroaryl group, an optionally substituted C5.20 aryl group, or an optionally substituted CMO alkyl group where RC2 are selected from H, an optionally substituted C5-20 aryl group, an optionally substituted C5.20heterocyclyl group, an optionally substituted C1.7 alkyl group or NRN11RNI2, where RNU

WO 2008/023161

PCT/GB2007/003179

and RN|2 are independently selected from H, an optionally substituted d.? alkyl group, an optionally substituted C5.20 heterocycly group, an optionally substituted C5-20 aryl group or RN1! and RN12 togetlier with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms; 5 Rm0a is selected from hydrogen or an optionally substituted CMO alkyl group; or
RNi0 and RN,0a together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms. In one embodiment, R7 is

10 wherein
Z is H, For OR03;
RN1° is selected from hydrogen, C(0)RC2, an optionally substituted C5-6 heteroaryl group, an
optionally substituted Ce aryl group, or an optionally substituted CMO alkyl group where R02
are selected from CH3 or CH2OH; 15 RN10a is selected from hydrogen or an optionally substituted CMO alkyl group; or
RNI° and RN10a together with the nitrogen to which they are bound form an optionally
substituted heterocyclic ring containing between 3 and 8 ring atoms;
and where the optional subtsituents are selected from cyano, halo, hydroxyl, C1.7alkyloxy,
C1-7alkylamino and di-C1_7alkylamino.
20 In one embodiment, R7 is

wherein
Z is H, For OR03;
Rmo is selected from hydrogen, -C(0)CH3, -C(0)CH2OH, -CH3, -CH2CH3) -CH2CH2OH, 25 -CH(CH3)2, -CH2CH2OMe, -CH2C(CH3)2, -CH2CH2C(CH3)2, -CH(CH3)CH2C(CH3)2,

WO 2008/023161

PCT/GB2007/003179

-CH2CH2CH2N(CH3)2, cycloproyl, cyclopentyl, cyclohexyl, cycloheptyl, -CH2cyclopropyl, methylcyclohexyl, cyanocyclohexyl, pyrazolyl, hydroxypytTolidinyl, -Ctkimidazole ; RNIOa is hydrogen; or
RN1° and RN,0a together with the nitrogen to which they are bound form an optionally 5 substituted heterocyclic ring containing between 5 or 6 ring atoms;
and where the optional subtsituents are selected from halo, hydroxy 1, Ci^alkyloxy.
In a further embodiment of the invention R7 is selected from


WO 2008/023161

PCT/GB2007/003179


R2
5 In one embodiment R2 is OR02 where R02 is an optionally substituted C1-7alkyl group.
In one embodiment R2 is OR02 where R02 is -CH3, -CH2CH3, -CH2CH2OR -CH2CH2OCH3, or -CH(CH3)CH2N(CH3)2.
Preferably R2 is NRN5RN6, where RN5 and RN6 are as previously defined, and more
preferably RND and RN6 together with the nitrogen to which they are bound form a
10 heterocyclic ring containing between 3 and 8 ring atoms, which may optionally be substituted.
The ring preferably has from 5 to 7 ring atoms. Preferred optionally substituted groups
include, but are not limited, to imidazolyl, morpholino, thiomorpholino, piperadinyl,
homopiperadinyl, piperazinyl (preferably N-substituted), homopiperazinyl (preferably N-
substituted) and pyrrolidinyl.
15 Preferred N-substituents for the piperazinyl and homopiperazinyl groups include
esters, in particular, esters bearing a C1.7 alkyl group as an ester substituent, e.g. -C(=0)OCH3, -C(=0)OCH2CH3 and -C(=0)OC(CH3)3.
Preferred N-substituents for the piperazinyl and homopiperazinyl groups include C1_7alkyl groups or esters, in particular, esters bearing a C1.7 alkyl group as an ester 20 substituent, e.g. -C(=0)OCH3, -C(=0)OCH2CH3 and -C(=0)OC(CH3)3.
Preferred C-substituents for the groups include CM alkyl, preferably methyl. The groups may bear one or more substituents, for example one or two substituents'.
Preferred C-substituents for the groups include phenyl, ester, amide and CM alkyl, preferably methyl, aminomethyl, hydroxymethyl or hydroxy ethyl,. The groups may bear one 25 or more substituents, for example one or two substituents.

WO 2008/023161

PCT/GB2007/003179

In one embodiment R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound form a heterocyclic ring containing 5 to 7 ring atoms which may be optionally be substituted, wherein the optional substituents are selected from amino, cyano, halo, hydroxyl, ester, a C3.7 cycloalkyl ring, a C6carboaryI ring, a heterocyclic ring containing 5 5 to 7 ring atoms and C1.7 saturated alkyl and C1.7 saturated alkoxy (wherein the heterocyclic ring, the cycloalkyl ring, the carboaryl ring, the saturated alkyl and alkoxy groups may be optionally substituted by one or more groups selected from halo, hydroxyl, C4.7 alkoxy. amino and Co-6 aryl)
10 In one embodiment R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to
which they are bound form a heterocyclic ring containing between 5 to 7 ring atoms which may be optionally be substituted, wherein the optional substituents are selected from cyano, halo, hydroxyl, and C1-7 saturated alkyl and C1.7 saturated alkoxy (wherein the saturated alkyl and alkoxy groups may be optionally substituted by one or more groups selected from halo,
15 hydroxyl, C1-7 alkoxy, amino and C5-6 aryl)
In one embodiment R2 is NRN5RN6, where RN5 is an optionally substituted C[.7alkyl group or an optionally substituted phenyl group, and RN6 is hydrogen.
In one embodiment R2 is NRN5RN6, where RN5 is -CH(CH3)CH2OCH3, cyclopentyl or a phenyl group, and RN6 is hydrogen.
20 Preferred R2 groups are pyrrolidinyl, morpholino, 'piperadinyl and homopiperadinyl
groups. More preferred groups are morpholino and piperadinyl. These are preferably substituted with one or more alkyl substituents, for example methyl or ethyl substituents. More preferably these are substituted with one or two methyl substituents. If these groups bear two methyl substituents, these are preferably on separate carbon atoms. The alkyl
25 substituents may also be optionally substituted. Examples of optional substituents of the alkyl substitutents include halo, hydroxy, ether or amino. Particularly preferred groups include methylmorpholino groups, dimethylmorpholino groups and methyl piperidinyl groups, for


WO 2008/023161

PCT/GB2007/003179

More preferred groups are morpholino and piperadinyl. These are preferably substituted with one or more alkyl substituents, for example methyl or ethyl substituents. More preferably these are substituted with one or two methyl substituents. If these groups bear two methyl substituents, these are preferably on separate carbon atoms. Particularly 5 preferred groups include methylmorpholino groups, dimethylmorpholino groups and methyl piperidinyl groups, for example:

Preferred R2 groups are pyrrolidinyl, morpholino, piperadinyl and homopiperadinyl groups. More preferred groups are morpholino and piperadinyl. These are preferably
10 substituted with one or more alkyl substituents, for example methyl or ethyl substituents. More preferably these are substituted with one or two methyl substituents. If these groups bear two methyl substituents, these are preferably on separate carbon atoms. The alkyl substituents may also be optionally substituted. Examples of optional substituents of the alkyl substitutents include halo, hydroxy, ether or amino. Particularly preferred groups include
15 methylmorpholino groups, dimethylmorpholino groups and methyl piperidinyl groups, for example:

Further preferred R2 groups are optionally substituted pyrrolidinyl, morpholino, piperadinyl and homopiperadinyl wherein the optional substituents are selected from 20 hydroxyl, C1.7 alkyl, Ci-7alkoxy, amino (for example -NH2, Csarylamino, C].7alkylamino, and di-(Ct.7alkyl)arnino), amido (for example -CONH2, -CONHC1-7alkyl, -CON(C1.7alkyi)2)3 ester (for example -COzC1-7alkyl), Cearyl and 3 to 7 membered heterocyclyl group and wherein the susbtitutent alkyl, alkoxy, aryl or heterocyclyl groups may be further optionally substituted by one or more groups selected from halo, hydroxyl, C1.7 alkyl, C1-7 alkoxy, -NH2,

WO 2008/023161

PCT/GB2007/003179

di-(Ci-7alkyl)amino and C17.Talkylamino. More preferred groups are morphobno, piperadinyl
and homopiperadinyl which may be optionally substited by one or more groups selected from
hydroxyl, methyl, ethyl, -C02Me, -C02Et, -CH2OH, -CH2Ome, -CH2NMe2! -CONH2,
-CONHMe, -CONMe2, phenyl, pyrrolidinyl, morphobno and piperadinyl.
5 In a further embodiment of the invention R2 is selected from


WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179


In an embodiment of the invention, there is provided a subset of compounds of
formula (I) or (la), and pharmaceutically acceptable salts thereof, in which:
10 only one of X5, X6 and Xs is N;

WO 2008/023161

PCT/GB20II7/003179

R7 is selected from an optionally substituted C5-20 aryl group, an optionally substituted 5- to 20-membered heteraryl group, OR01, NRN1RN2, NRN7aC(=0)Rcl and NRN7bS02RS2a; and
R2 is selected from OR02, NRN5RN6, an optionally substituted C5.20 heteroaryl group,
5 and an optionally substituted 5-20 aryl group.
In another embodiment, there is provided a subset of compounds of formula (I) or (la), and pharmaceutically acceptable salts thereof, in which: only one of X5, X6 and X8 is N; R7 is an optionally substituted C5_6 aryl group or an optionally substituted 5 or 6
10 membered heteraryl group, wherein the optional substituents are selected from halo,
hydroxyl, cyano, C1-7 alkyl, d-7alkoxy, amino (for example -NH2, C5.6arylamino, C1-7alkylamino, and di-(C1_7aucyl)amino), and amido (for example -CONH2, -CONHC].7alkyl, -CON(C,.7alkyl)2 and -CONHheterocycyl) and wherein the susbtitutent allcyl, alkoxy, or aryl groups may be further optionally substituted by one
15 or more groups selected from halo, hydroxyl, C1.7 alkyl, C1-7 alkoxy, C5-6aryl,
Cs-earylamino, di-(Ci-7alkyl)amino and C{.7alkylamino; and
R2 is selected from OR02, NRN5RN6, an optionally substituted C5-6 heteroaryl group, and an optionally substituted d aryl group. In another embodiment, there is provided a subset of compounds of formula (I) or (la),
20 and pharmaceutically acceptable salts thereof, in which; only one of X5, X6 and X8 is N;
R7 is an optionally substituted Cg^ aryl group or an optionally substituted 5 or 6 membered heteraryl group, wherein the optional substituents are selected from halo, hydroxyl, cyano, C1-7 alkyl, C1.7alkoxy, amino (for example -NH2, d^arylamino,
25 Ci-7alkylamino, and di-(Ci.7alkyl)amino), and amido (for example -CONH2,
-CONHC1.7alkyl, -CON(C1.7alkyl)2 and -CONHheterocycyl) and wherein the susbtitutent alkyl, alkoxy, or aryl groups may be further optionally substituted by one or more groups selected from halo, hydroxyl, C17 alkyl, C1-7 alkoxy, C5-6aryl, d-earyl amino, di-(d-7alkyI)amino and C[.7alkylamino; and
30 R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound
form a heterocyclic ring containing between 5 to 7 ring atoms which may be optionally be substituted, wherein the optional substituents are selected from cyano,

WO 2008/023161

PCT/GB20W7/003179

halo, hydroxyl, and C1.7 saturated alkyl and C1-7 saturated alkoxy (wherein the saturated alkyl and alkoxy groups may be optionally substituted by one or more groups selected from halo, hydroxyl, C1.7 alkoxy, amino and C5.6 aryl). In a further embodiment, there is provided a subset of compounds of formula (I) or 5 (la), and pharmaceutically acceptable salts thereof, in which: only one of X5, X6 and X8 is N;
R7 is an optionally substituted C5.6 aryl group or an optionally substituted 5 or 6 membered heteraryl group, wherein the optional substituents are selected from halo, hydroxyl, cyano, C1.7 alkyl, C^alkoxy, amino (for example -NH2, Csarylamino,
10 C1.7alkylamino} and di-(C1.7alkyl)amino), and amido (for example -CONH2,
-CONHCi.yalkyl, -CON(C1.7alkyl)2 and -CONHheterocycyl) and wherein the susbtitutent alkyl, alkoxy, or aryl groups may be further optionally substituted by one or more groups selected from halo, hydroxyl, C1-7 alkyl. C1.7 alkoxy, C5-6aryl, Cs^arylamino, di-(Ci.7alkyl)amino and Chalkylamino; and
15 R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound
form an optionally substituted imidazolyl, morpholino, thiomorpholino, piperadinyl, homopiperadinyl, piperazinyl (preferably N-substituted), homopiperazinyl (preferably N-substituted) or pyrrolidinyl, wherein optional N-substituents on the piperazinyl and homopiperazinyl groups include Ci.7alkyl groups or esters, in particular, esters bearing
20 a C[-7 alkyl group as an ester substituent, e.g. -C(=0)OCH3, -C(=0)0CHZCH3 and
-C(=0)OC(CH3)3, and optional C-substituents for the imidazolyl, morpholino, thiomorpholino, piperadinyl, homopiperadiny], piperazinyl, homopiperazinyl or pyrrolidinyl groups include phenyl, ester, amide and Ci_4 alkyl, preferably methyl, aminomethyl, hydroxymethyl or hydroxyethyl.
25 In an embodiment of the invention, there is provided a subset of compounds of
formula (I), (la) or (lb), and pharmaceutically acceptable salts thereof, in which: only one of X5, X6 and X8 is N;
R7 is selected from an optionally substituted C5.20 aryl group, an optionally substituted 5- to 20-membered heteraryl group, OR01, NRNIRN2, NRN7flC(=0)Rc' and
30 NRN7bS02RS2a; and
R2 is selected from OR02, NR^R*6, an optionally substituted C5-20 heteroaryl group, and an optionally substituted C5-20 aryl group.

WO 2008/023161

PCT7GB2007/003179

In another embodiment, there is provided a subset of compounds of formula (I), (la) or (lb), and pharmaceutically acceptable salts thereof, in which: only one of X3, X6 and X8 is N;
R7 is an optionally substituted C5_6 aryl group or an optionally substituted 5 or 6
5 membered heteraryl group, wherein the optional substituents are selected from halo,
hydroxyl, cyano, C1.7 alkyl, C[.7aIkoxy, amino (for example -NH2, Csaryiamino, C^alkylamino, and di-(Ci.7alkyl)aniino), and amido (for example -CONH2, -CONHCi.7alkyL, -CON(Ci-7alkyl)2 and -CONHheterocycyl) and wherein the susbtitutent alkyl, alkoxy, or aryl groups may be further optionally substituted by one
10 or more groups selected from halo, hydroxyl, C1-7 alkyl, C1.7 alkoxy, C5-6aryl,
Cs^arylamino, di-(C[.7alkyl)amino and C1.7alkylamino; and
R2 is selected from OR02, NRN5RN6, an optionally substituted C5-6 heteroaryl group, and an optionally substituted Ce aryl group. In another embodiment, there is provided a subset of compounds of formula (I), (la) or
15 (lb), and pharmaceutically acceptable salts thereof, in which: only one of X5, X6 and X8 is N;
R7 is an optionally substituted C5-6 aryl group or an optionally substituted 5 or 6 membered heteraryl group, wherein the optional substituents are selected from halo, hydroxyl, cyano, C1.7 alkyl, Ci^alkoxy, amino (for example -NH2 C5-6aryIamino,
20 C].7alkylamino, and di-(C1.7alkyl)amino), and amido (for example -CONH2,
-CONHC1-7alkyl, -CON(C1.7alkyl)2 and -CONHheterocycyl) and wherein the susbtitutent alkyl, alkoxy, or aryl groups may be further optionally substituted by one or more groups selected from halo, hydroxyl, Cj_7 alkyl, C1.7 alkoxy, Cs-garyl, C5-6arylamino» di-(C1.7alkyl)amino and Chalkylamino; and
25 R2 is NRN5RN6 where RNS and RN6 together with the nitrogen to which they are bound
form a heterocyclic ring containing between 5 to 7 ring atoms which may be optionally be substituted, wherein the optional substituents are selected from cyano, halo, hydroxyl, and C1.7 saturated alkyl and C1.7 saturated alkoxy (wherein the saturated alkyl and alkoxy groups may be optionally substituted by one or more
30 groups selected from halo, hydroxyl, C1.7 alkoxy, amino and C5.6 aryl).
In a further embodiment, there is provided a subset of compounds of formula (I), (la) or (lb), and pharmaceutically acceptable salts thereof, in which:

WO 2008/023161

PCT/GB2007/003179

only one of X5, X6 and X8 is N;
R7 is an optionally substituted C5-6 aryl group or an optionally substituted 5 or 6
membered heteraryl group, wherein the optional substituents are selected from halo.
hydroxy 1, cyano, C1.7 alkyl, C^alkoxy, amino (for example -NH2i C5_6arylamino,
5 Chalky lamino, and di-(C1.7alkyl)amino), and arnido (for example -CONH2,
-CONHCi.7alkyl, -CON(C1-7alkyl)2 and -CONHheterocycyl) and wherein the susbtitutent alkyl, alkoxy, or aryl groups may be further optionally substituted by one or more groups selected from halo, hydroxy!, C1.7 alkyl, C1.7 alkoxy, Csaryl, C^arylamino, di-(Ci.7alkyl)amino and C1..7alkylamino; and
10 R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound
form an optionally substituted imidazolyl, morpholino, thiomorpholino, piperadinyl, homopiperadinyl, piperazinyl (preferably N-substituted), homopiperazinyl (preferably N-substituted) or pyrroUdinyl, wherein optional N-substituents on the piperazinyl and homopiperazinyl groups include Ci^alkyl groups or esters, in particular, esters bearing
15 a C].7 alkyl group as an ester substituent, e.g. -C(=0)OCH3, -C(=0)OCH2CH3 and
-C(=0)OC(CH3)3, and optional C-substituents for the imidazolyl, morpholino, thiomorpholino, piperadinyl, homopiperadinyl, piperazinyl, homopiperazinyl or pyrrolidinyl groups include phenyl, ester, amide and CM alkyl, preferably methyl, aminomethyl, hydroxymethyl or hydroxyethyl.
20 In a further embodiment, there is provided a subset of compounds of formula (I), (la)
or (lb), and pharmaceutically acceptable salts thereof, in which: X5 and X6 are each CH; X8isN; R7 is an optionally substituted phenyl or pyridinyl group, wherein the optional
25 substituents are preferably selected from fluoro, hydroxyl, cyano, nitro, methyl,
methoxy, -OCH2CH3, -NH2, -NHS02CH3, -CH2NHS02CH3! -OCHF2) -CH2OH, -C02H -CONHj, -CONHMe, -CONHEt, -CONHCH(CH3)2j -CONHCH2CH2F, -CONHCH2CHF2, -CONHCH2CH2OH, -CONMeEt, -CONMe2, N-methylpiperazinylcarbonyl and 4-hydroxypiperidinylcarbonyl; and
30 R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound
form a heterocyclic ring containing 5 to 7 ring atoms which may be optionally be substituted, wherein the optional substituents are selected from amino, cyano, halo,

WO 2008/023161

PCT/GB2007/003179

hydroxyl, ester, a C3-7 cycloalkyl ring, a Cecarboaryl ring, a heterocyclic ring
containing 5 to 7 ring atoms and C1_7 saturated alkyl and C].7 saturated alkoxy
(wherein the heterocyclic ring, the cycloalkyl ring, the carboaryl ring, the saturated
alkyl and alkoxy groups may be optionally substituted by one or more groups selected
5 from halo, hydroxyl, C1.7 alkoxy, amino and C5-6 aryl).
In a further embodiment, there is provided a subset of compounds of formula (I), (la) or (lb), and pharmaceutically acceptable salts thereof, in which: X5 and X6 are each CH;X8 is N; R7 is an optionally substituted phenyl or pyridinyl group, wherein the optional
10 substituents are preferably selected from fluoro, hydroxyl, cyano, nitro, methyl,
methoxy, -OCH2CH3, -NH2, -NHS02CH3, -CH2NHS02CH3, -OCHF2, -CH2OH, -C02H -CONH2, -CONHMe, -CONHEt, -CONHCH(CH3)2, -CONHCH2CH2F, -CONHCH2CHF2, -CONHCH2CH2OH, -CONMeEt, -CONMe2, N-methylpiperazinylcarbonyl and 4-hydroxypiperidinylcarbonyl; and
15 R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound
form an optionally substituted imidazolyl, morpholino, thiomorpholino, piperadinyl, homopiperadinyl, piperazinyl (preferably N-substituted), homopiperazinyl (preferably N-substituted) or pyrrolidinyl, wherein optional N-substituents on the piperazinyl and homopiperazinyl groups include Ci-7alkyl groups or esters, in particular, esters bearing
20 a C1.7 alkyl group as an ester substituent, e.g. -C(=0)OCH3, -C(=0)OCH2CH3 and
-C(=0)OC(CH3)3, and optional C-substituents for the imidazolyl, morpholino, thiomorpholino, piperadinyl, homopiperadinyl, piperazinyl, homopiperazinyl or pyrrolidinyl groups include phenyl, ester, amide and CM alkyl, preferably methyl, aminomethyl, hydroxymethyl or hydroxyethyl.
25 In a further embodiment, there is provided a subset of compounds of formula (I) or
(la), and pharmaceutically acceptable salts thereof, in which: X5 and X6 are each CH; X8isN; R7 is an optionally substituted phenyl or pyridinyl group, wherein the optional
30 substituents are preferably selected from -NH2) fluoro, hydroxyl, cyano, nitro, methyl,
methoxy, -CH2OH, -CQ2H -CONH2, -CONHMe, -CONHEt, -C0NHCH2CHZF,

WO 2008/023161

PCT/GB2007/003179

-CONHCH2CHF2, -CONHCH2CH2OH, -CONMeEt, -CONMe2; N-
methylpiperazinylcarbonyl and 4-hydroxypiperidinyIcarbonyl; and
R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound
form an optionally substituted imidazolyl, morpholino, thiomorpholino, piperadinyl,
5 homopiperadinyl, piperazinyl (preferably N-substituted), homopiperazinyl (preferably
N-substituted) or pyrrolidinyl, wherein optional N-substituents on the piperazinyl and homopiperazinyl groups include Ci.ialkyi groups or enters, in particular, esters bearing a Ci-7 alkyl group as an ester substituent, e.g. -C(=*0)OCH3, -C(=0)OCH2CH3 and -C(=0)OC(CH3)3, and optional C-substituents for the imidazolyl, morpholino,
10 thiomorpholino, piperadinyl, homopiperadinyl, piperazinyl, homopiperazinyl or
pyrrolidinyl groups include phenyl, ester, amide and Ci^ alkyl, preferably methyl, aminomethyl, hydroxymethyl or hydroxyethyl.
In a further embodiment, there is provided a subset of compounds of formula (I), (la) or (lb), and pharmaceutically acceptable salts thereof, in which:
15 X5 and X6 are each CH;
X8 is N;
R7 is an optionally substituted phenyl or pyridinyl group, wherein the optional substituents are preferably selected from -NH2, fluoro, hydroxyl, cyano, nitro, methyl, methoxy, -CH2OH, -C02H -CONH2, -CONHMe, -CONHEt, -CONHCH2CH2F,
20 -CONHCH2CHF2, -CONHCH2CH2OH, -CONMeEt, -CONMe2, N-
methylpiperazinylcarbonyl and 4-hydroxypiperidinylcarbonyl; and R2 is NRN5RN6 where RN5 and RNfi together with the nitrogen to which they are bound form an optionally substituted imidazolyl, morpholino, thiomorpholino, piperadinyl, homopiperadinyl, piperazinyl (preferably N-substituted), homopiperazinyl (preferably
25 N-substituted) or pyrrolidinyl, wherein optional N-substituents on the piperazinyl and
homopiperazinyl groups include Ci^alkyl groups or esters, in particular, esters bearing a C1.7 alkyl group as an ester substituent, e.g. -C(=0)OCH3, -C(=0)OCH2CH3 and -C(=0)OC(CH3)3, and optional C-substituents for the imidazolyl, morpholino, thiomorpholino, piperadinyl, homopiperadinyl, piperazinyl, homopiperazinyl or
30 pyrrolidinyl groups include phenyl, ester, amide and CM alkyl, preferably methyl,
aminomethyl, hydroxymethyl or hydroxyethyl,

WO 2008/023161

PCT/GB2007/003179



10

In a further embodiment, there is provided a subset of compounds of formula (I), (la) or (lb), and pharmaceutically acceptable salts thereof, in which: X5 and X6 are each CH; X8isN;
R7 is an optionally substituted phenyl or pyridinyl group, wherein the optional substituents are preferably selected from fluoro, hydroxyl, cyano, nitro, methyl, methoxy, -OCH2CH3, -NH2, -NHS02CH3, -CH2NHS02CH3, -OCHF2, -CH2OH, -C02H -CONH2, -CONHMe, -CONHEt, -CONHCH(CH3)2, -CONHCH2CH2F, ^CONHCH2CHF2, -CONHCH2CH2OH, -CONMeEt, -CONMe2, N-methylpiperazinylcarbonyl and 4-hydroxypiperidinylcarbonyl; and R2 is a group selected from





WO 2008/023161

PCT/GB2007/003179


In a further embodiment, there is provided a subset of compounds of formula (I) or 5 (la), and pharmaceutical^ acceptable salts thereof, in which: Xs and X6 are each CH; XsisN;
R7 is an optionally substituted phenyl or pyridinyl group, wherein the optional
substiruents are preferably selected from -NH2. fluoro, hydroxy!, cyano, nitro, methyl,
10 methoxy, -CH2OH, -C02H -CONH2, -CONHMe, -CONHEt, -CONHCH2CH2F,
-CONHCH2CHF2) -CONHCH2CH2 OH, -CONMeEt, -CONMe2, N-methylpiperazinylcarbonyl and 4-hydroxypiperidinylcarbonyl; and R2 is a group selected from


WO 2008/023161

PCT/GB2007/003179


Tn a further embodiment, there is provided a subset of compounds of formula (I), (la) or (lb), and pharmaceutically acceptable salts thereof, in which: X5 and X6 are each CH;
XBisN;

WO 2008/023161

PCT/GB2007/003179

R7 is an optionally substituted phenyl or pyridinyl group, wherein the optional substituents are preferably selected from-NH2, fluoro, hydroxyl, cyano, nitro, methyl, methoxy, -CH2OH, -C02H -CONH2, -CONHMe, -CONHEt, -CONHCH2CH2F, -CONHCH2CHF2s -CONHCH2CH2OH, -CONMeEt, -CONMe2, N-methylpiperazinylcarbonyl and 4-hydroxypiperidinylcarbonyl; and R2 is a group selected from


WO 2008/023161

PCT/GB2007/003179






10

In a further embodiment, there is provided a subset of compounds of formula (I), (la) or (lb), and pharmaceutical^ acceptable salts thereof, in which: X5 and X6 are each CH; X8isN;
R7 is a 4-chlorophenyl, 4-methylphenyl, 4-methoxyphenyl, 3-hydroxy methy 1-4-methoxy-pheny 1, 3,5-dimethoxy-4-hydroxyphenyI, 4-hydrox yphenyl, 3 -hydroxyphenyl or a 3-hydroxymethylphenyl group; and R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound




o
15


iorm a w group.
In a further embodiment, there is provided a subset of compounds of formula (I) or (la), and pharmaceutically acceptable salts thereof, in which: X5 and X6 are each CH; X8isN;
R7 is a 4-chlorophenyl, 4-methylphenyl, 4-methoxyphenyl, 3-hydroxymethyl-4-
metboxy-phenyl, 3,5-dimethoxy-4-hydroxyphenyl, 4-hydroxy phenyl, 3-
hydroxyphenyl or a 3-hydroxymethylphenyI group; and R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound




20 form a

WO 2008/023161

PCT/GB2007/003179

In a fuither embodiment, there is provided a subset of compounds of formula (I), (la) or (lb), and pharmaceutically acceptable salts thereof, in which: X5 and X6 are each CH; X8isN;
R7 is a 4-chlorophenyl, 4-methylphenyl, 4-methoxy phenyl, 3-hydroxymethyl-4-methoxy-phenyl, 3,5-dimethoxy-4-hydroxyphenyls 4-hydroxyphenyl, 3-hydroxyphenyl or a 3-hydroxymethylphenyl group; and R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound


forma
10 In a further embodiment, there is provided a subset of compounds of formula (I), (la)
or (lb), and pharmaceutically acceptable salts thereof, in which: X5 and X6 are each CH;
X8 is N;


15

WO 20(18/023161

PCT/GB2007/00J179


group; ana
5 R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound
forma

In an embodiment of the invention, there is provided a subset of compounds of 10 formula (II) or (Ila), and pharmaceuticaly acceptable salts thereof,

WO 2008/023161 PCT/GB2007/003179

wherein:
only one of X5, X6 and X8 is N, and the others are CH;
Z is H, For OR03;
5 RN1° is selected from hydrogen, C(0)RC2, an optionally substituted C5-20 heteroaryl
group, an optionally substituted C5.20 aryl group, or an optionally substituted CMO
alkyl group where R02 are selected from H, an optionally substituted C5-20 aryl group,
an optionally substituted C5.20 heterocyclyl group, an optionally substituted C1-7 alkyl
group or NRNllRNI2, where RNU and RNI2 are independently selected from H, an
10 optionally substituted C1.7 alkyl group, an optionally substituted C5-20 heterocycly
group, an optionally substituted C5.2o aryl group or RNn and RNI2 together with the
nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8
ring atoms;
RN10a is selected from hydrogen or an optionally substituted C1-10 alkyl group; or
15 RN1° and RN,0a together with the nitrogen to which they are bound form an optionally
substituted heterocyclic ring containing between 3 and 8 ring atoms;
R03 is an optionally substituted d-g alkyl group; and
R2 is selected from NRN5RN6, an optionally substituted C5.20 heteroaryl group, and an
optionally substituted C3-20 aryl group.
20 In another embodiment, there is provided a subset of compounds of formula (II) or
(Ha), and pharmaceutical^ acceptable salts thereof, in which:
only one of X5, X6 and X8 is N, and the others are CH:
Z is H, For OR03
RNIO ig RNIO ig seiected from hydrogen, C(0)RC2, an optionally substituted C5.6
25 heteroaryl group, an optionally substituted Ce aryl group, or an optionally substituted
CMO alkyl group where RC2 are selected from CH3 or CH2OH where the optional
subtsituents are selected from cyano, halo, hydroxyl, C^alkyloxy, Chalkylamino and
di-Cwaikylamino;

WO 2008/023161

PCT/GB2O07/003179

RNl0a is selected from hydrogen or an optionally substituted CMO alkyl group where
the optional subtsituents are selected from cyano, halo, hydroxyl, C(.alkyloxy,
Ci^alkylamino and di-C].7alkylamino; or
RNI° and RN10a together with the nitrogen to which they are bound form an optionally
5 substituted heterocyclic ring containing between 3 and 8 ring atoms, where the
optional subtsituents are selected from cyano, halo, hydroxyl, C^alkyloxy,
C].7alkylamino and di-C1-7alkylamino;
R°3 is an unsubstituted C1.3 alkyl group; and
R2 is selected from NRN5RN6, an optionally substituted C5.6 heteroaryl group, and an
10 optionally substituted C6 aryl group.
In another embodiment, there is provided a subset of compounds of formula (II) or (Ha), and pharmaceutically acceptable salts thereof, in which:
only one of X5, X6 and X8 is N, and the others are CH;
Z is H, For OR03
15 RN1° is selected from hydrogen, -C(0)CH3, -C(0)CH2OH: -CH3, -CH2CH3,
-CH2CH2OH, -CH(CH3)2, -CH2CH2OMe, -CH2C(CH3)2, -CH2CH2C(CH3)2l
-CH(CH3)CH2C(CH3)2, -CH2CH2CH2N(CH3)2, cycloproyl, cyclopentyl, cyclohexyl,
cycloheptyl, -CH2cyclopropyl, methylcyclohexyl, cyanocyclohexyl, pyrazolyl,
hydroxypyiTolidinyl, -CH2imidazole ;
20 RN10a is hydrogen; or
RN1° and RNl0a together with the nitrogen to which they are bound form an optionally
substituted heterocyclic ring containing between 5 or 6 ring atoms, where the optional
subtsituents are selected from halo, hydroxyl, Ci.7alkyloxy;
R03 is a methyl group; and
25 R2 is NRN5RK6 where RN5 and RN6 together with the nitrogen to which they are bound
form a heterocyclic ring containing between 5 to 7 ring atoms which may be
optionally be substituted, wherein the optional substituents are selected from cyano,
halo, hydroxyl, and C1.7 saturated alkyl and C1.7 saturated alkoxy (wherein the
saturated alkyl and alkoxy groups may be optionally substituted by one or more
30 groups selected from halo, hydroxyl, C1-7 alkoxy, amino and C5.6 aryl).
In a further embodiment, there is provided a subset of compounds of formula (II) or (Ila), and pharmaceutically acceptable salts thereof, in which:

WO 2008/023161

PCT/GB2007/003179

only one of X3, X6 and X8 is N, and the others are CH;
Z is H, For OR03
Rmo is selected from hydrogen, -C(0)CH3, -C(0)CH2OH, -CH3, -CH2CH3,
-CH2CH2OH, -CH(CH3)2, -CH2CH2OMe, -CH2C(CH3)2, ~CH2CH2C(CH3)2,
5 -CH(CH3)CH2C(CH3)2, -CH2CH2CH2N(CH3)2, cycloproyl, cyclopentyl, cyclohexyl,
cycloheptyl, -CH2cyclopropyI, methylcyclohexyl, cyanocyclohexyl, pyrazolyl,
hydvaxypyrroliduiyl, -CHiimidazole;
RN10a is hydrogen; or
RNI° and RNIOa together with the nitrogen to which they are bound form an optionally
10 substituted heterocyclic ring containing between 5 or 6 ring atoms, where the optional
subtsituents are selected from halo, hydroxyl, Ci^alkyloxy;
R03 is a methyl group; and
R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound
form an optionatty subslita'teu xnn6azcftly morpholino thiropipephoting, pleperadiny,
15 homopiperadinyl, piperazinyl (preferably N-substituted), homopiperazinyl (preferably
N-substituted) or pyrrolidinyl, wherein optional N-substituents on the piperazinyl and
homopiperazinyl groups include Ci^alkyl groups or esters, in particular, esters bearing
a C|.7 alkyl group as an ester substiruent, e.g. -C(^0)OCH3, -C(=0)OCH2CH3 and
-C(=0)OC(CH3)3, and optional C-substituents for the iraidazolyl, morpholino,
20 thiomorpholino, piperadinyl, homopiperadinyl, piperazinyl, homopiperazinyl or
pyrroBdinyl groups include phenyl, ester, amide and CM alkyl, preferably methyl,
aminomethyl, hydroxymethyl or hydroxyethyl.
In a further embodiment, there is provided a subset of compounds of formula (II) or
(Ha), and pharmaceutical^ acceptable salts thereof, in which:
25 X5 and X6 are each CH;
X8isN;
ZisH5ForOR°3
Rmo is selected from hydrogen, -C(0)CH3, -C(0)CH2OH, -CH3t -CH2CH3,
-CH2CH2OH, -CH(CH3)2, -CH2CH2OMe, -CH2C(CH3)2, -CH2CH2C(CH3)23
30 -CH(CH3)CH2C(CH3)2, -CH2CH2CH2N(CH3)2, cycloproyl, cyclopentyl, cyclohexyl,
cycloheptyl, -CH2cyclopropyl, methylcyclohexyl, cyanocyclohexyl, pyrazolyl,
hydroxypyrrolidinyl, -CH2hnidazole;

WO 2008/023161

PCT/GB2007/003179

RMVUa is hydrogen; or
RNi0 and RNIOa together with the nitrogen to which they are bound form an optionally
substituted heterocyclic ring containing between 5 or 6 ring atoms, where the optional
subtsituents are selected from halo, hydroxyl, C|_7alkyloxy;
5 R 3 is a methyl group; and
R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound
form an optionally substituted morpholino, thiomorpholino, piperidinyl,
homopiperidinyl, piperazinyl (preferably N-substituted), homopiperazinyl (preferably
N- substituted) or pyrrolidinyl group, wherein the optional substituents are selected
10 from cyano, halo, hydroxyl, and C1.7 saturated alkyl and C1-7 saturated alkoxy
(wherein the saturated alkyl and alkoxy groups may be optionally substituted by one
or more groups selected from halo, hydroxyl, C1-7 alkoxy, amino and C5.6 aryl).
In a further embodiment, there is provided a subset of compounds of formula (II) or
(Ha), and pharmaceutical acceptable salts thereof, in which:
15 X5 and X6 are each CH;
X8isN;
Z is H, For OR03
RN1° is a RN1° is selected from hydrogen, -C(0)CH3, -C(0)CH2OH, -CH3, -CH2CH3)
-CH2CH2OH, -CH(CH3)2, -CH2CH2OMe, -CH2C(CH3)2, -CH2CH2C(CH3)2,
20 -CH(CH3)CH2C(CH3)2, -CH2CH2CH2N(CH3)2, cycloproyl, cyclopentyl, cyclohexyl,
cycloheptyl, -CH2cyclopropyl, methylcyclohexyl, cyanocyclohexyl, pyrazolyl,
hydroxypyrrolidinyl, -CH2imidazole;
RNIOn is hydrogen; or
RNi0 and RN10a together with the nitrogen to which they are bound form an optionally
25 substituted heterocyclic ring containing between 5 or 6 ring atoms, where the optional
subtsituents are selected from halo, hydroxyl, C1-7alkyloxy;
R03 is a methyl group; and
R2 is a group selected from

WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179


In a further embodiment, there is provided a subset of compounds of formula (H) or (Ila), and pharrhaceutically acceptable salts thereof, in which: X5 and X6 are each CH;
X8isN;

03
ZisH,ForOR'
RN1° is selected from hydrogen, -C(0)CH3, -C(0)CH2OH, -CH3, -CH2CH3,
-CH2CH2OH, -CH(CH3)2, -CH2CH2OMe, -CH2C(CH3)2, -CH2CH2C(CH3)2,
-CH(CH3)CH2C(CH3)2, -CH2CH2CH2N(CH3)2, cycloproyl, cyclopentyl, cyclohexyl,
10
cycloheptyl, -CH2cyclopropyl, methylcyclohexyl, cyanocyclohexyl. pyrazolyl,
hydroxypyrrolidinyl, -CH2imidazole;
RNIOa is hydrogen; or
RNI° and RN10a together with the nitrogen to which they are bound form an optionally
substituted heterocyclic ring containing between 5 or 6 ring atoms, where the optional
15
subtsituents are selected from halo, hydroxyl, C1-7alkyloxy;
R03 is a methyl group; and
R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound

In a further embodiment, there is provided a subset of compounds of formula (II) or 20 (Ila), and pharmaceutically acceptable salts thereof, in which: X5 and X6 are each CH; X8isN: ZisH, ForOR°3

WO 201)8/023161

PCT/GB2007/003179

Rmo is selected from hydrogen, -C(0)CH3, -C(0)CH2OH, -CH3, -CH2CH3,
-CH2CH2OH, -CH(CH3)2j -CH2CH2OMe, -CH2C(CH3)2, -CH2CH2C(CH3)2,
-CH(CH3)CH2C(CH3)2, -CH2CH2CH2N(CH3)2, cycloproyi, cyclopentyl, cyclohexyl,
cycloheptyl, -CH2cyclopropyl, methylcyclohexyl, cyanocyclohexyl, pyrazolyl,
5 hydroxypyrrolidinyl, -CH2imidazole;
RNJOa is hydrogen; or
Rmo and RN10a together with the nitrogen to which they are bound form an optionally
substituted heterocyclic ring containing between 5 or 6 ring atoms, where the optional
subtsituents are selected from halo, hydroxyl, Ci.7alkyloxy;
10 R03 is a methyl group; and
R2 is NRN5RN6 where RN3 and RN6 together with the nitrogen to which they are bound


In an embodiment of the invention, there is provided a subset of compounds of
formula (I), (la) or (lb) wherein the compound is a compound of formula (II). (Ila) or (lib),
15 and pharmaceutical^ acceptable salts thereof,
,0,
N
DN10 _N10a v5 I

•R2
z
N ■'" N*
wherein:
only one of X5, X6 and Xs is N, and the others are CH;
20 Z is H, For OR03;

R2

WO 2008/023161

PCT/GB2007/003179

RN1° is selected from hydrogen, C(0)RC2, an optionally substituted C5-20 heteroaryl
group, an optionally substituted C5.20 aryl group, or an optionally substituted Ci_l0
alkyl group where RC2 are selected from H, an optionally substituted C5.20 aryl group,
an optionally substituted C5.20 heterocyclyl group, an optionally substituted C1-7 alkyl
5 group or NRN11RN12, where RNU and RN12 are independently selected from H, an
optionally substituted C1-7 alkyl group, an optionally substituted 05.20 heterocycly group, an optionally substituted C5.20 aryl group or RNU and RN12 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms;
10 RNIOa is selected from hydrogen or an optionally substituted CMO allcyl group; or
RN1° and RN10a together with the nitrogen to which they are bound form an optionally
substituted heterocyclic ring containing between 3 and 8 ring atoms;
R03 is an optionally substituted C1-7alkyl group; and
R2 is selected from NRN5RNd, an optionally substituted C5-20 heteroaryl group, and an
15 optionally substituted C5-20 aryl group.
In another embodiment, there is provided a subset of compounds of formula (II), (Ha) or (lib), and pharmaceutical^ acceptable salts thereof, in which: only one of X5, X6 and X8 is N, and the others are CH; Z is H, For OR03
20 RN'° is RN'° is selected from hydrogen, C(0)RC2, an optionally substituted C5.6
heteroaryl group, an optionally substituted C6 aryl group, or an optionally substituted CMO alkyl group where RC2 are selected from CH3 or CH2OH where the optional subtsituents are selected from cyano, halo, hydroxyl, C1.7alkyloxy, Chalky lamino and di-Ci.7alkylamino;
25 |^Ni0a -S se]ec(e£j from hydrogen or an optionally substituted C1-7 alkyl group where
the optional subtsituents are selected from cyano, halo, hydroxyl, Ci^alkyloxy, Ci_7alkylamino and di-Ci-7alkylamino; or
RN1° and RN10a together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms, where the
30 optional subtsituents are selected from cyano, halo, hydroxyl, C1-7alkyloxy,
C1-7alkylamino and di-C1-7alkylamino; R03 is an unsubstituted C1.3 alkyl group; and

WO 2008/023161

PCT/GB2007/003179

R2 is selected from NRN5RN6, an optionally substituted C5-6 heteroaryl group, and an optionally substituted Cg aryl group.
In another embodiment, there is provided a subset of compounds of formula (II), (Ila)
or (lib), and pharmaceuticaUy acceptable salts thereof, in which:
5 only one of X5, X6 and Xs is N, and the others are CH;
ZisHsForOR03
RNl° is selected from hydrogen, -C(0)CH3) -C(0)CH2OH, -CH3, -CH2CH3, -CH2CH2OH, -CH(CH3)2; -CH2CH2OMe, -CH2C(CH3)2, -CH2CH2C(CH3)2) -CH(CH3)CH2C(CH3)2, -CH2CH2CH2N(CH3)2, cycloproyl, cyclopentyl, cyclohexyl,
10 cycloheptyl, -CH2cyclopropyl, raethylcyclohexyl, cyanocyclohexyl, pyrazolyl,
hydroxypyrrolidinyl, -CH2imidazole; RNl0a is hydrogen; or
RNI° and RNl0a together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 5 or 6 ring atoms, where the optional
15 subtsituents are selected from halo, hydroxy!, Ci^alkyloxy;
R03 is a methyl group; and
R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound form a heterocyclic ring containing between 5 to 7 ring atoms which may be optionally be substituted, wherein the optional substituents are selected from cyano,
20 halo, hydroxyl, and C1-7 saturated alkyl and C1.7 saturated alkoxy (wherein the
saturated alkyl and alkoxy groups may be optionally substituted by one or more groups selected from halo, hydroxyl, C1.7 alkoxy, amino and C5.6 aryl). In a further embodiment, there is provided a subset of compounds of formula (II), (Ila) or (lib), and pharmaceuticaUy acceptable salts thereof, in which:
25 only one of X5, X6 and X8 is N, and the others are CH;
Z is H, F or OR03
RN,° is selected from hydrogen, -C(0)CH3s -C(0)CH2OH, -CH3, -CH2CH3, -CH2CH2OH, -CH(CH3)2, -CH2CH2OMe, ~CH2C(CH3)2) -CH2CH2C(CH3)2: -CH(CH3)CH2C(CH:,)2, -CH2CH2CH2N(CH3)2, cycloproyl, cyclopentyl, cyclohexyl,
30 cycloheptyl, -CH2cyclopropyl, methylcyclohexyl, cyanocyclohexyl, pyrazolyl,
hydroxypyrrolidinyl, -CH2imidazole; RN10a is hydrogen; or

WO 2008/0231fil

PCT7GB2007/003179

RN,° and RNt0a together with the nitrogen to which they are bound form an optionally
substituted heterocyclic ring containing between 5 or 6 ring atoms, where the optional
subtsituents are selected from halo, hydroxyl, Ci.7alkyloxy;
R03 is a methyl group; and
5 R2 is NRN5RN6 where RN3 and RN6 together with the nitrogen to which they are bound
form an optionally substituted imidazolyl, morpholino, thiomorpholino, piperadinyl, homopiperadinyl, piperazinyl (preferably N-substituted), homopiperazinyl (preferably N-substituted) or pyrrolidinyl, wherein optional N-substituents on the piperazinyl and homopiperazinyl groups include Ci_7alkyl groups or esters, in particular, esters bearing
10 a Ci_7 alkyi group as an ester substituent, e.g. -C(-0)OCH3s -C(=0)OCH2CH3 and
-C(=0)OC(CH3)3) and optional C-substituents for the imidazolyl, morpholino, thiomorpholino, piperadinyl, homopiperadinyl, piperazinyl, homopiperazinyl or pyrrolidinyl groups include phenyl, ester, amide and CM alkyi, preferably methyl, aminomethyl, hydroxymethyl or hydroxyethyl.
15 In a further embodiment, there is provided a subset of compounds of formula (II),
(Ila) or (lib), and pharmaceutical^ acceptable salts thereof, in which: X5 and X6 are each CH; X8 is N; Z is H, For OR03
20 RN,° is selected from hydrogen, -C(0)CH3j -C(0)CH2OH, -CH3, -CH2CH3,
-CH2CH2OH, -CH(CH3)2, -CH2CH2OMe, -CH2C(CH3)2, -CH2CH2C(CH3)2; -CH(CH3)CH2C(CH3)2, -CH2CH2CH2N(CH3)2) cycloproyl, cyclopentyl, cyclohexyl, cycloheptyl, -CH2cyclopropyl, methylcyclohexyl, eyanocyclohexyl, pyrazolyl, hydroxypyrrolidinyl, -CH2imidazole;
25 RNl0a is hydrogen; or
RN1° and RNl0a together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 5 or 6 ring atoms, where the optional subtsituents are selected from halo, hydroxyl, Ci^alkyloxy; R03 is a methyl group; and
30 R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound
form an optionally substituted morpholino, thiomorpholino, piperidinyl, homopiperidinyl, piperazinyl (preferably N-substituted), homopiperazinyl (preferably

WO 2008/023161

PCT/GB2007/003179

N-substituted) or pyrrolidinyl group, wherein the optional substituents are selected
from cyano, halo, hydroxyl, and C1 .7saturated alkyl and C1.7 saturated alkoxy
(wherein the saturated alkyl and alkoxy groups may be optionally substituted by one
or more groups selected from halo, hydroxyl, C1.7 alkoxy, amino and C5-6 aryl).
5 In a further embodiment, there is provided.a subset of compounds of formula (II).
(Ila) or (lib), and pharmaceutically acceptable salts thereof, in which: X5 and X6 are each CH; X8isN; Z is H, F or OR03
10 RN,° is a RN1° is selected from hydrogen, -C(0)CH3, -C(0)CH2OH, -CH3, -CH2CH3,
-CH2CH2OH, -CH(CH3)2, -CH2CH2OMe, -CH2C(CH3)2! -CH2CH2C(CH3)2, -CH(CH3)CH2C(CH3)2, -CH2CH2CH2N(CH3)2, cycloproyl, cyclopentyl, cyclohexyl, cycloheptyl, -CH2cyclopropyl, methylcyclohexyl, cyanocyclohexyl, pyrazolyl, hydroxypyrrolidinyl, -CH2imidazole;
15 RN10a is hydrogen; or
Rmo and RN,0a together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 5 or 6 ring atoms, where the optional subtsituents are selected from halo, hydroxyl, Ct^alkyloxy; R03 is a methyl group; and
20 R2 is a group selected from


WO 2008/023161

PCT/GB2007/003179






10

In a further embodiment, there is provided a subset of compounds of formula (II), (Ila) or (lib), and pharmaceutical^ acceptable salts thereof, in which: X5 and X6 are each CH; X8isN;
Z is H, For OR03
RN1° is selected from hydrogen, -C(0)CH3j -C(0)CH2OH, -CH3, -CH2CH3, -CH2CH2OH, -CH(CH3)2, -CH2CH2OMe, -CH2C(CH3)23 -CH2CH2C(CH3)2, -CH(CH3)CH2C(CH3)2, -CH2CH2CH2N(CH3)2, cycloproyl, cyclopentyl, cyclohcxyl,

WO 2008/023161

PCT/GB2007/003179

cycloheptyl, -CH2cyclopropyl, methylcyclohexyl, cyanocyclohexyl, pyrazolyl,
hydroxypyrrolidinyl, -CHiimidazole;
RN10a is hydrogen; or
RN,° and RNIOa together with the nitrogen to which they are bound form an optionally
substituted heterocyclic ring containing between 5 or 6 ring atoms, where the optional
subtsituents are selected from halo, hydroxyl, Ci^alkyloxy;
R03 is a methyl group; and
R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound



10 In a further embodiment, there is provided a subset of compounds of formula (II),
(Ua) or (lib), and pharmaceutically acceptable salts thereof, in which:
X5 and X6 are each CH;
X8isN;
Z is H, F or OR03
15 RNI° is selected from hydrogen, -C(0)CH3, -C(0)CH2OH, -CH3, -CH2CH3,
-CH2CH2OH, -CH(CH3)2, -CH2CH2OMe, -CH3C(CH3)2, -CH2CH2C(CH3)2,
-CH(CH3)CH2C(CH3)2, -CH2CH2CH2N(CH3)2, cycloproyl, cyclopentyl, cyclohexyl,
cycloheptyl, -CH2cyclopropyl, methylcyclohexyl, cyanocyclohexyl, pyrazolyl,
hydroxypyrrolidinyl, -CH2imidazole;
20 RNIOa is hydrogen; or
RN1° and RN10a together with the nitrogen to which they are bound form an optionally
substituted heterocyclic ring containing between 5 or 6 ring atoms, where the optional
subtsituents are selected from halo, hydroxyl, Ci-7alkyloxy;
R03 is a methyl group; and
25 R2 is NRN5RN5 where RN5 and RN6 together with the nitrogen to which they are bound
; i r
N

*Q" or ^ -O' "" or \^/ or . .
form a u group.

WO 2008/023161

PCT/GB2007/003179

In a further embodiment, there is provided a subset of compounds of formula (II),
(Ila) or (lib), and pharmaceutically acceptable salts thereof, in which:
X5 and X6 are each CH;
X8isN;
5 Z is H, F or OR03
RN,° is selected from hydrogen, -C(0)CH3, -C(0)CH2OH, -CH3, -CH2CH3, -CH2CH2OH, -CH(CH3)2, -CH2CH2OMe, -CH2C(CH3)2, -CH2CH2C(CH3)2: -CH(CH3)CH2C(CH3)2, -CH2CH2CH2N(CH3)2, cycloproyl, cyclopentyl, cyclohexyl, cycloheptyl, -CH2cyclopropyl, methylcyclohexyl, cyanocyclohexyl, pyrazolyl,
10 hydroxypyrrolidinyl, -CH2imidazole;
RNIOa is hydrogen; or
RNi0 and RNIOa together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 5 or 6 ring atoms, where the optional subtsituents are selected from halo, hydroxyl, C1-7alkyloxy;
15 R°3 is a methyl group; and
R2 is NRN5RN6 where RN5 and RN6 together with the nitrogen to which they are bound

In another aspect of the invention, there is provided a compound, or a pharmaceutical salt thereof, selected from any one of the Examples.
20 In a further aspect of the invention, there is provided a compound, or a pharmaceutical
salt thereof, selected from Examples lbu, Ice, 12b, 18de, 18dg, 18j, lar, I9e, 19h, I9i, 191, 19m, 19n, 19o, 18n, 18o, 18z, 18aa, 18ag, 18ai, ISal, lv, 18az, lah, 7e, 7i, 7j, 5d, 5f, 4v, 4ab, 4aj, 5t, 5u, 5w, 5x, 5y, 5z, 3f, 3g, 18bp, 18bs318bv, 18by; 18cb, 18cv, law, 3u, lbf, ISct, 19q, 19s, 19u, 19v, 19w, lau, 5r, 4t, 18dj, lcl, 2d, 2e, les, 2h, 2j, lew, lbo, lbp, lj, lbx, Iby, lei
25 lei, lcj, 4an, 4ap, 4av, 12d, 18dh, 18di, 6a, In, lp, lq, 18e, 18h, 19b, 19c, 19f, 19k, 18p, lbd, lSw, ISab, 18af, ISaj, 18aq, 18as, 18av, ISay, 18bb; 18bc, 18bf, lSbl, lab, 4p, 9a, lav, 3a, 5b, 5c, 5e, 5g, 4aa, 4ad, 4ah, 5v, 3e, 18bq3 18bt, 18bz, 18ca, 18cd, IScg, 18ci, 18bx, 5n, lam, lao, 18cn, 18cx, lbk, 13b, 4g, 5s, 4q, 18dd, lep, lcq, 2f, 2g, 13g, lev, let, lb, la, 1c, Id, lbl, lbm, If, li, lg, lh, lbr, lbs, lbv, le, lbz, Ice, Ik, leg, II, 4al, 4am, 4ao, 4aq, 4as, 4at, 4au3
30 4aw, 4ax, 4ay, 4az, 4ba, 4bb, 4bc, 4bd, 4be, 4bf, 12c, 12a, 18a, las, Is, 18c, 18d, 18f, 18g,

WO 2008/023161

PCT/GB2007/003179

18L 18k, 19j, 18m, 18q, 18r, 18s, 18t, ISu,. 18v, 18x, 18y, 18ac, 18ad, 18ae, 18ah, 18ak, 18am, 18an, 18ap, ISar, 18au, 18aw, 18ax, 18ba, 18bd, 18be, 18bg, 18bi, 18bk, 18bh, lSbj, 18bm, lbg, 8b, 4h, lba, 8a, laa, lac, lae, laf, lag, 14b, lbc, 4i, 4j, 4k, 41, 4m, 4n, 4o, 18bn, 18bo, 4u, lbb, lat, 7b, 7c, 7d, 7f, 7g, 7k, 5a, 4w, 4x, 4y, 4z, 4ac, 4af, 4ai, 18br, 18bw, 18cc, 5 18cf, 18ch, 18cj, 18ck, 18cl, 4ak, 18cm, 4a, 3i, 3y, lak, lat, lap, lbe, 18co, 18cr, 18cs, 18db, 19p, 31, lu, 4b, 5q, 4c, 4e, 4f, 4d, laz, 4r, 4s, lcn, lco, 3ad, lcr, lew, ley, ldv, 15c, lcl, 1cm, lcn, lcq, lev, lex, Idi, ldj, leb, lcj, lck, let, leu, lcz, ldb, ldc, ldd, lde, ldg, ldh, ldk, ldl, 1dm, ldn, ldo, ldp, ldq, ldt, ldu, ldw, ldy, ldz, lea, lee, led, lee, 18dm, 18dnand 18do.
10 In a further aspect of the invention, there is provided a compound, or a pharmaceutical
salt thereof, selected from Examples'lbo, lbp, Ij, lbx, Iby, lef, lei, lcj, 4an, 4ap, 4av, 12d, 18dh, 18di, 6a, In, lp, lq, 18e, 18h, 19b, 19c, 19f, 19k, 18p, lbd, 18w, 18ab, 18af, 18aj, 18aq, 18as, 18av, ISay, 18bb, 18bc, 18bf, 18bl, lab, 4p, 9a, lav, 3a, 5b, 5c, 5e, 5g, 4aa, 4ad, 4ah, 5v, 3e, 18bq, 18bt, 18bz, ISca, 18cd, 18cg, 18ci, 18bx, 5n, lam, lao, IScn, 13cx, lbk,
15 13b, 4g, 5s, 4q, 18dd, lep, lcq, 2f, 2g, 13g, lev, let, lb, la, lc, id, lbl, lbm, If, li, lg, lh, Ibr, lbs, Ibv, le, lbz, Ice, Ik, leg, 11, 4al, 4am, 4ao, 4aq, 4as, 4at, 4au, 4aw, 4ax, 4ay, 4az, 4ba, 4bb, 4bc, 4bd, 4be, 4bf, 12c, 12a, 18a, las, Is, 18c, 18d, 18f, 18g, ISi, 18k, 19j, 18m, 18q, 18r, 18s, 18t, 18u, 18v, 18x, 18y, 18ac, 18ad, 18ae, 18ah, 18ak, 18am, 18an, 18ap, 18ar, 18au, 18aw, 18ax, 18ba, 18bd, 18be, 18bg, 18bi, 18bk, 18bh, 18bj, 18bm, lbg, 8b, 4b; lba,
20 8a, laa, lac, lae, laf, lag, 14b, lbc, 4i, 4j, 4k, 41, 4m, 4n, 4o, I8bn, 18bo, 4u, lbb, lat 7b, 7c, 7d, 7f, 7g, 7k, 5a, 4w, 4x, 4y, 4z, 4ac, 4af, 4ai, 18br, 18bw, 18cc, 18cf, 18ch, 18cj, 18ck, 18cl, 4ak, 18cm, 4a, 3i, 3y, lak, lal, lap, lbe, 18co, 18cr, 18cs, 18db, 19p, 31, lu, 4b, 5q, 4c, 4e, 4f, 4d, laz, 4r, 4s, lcn, lco, 3ad, lcl, 1cm, lcn, lcq, lev, lex, ldi, ldj, leb, lcj, lck, let leu, lcz, ldb, ldc, ldd, lde, ldg, ldh, ldk, ldl, 1dm, ldn, ldo, ldp, ldq, ldt, ldu, ldw, ldy, ldz,
25 lea, lee, led, lee, 18dm, 18dnand 18do.
In a further aspect of the invention, there is provided a compound, or a pharmaceutical salt thereof, selected from Examples lb, la, lc, Id, lbl, lbm; If, li, lg, lh, lbr, lbs, lbv, le, lbz, lec, Ik, leg, 11,4al, 4am, 4ao, 4aq, 4as, 4at, 4au, 4aw, 4ax, 4ay, 4az, 4ba, 4bb, 4bc, 4bd, 4be,4bf, 12c, 12a, 18a, las, Is, 18c, 18d, 18f, 18g, 18i, 18k, 19j, 18m, 18q, I8r, 18s, 18t, 18u,
30 18v, 18x, 18y, 18ac, ISad, 18ae, 18ah, 18ak, 18am, 18an, 18ap, 18ar, 18au: I8aw, ISax, 18ba, 18bd, 18be, 18bg, 18bi, 18bk, 18bh, 18bj, 18bm, lbg, 8b, 4h, lba, 8a, laa, lac, lae, laf, lag, 14b, lbc, 4i, 4j, 4k, 41, 4m, 4n, 4o, 18bn, 18bo, 4u, lbb, lat, 7b, 7c, 7d, 7f, 7g? 7k, 5a, 4w, 4x,

WO 2008/023161

PCT/GB2WV7/003179

4y, 4z, 4ac, 4af, 4ai, 18br, 18bw, 18cc, 18cf, 18ch, 18cj, I8ck, 18cl, 4ak, 18cm, 4a, 3i, 3y,
lak, lal, lap, lbe, 18co, 18cr, IScs, 18db, 19p, 31, lu, 4b, 5q, 4c, 4e, 4f, 4d, laz, 4r, 4s, lcn,
lco, 3ad, lcj, lck, let, leu, Icz, Idb, Idc, Idd, Ide, ldg, ldh, Idk, ldl, 1dm, ldn, Ido, ldp,
ldq, ldt, ldu, ldw, ldy, ldz, lea, lee, led, lee, 18dm, 18dnand 18do.
5 In a further aspect of the invention, there is provided a compound, or a pharmaceutical
salt thereof, selected from Examples la, lu, lal, lap, lat, laz, lco, Ide, ldg, ldh, ldk, ldl, ldp, ldq, ldr, Ids, ldt, ldu, ldy, lee, lee, 12d, 14b, 18dn and 18do. Includes Other Forms
Included in the above are the well known ionic, salt, solvate, and protected forms of
10 these substituents. For example, a reference to carboxylic acid (-COOH) also includes the anionic (carboxylate) form (-COO'), a salt or solvate thereof, as well as conventional protected forms. Similarly, a reference to an amino group includes the protonated form (-N+HR1R2), a salt or solvate of the amino group, for example, a hydrochloride salt, as well as conventional protected forms of an amino group. Similarly, a reference to a hydroxyl group
15 also includes the anionic form (-0"), a salt or solvate thereof, as well as conventional protected forms of a hydroxyl group. Isomers, Salts, Solvates, Protected Forms, and Prodrugs
Certain compounds may exist in one or more particular geometric, optical, enantiomeric, diasteriomeric, epimeric, stereoisomeric, tautomeric, conformational, or
20 anomeric forms, including but not limited to, cis- and trans-iotros\ E- and Z-forms; c-, t-, and r-forms; endo- and exo-forms; R-, S-, and mes-o-forms; D- and L-forms; d- and /-forms; (+) and (-) forms; keto-, enol-, and enolate-forms; syn- and anti-forms; synclinal- and anticlinal-forms; a- and p-forms; axial and equatorial forms; boat-, chair-, twist-, envelope-, and half chair-forms; and combinations thereof, hereinafter collectively referred to as "isomers"
25 (or "isomeric forms").
If the compound is in crystalline form, it may exist in a number of different polymorphic ' forms. For example, for Example la was isolated as Form A: 2-Theta° 6.9 (46%), 8.53 (100%), 10.1 (21%), 10.86 (24%), 11.65 (11%), 13.31 (14%), 13.75 (7%), 14.37 (54%), 15.21 (5%), 16.19 (13%), 16.81 (39%), 17.19 (40%), 17.97 (21%), 18.41 (65%), 18.78 (80%), 20.66
30 (8%), 21.07 (89%), 22.05 (19%), 22.36 (42%), 24 (7%), 24.36 (33%), 25.25 (31%), 25.54 (16%), 26.92 (18%), 27.26 (8%), 28.03 (8%), 28.39 (21%), 29 (8%), 29.91 (13%), 30.62 (23%), 31.48 (9%), 32.72 (5%), 33.27 (11%), 34.88 (4%), 35,48 (5%), 36.16 (4%), 36.88

WO 2008/023161

PCT/GB2007/003179

(4%), 37.37 (4%), 37.91 (6%), 38.65 (4%) and 39.83 (4%). A less stable form, 'Form B, has also been isolated from water / THF: 2-Theta° 3.67 (7%), 7.28 (7%), 8.52 (7%), 9.22 (30%), 11.42 (78%), 12.69 (24%), 13 (15%), 13.41 (44%), 13.6 (26%), 14.51 (19%), 15.56 (13%), 16.25 (9%), 17.11 (13%), 17.55 (18%), 18.24 (64%), 18.59 (56%), 19.51 (33%), 19.85 (26%), 5 20.32 (13%), 21.49 (17%), 21.79 (13%), 22.23 (18%), 22.84 (26%), 23.72 (23%), 25.46 (74%), 26.1 (100%), 26.72 (43%), 27.94 (16%), 28.35 (8%), 34.74 (10%), 35.34 (6%), 36.72 (9%) and 38.55 (4%).
Note that, except as discussed below for tautomeric forms, specifically excluded from the term "isomers", as used herein, are structural (or constitutional) isomers (i.e. isomers
10 which differ in the connections between atoms rather than merely by the position of atoms in space). For example, a reference to a methoxy group, -OCH3, is not to be construed as a reference to its structural isomer, a hydroxymethyl group, -CH2OH. Similarly, a reference to ortho-chlorophenyl is not to be construed as a reference to its structural isomer, meta-chlorophenyl. However, a reference to a class of structures may well include structurally
15 isomeric forms falling within that class (e.g., C1.7 alkyl includes «-propyl and wo-propyl; butyl includes n-, iso-, sec-, and tert-butyl; methoxyphenyl includes ortho-, meta-, and para-methoxyphenyl).
The above exclusion does not pertain to tautomeric forms, for example, keto-, cnol-, and enolate-forms, as in, for example, the following tautomeric pairs: keto/enol,
20 imine/enamine, amide/imino alcohol, amidine/amidine, nitroso/oxime, thioketone/enethiol, N-nitroso/hyroxyazo, and nitro/aci-nitro.
Note that specifically included in the term "isomer" are compounds with one or more isotopic substitutions. For example, H may be in any isotopic form, including 'H, 2H (D), and 3H (T); C may be in any isotopic form, including 12C, l3C, and l4C; O may be in any isotopic
25 form, including l60 and I80; and the like.
Unless otherwise specified, a reference to a particular compound includes all such isomeric forms, including (wholly or partially) racemic and other mixtures thereof. Methods for the preparation (e.g. asymmetric synthesis) and separation (e.g. fractional crystallisation and chromatographic means) of such isomeric forms are either known in the art or are readily
30 obtained by adapting the methods taught herein, or known methods, in a known manner.

WO 2008/023161

PCT/GB2007/003179

Unless otherwise specified, a reference to a particular compound also includes ionic, salt, solvate, and protected forms of thereof, for example, as discussed below, as well as its different polymorphic forms.
It may be convenient or desirable to prepare, purify, and/or handle a corresponding 5 salt of the active compound, for example, a pharmaceutically-acceptable salt. Examples of ■ pharmaceutical^ acceptable salts are discussed in ref. 25.
For example, if the compound is anionic, or has a functional group which may be anionic (e.g., -COOH may be -COO"), then a salt may be formed with a suitable cation. Examples of suitable inorganic cations include, but are not limited to, alkali metal ions such
10 as Na+ and K+, alkaline earth cations such as Ca2+ and Mg2+, and other cations such as Al3+. Examples of suitable organic cations include, but are not limited to, ammonium ion (i.e.., NH4+) and substituted ammonium ions (e.g., NH3R+, NH2R2+, NHR3+, NR44). Examples of some suitable substituted ammonium ions are those derived from: ethylamine, diethylamine, dicyclohexylamine, tri ethyl amine, butylamine, ethylenediamine, ethanolamine.
15 diethanolamine, piperazine, benzylamine, phenylbenzylamine, choline, meglumine, and tromethamine, as well as amino acids, such as lysine and arginine. An example of a common quaternary ammonium ion is N(CH3)4+.
If the compound is cationic, or has a functional group which may be cationic (e.g., -NH2 may be -NH3"*), then a salt may be formed with a suitable anion. Examples of suitable
20 inorganic anions include, but are not limited to, those derived from the following inorganic acids: hydrochloric, hydrobromic, hydroiodic, sulfuric, sulfurous, nitric, nitrous, phosphoric, and phosphorous. Examples of suitable organic anions include, but are not limited to, those derived from the following organic acids: acetic, propionic, succinic, gycolic, stearic, palmitic, lactic, malic, pamoic, tartaric, citric, gluconic, ascorbic, maleic, hydroxymaleic,
25 phenylacetic, glutamic, aspartic, benzoic, cinnamic, pyruvic, salicyclic, sulfanilic, 2-ace tyoxy benzoic, fiimaric, toluenesulfonic, methanesulfonic, ethane sulfonic, ethane disulfonic, oxalic, isethionic, valeric, and gluconic. Examples of suitable polymeric anions include, but are not limited to, those derived from the following polymeric acids: tannic acid, carboxymethyl cellulose.
30 It may be convenient or desirable to prepare, purify, and/or handle a corresponding,
solvate of the active compound. The term "solvate" is used herein in the conventional sense to refer to a complex of solute (e.g. active compound, salt of active compound) and solvent.

WO 2008/023161

PCT/GB2(in7/0«3179

If the solvent is water, the solvate may be conveniently referred to as a hydrate, for example, a mono-hydrate, a di-hydrate, a tri-hydrate, etc.
It may be convenient or desirable to prepare, purify, and/or handle the active compound in a chemically protected form. The term "chemically protected form," as used 5 herein, pertains to a compound in which one or more reactive functional groups are protected from undesirable chemical reactions, that is, are in the form of a protected or protecting group (also known as a masked or masking group or a blocked or blocking group). By protecting a reactive functional group, reactions involving other unprotected reactive functional groups can be performed, without affecting the protected group; the protecting group may be
10 removed, usually in a subsequent step, without substantially affecting the remainder of the molecule. See, for example, ref. 26.
For example, a hydroxy group may be protected as an ether (-OR) or an ester (-OC(=0)R), for example, as: a /-butyl ether; a benzyl, benzhydryl (diphenylmethyl), or trityl (triphenyhnethyl) ether; a trimethylsiiyl or /-butyldimethylsilyl ether; or an acetyl ester
15 (-OC(=0)CH3, -OAc).
For example, an aldehyde or ketone group may be protected as an acetal or ketal, respectively, in which the carbonyl group (>C=0) is converted to a diether (>C(OR)2), by reaction with, for example, a primary alcohol. The aldehyde or ketone group is readily regenerated by hydrolysis using a large excess of water in the presence of acid.
20 For example, an amine group may be protected, for example, as an amide or a
urethane, for example, as: a methyl amide (-NHCO-CH3); a benzyloxy amide (-NHCO-OCH2C6H5, -NH-Cbz); as a t-butoxy amide (-NHCO-OC(CH3)3, -NH-Boc); a 2-biphenyl-2-propoxy amide (-NHCO-OC(CH3)2CfiH4C6H5, -NH-Bpoc), as a 9-fluorenylmethoxy amide (-NH-Fmoc), as a 6-nitroveratryloxy amide (-NH-Nvoc), as a 2-trimethylsilyIethyloxy amide (-
25 NH-Teoc), as a 2,2,2-trichloroethyloxy amide (-NH-Troc), as an allyloxy amide (-NH-Alloc), as a 2(-phenylsuIphonyl)ethyloxy amide (-NH-Psec); or, in suitable cases, as an iV-oxide (>NO).
For example, a carboxylic acid group may be protected as an ester for example, as: an C1.7 alkyl ester (e.g. a methyl ester; a /-butyl ester); a C1-.7 haloalkyl ester (e.g. a C1-7
30 trihaloalkyl ester); a triCi-7 alkyIsilyl-Ci.7 alkyl ester; or a C5.20 aryl-C1-7 aikyl ester (e.g. a benzyl ester; a nitrobenzyl ester); or as an amide, for example, as a methyl amide.

WO 2008/023161

PCT/GB2007/003179

For example, a thiol group may be protected as a thioether (-SR), for example, as: a
benzyl thioether; an acetamidornethyl ether (-S-CH2NHC(=0)CH3).
It may be convenient or desirable to prepare, purify, and/or handle the active
compound in the form of a prodrug. The term "prodrug", as used herein, pertains to a 5 compound which, when metabolised (e.g. in vivo), yields the desired active compound.
Typically, the prodrug is inactive, or less active than the active compound, but may provide
advantageous handling, administration, or metabolic properties.
For example, some prodrugs are esters of the active compound (e.g. a physiologically
acceptable metabolically labile ester). During metabolism, the ester group (-C(=0)OR) is 10 cleaved to yield the active drug. Such esters may be formed by esterification, for example, of
any of the carboxylic acid groups (-C(=0)OH) in the parent compound, with, where
appropriate, prior protection of any other reactive groups present in the parent compound,
followed by deprotection if required. Examples of such metabolically labile esters include
those wherein R is C1-20 alkyl (e.g. -Me, -Et); C1-7 aminoalkyl (e.g. aminoethyl; 2-(N,N-15 diethylamino)ethyl; 2-(4-morpholino)ethyl); and acyloxy-Ci-7 alkyl (e.g. acyloxymethyl;
acyloxyethyl; e.g. pivaloyloxymethyl; acetoxymethyl; 1-acetoxyethyl; l-(l-methoxy-l-
methyl)ethyl-carbonxyloxyethyl; l-(benzoyloxy)ethyl; isopropoxy-carbonyloxymethyl;
1-isopropoxy-carbonyloxyethyl; cyclohexyl-carbonyloxymethyl; 1-cyclohexyl-
carbonyloxyethyl; cyclohexyloxy-carbonyloxymethyl; l-cyclohexyloxy-carbonyloxyethyl; (4-20 tetrahydropyranyloxy) carbonyloxymethyl; l-(4-terrahydropyranyloxy)carbonyIoxyethyl; (4-
tetrahydropyranyl)carbonyloxymethyl; and 1 -(4-tetrahydropyranyl)carbonyloxyethyl).
Further suitable prodrug forms include phosphonate and glycolate salts. In particular,
hydroxy groups (-Orl), can be made into phosphonate prodrugs by reaction with
chlorodibenzylphosphite, followed by hydrogenation, to form a phosphonate group -O-25 P(=0)(OH)2. Such a group can be cleared by phosphotase enzymes during metabolism to
yield the active drug with the hydroxy group.
Also, some prodrugs are activated enzymatically to yield the active compound, or a
compound which, upon further chemical reaction, yields the active compound. For example.
the prodrug may be a sugar derivative or other glycoside conjugate, or may be an amino acid 30 ester derivative.

WO2008/023T61

PCT/GB2007/003179

Acronyms
For convenience, many chemical moieties are represented using well known
abbreviations, including but not limited to, methyl (Me), ethyl (Et), ^-propyl (nPr), iso-propyl
(iPr), /7-butyl (nBu), tert-butyl (tBu), >?-hexyl (nHex), cyclohexyl (cHex), phenyl (Ph),
5 biphenyl (biPh), benzyl (Bn), naphthyl (naph), methoxy (MeO), ethoxy (EtO), benzoyl (Bz),
and acetyl (Ac).
For convenience, many chemical compounds are represented using well known abbreviations, including but not limited to, methanol (MeOH), ethanol (EtOH), iso-propanol (i-PrOH), methyl ethyl ketone (MEK), ether or diethyl ether (Et20), acetic acid (AcOH), 10 dichlorom ethane (methylene chloride, DCM), trifluoroacetic acid (TFA), dimethylformamide (DMF), tetrahydrofuran (THF), and dimethylsulfoxide (DMSO). General Synthesis
Compounds of formula I can be represented by Formula 1:

15 wherein R represents
Compounds of Formula 1 can be synthesised from compounds of Formula 2:
ii i.

When R7 is NRN,RN2, this is by reaction with R7H. When R7 is an amide, urea or sulfonamide group, this is by reaction with ammonia followed by reaction of the resulting primary amide
20 with the appropriate acid chloride, isocyanate or sulfonyl chloride. When R7 is OR01 or SRSI, this is by reaction with potassium carbonate in the appropriate alcohol or thiol solvent. When R7 is an optionally substituted C3.20 heterocyclyl group or C5.2o aryl group, this is by reaction with R7B(OAlk)2, where each Alk is independently C1.7 alkyl or together with the oxygen to which they are attached form a C5.7 heterocyclyl group.
25 Compounds of Formula 2 can be synthesised from compounds of Formula 3:

WO 2008/023161

PCT/GB2007/003179


by reaction with ,^v V~D. , *v„v.,v~ »j .~«~«v,.. u..
Compounds of Formula 3 can be synthesised from compounds of Formula 4:

5 by treatment with POClj and N,N-diisopropylamine, for example.
Compounds of Formula 4 can be synthesised from compounds of Formula 5:
O
,X5
x° ^r NH2
J\ fl-^L Formula 5
or x NH2
by treatment with oxalyl chloride, for example.
. Compounds of Formula 5 can be synthesised from compounds of Formula 6, for 10 example by reaction with liquid ammonia followed by reaction with thionyi chloride and ammonia gas:
O
x6'Y^(
OH
I Formula 6
cr ^x? cAlternatively compounds of Formula 1 can be synthesised from compounds of
Formula 7:
.-.4
XXA, Formula7
X6'
15 R" "X N CI
by reaction with HR2.

WO 2008/023161

PCT7GB2007/003179

Compounds of Formula 7 can be synthesised from compounds of Formula 8:

When R7 is NRN1RN2, this is by reaction with R7H. When R7 is an amide, urea or sulfonamide
group, this is by reaction with ammonia followed by reaction of the resulting primary amide
5 with the appropriate acid chloride, isocyanate or sulfonyl chloride. When R7 is OR01 or SRSI,
this is by reaction with potassium carbonate in the appropriate alcohol or thiol solvent. When
R7 is an optionally substituted C3.20 heterocyclyl group or C5-20 aryl group, this is by reaction
with R7B(OAlk)2, where each Alk is independently C1.7 alkyl or together with the oxygen to
which they are attached form a C5.7 heterocyclyl group.
10 Compounds of Formula 8 can be synthesised ftom compounds of Formula 3:
CI


WO 2008/023161

PCT/GB20fl7/003179






wherein Lv is a leaving group, such as a halogen, for example chlorine, or a OSO2 group, where R is allcyl or aryl, such as methyl, by reaction with RN10NH2. 5 Compounds of Formula la can be synthesised by reaction of a compound of Formula lb
R4

with an alkyl or aryl sulphonyl chloride in the presence of a base.
10 For Example:

WO 2008/023161

PCT7GB20tt7/«Mtt179



Compounds of Formula lb can be prepared by reaction with R7B(OAlk)23 where each Alk is independently Ci.7 alkyl or together with the oxygen to which they are attached form a C5_7 heterocyclyl group. 5 Use
The present invention provides active compounds, specifically, active in inhibiting the activity of mTOR.
The term "active" as used herein, pertains to compounds which are capable of inhibiting mTOR activity, and specifically includes both compounds with intrinsic activity 10 (drugs) as well as prodrugs of such compounds, which prodrugs may themselves exhibit little or no intrinsic activity.
One assay which may. conveniently be used in order to assess the mTOR inhibition offered by a particular compound is described in the examples below.
The present invention further provides a method of inhibiting the activity of mTOR in 15 a cell, comprising contacting said cell with an effective amount of an active compound, preferably in the form of a pharmaceutically acceptable composition. Such a method may be practised in vitro or in vivo.
For example, a sample of cells may be grown in vitro and an active compound brought
into contact with said cells, and the effect of the compound on those cells observed. As
20 examples of "effect", the inhibition of cellular growth in a certain time or the accumulation of
cells in the Gl phase of the cell cycle over a certain time may be determined. Where the
active compound is found to exert an influence on the cells, this may be used as a prognostic

WO 2008/023161

PCT7GB2007/003179

or diagnostic marker of the efficacy of the compound in methods of treating a patient carrying cells of the same cellular type.
The term "treatment", as used herein in the context of treating a condition, pertains generally to treatment and therapy, whether of a human or an animal (e.g. in veterinary 5 applications), in which some desired therapeutic effect is achieved, for example, the inhibition of the progress of the condition, and includes a reduction in the rate of progress, a halt in the rate of progress, amelioration of the condition, and cure of the condition. Treatment as a prophylactic measure (i.e. prophylaxis) is also included.
The term "adjunct" as used herein relates to the use of active compounds in
10 conjunction with known therapeutic means. Such means include cytotoxic regimes of drugs and/or ionising radiation as used in the treatment of different cancer types. Examples of adjunct anti-cancer agents that could be combined with compounds from the invention include, but are not limited to, the following: alkylating agents: nitrogen mustards, mechlorethamine, cyclophosphamide, ifosfamide, melphalan, chlorambucil: "Nitrosoureas:
15 carmustine (BCNXJ), lomustine (CCNU), semustine (methyI-CCNU), ethylenimine/methylmelamine, thriethylenemelamine (TEM), triethylene thiophosphoramide (thiotepa), hexamethylmelamine (HMM, altretamine): Alkyl sufonates; busulfan; Triazines, dacarbazine (DTIC): Antimetabolites; folic acid analogs, methotrexate, trimctrexate, pyrimidine analogs, 5-fluorouracil, fluorodeoxyuridine, gemcitabine, cytosine arabinoside
20 (AraC, cytarabine). 5-azacytidine, 2,2'-di£luorodeoxycytidine: Purine analogs; 6-mercaptopurine, 6-thioguanine, azathioprine, 2!-deoxycoformycin (pentostatin, erythrohydroxynonyladenine (EHNA), fludarabine phosphate, 2-Chlorodeoxyadenosine (cladribine, 2-CdA): Topoisomerase I inhibitors; camptothecin, topotecan, irinotecan, rubitecan: Natural products; antimitotic drugs, paclitaxel, vinca alkaloids, vinblastine (VLB),
25 vincristine, vinorelbine, Taxotere™ (docetaxel), estramustine, estramustine phosphate; epipodophylotoxins, etoposide, teniposide: Antibiotics; actimomycin D, daunomycin (rubidomycin), doxorubicin (adriamycin), mitoxantrone, idarubicin, bleomycins, plicamycin (mithi-amycin), mitomycin C, dactinomycin: Enzymes; L-asparaginase, RNAse A: Biological response modifiers; interferon-alpha, IL-2, G-CSF, GM-CSF: Differentiation Agents; retinoic
30 acid derivatives: Radiosensitizers;, metronidazole, misonidazole, desmethylmisonidazole, pimonidazole, etanidazole, nimorazole, RSU 1069, E09, RB 6145, SR4233, nicotinamide, 5-bromodeozyuridine, 5-iododeoxy uridine, bromodeoxycytidine: Platinium coordination

WO 2008/023161

PCT/GB2007/003179

complexes; cisplatin, carboplatin: Anthracenedione; mitoxantrone, AQ4N Substituted urea,
hydroxyurea; Methylhydrazine derivatives, N-methylhydrazine (MIH), procarbazine;
Adrenocortical suppressant, mitotane (o.p'-DDX)), aminoglutethimide: Cytokines; interferon
(a, P, y), interleukin; Hormones and antagonists; adrenocorricosteroids/antagonists, 5 prednisone and equivalents, dexamethasone, aminoglutethimide; Progestins,
hydroxyprogesterone caproate, medroxyprogesterone acetate, megestrol acetate; Estrogens,
diethyl stilbestrol, ethynyl estradiol/equivalents; Antiestrogen, tamoxifen; Androgens,
testosterone propionate, fluoxymesterone/equivalents; Anti androgens, flutamide,
gonadotropin-releasing hormone analogs, leuprolide; Nonsteroidal anti androgens, flutamide; 10 EGFR inhibitors, VEGF inhibitors; Proteasome inhibitors.
Active compounds may also be used as cell culture additives to inhibit mTOR, for
example, in order to sensitize cells to known chemotherapeutic agents or ionising radiation
treatments in vitro.
Active compounds may also be used as part of an in vitro assay, for example, in order 15 to determine whether a candidate host is likely to benefit from treatment with the compound
in question.
Cancer
The present invention provides active compounds which are anticancer agents or
adjuncts for treating cancer. One of ordinary skill in the art is readily able to determine 20 whether or not a candidate compound treats a cancerous condition for any pai'ticular cell type,
either alone or in combination.
Examples of cancers include, but are not limited to, lung cancer, small cell lung
cancer, gastrointestinal cancer, bowel cancer, colon cancer, breast. carinoma, ovarian
carcinoma, prostate cancer, testicular cancer, liver cancer, kidney cancer, bladder cancer, 25 pancreas cancer, brain cancer, sarcoma, osteosarcoma, Kaposi's sarcoma, melanoma and
leukemias.
Any type of cell may be treated, including but not limited to, lung, gastrointestinal
(including, e.g., bowel, colon), breast (mammary), ovarian, prostate, liver (hepatic), kidney
(renal), bladder, pancreas, brain, and skin.
30 The anti cancer treatment defined hereinbefore may be applied as a sole therapy or
may involve, in addition to the compound of the invention, conventional surgery or

WO 2008/023161

PCT/GB2007/G0M79

radiotherapy or chemotherapy. Such chemotherapy may include one or more of the following categories of anti-tumour agents:-
(i) other antiproliferative/antineoplastic drugs and combinations thereof, as used in medical oncology, such as alkylating agents (for example cis platin, oxaliplatin, 5 carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan, temozoiamide and nitrosoureas); antimetabolites (for example gemcitabine and antifolates such as fluoropyrimidines like 5 fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, and hydroxyurea); antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C,
10 dactinomycin 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, topotecan and camptothecin);
(ii) cytostatic agents such as antioestrogens (for example tamoxifen, fulvestrant,
15 toremifene, raloxifene, droloxifene and iodoxyfene), antiandrogens (for example bicalutamide, flutamide, nilutamide and cyproterone acetate). LHRH antagonists or LHRH agonists (for example goserelin, leuprorelin and buserelin), progestogens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrozole, vorazole and exemestane) and inhibitors of 5*-reductase such as finasteride;
20 (iii) anti-invasion agents (for example c-Src kinase family inhibitors like 4-(6-
chloro-2,3-memylenedioxyanilino)-7-[2~(4-memylpiperazin-l-yl)ethoxy]-5-tetrahydropyran-4-yloxyquinazoline (AZD0530; International Patent Application WO 01/94341) and N-(2-chloro-6-memylphenyl)-2-{6-[4-(2-hydroxyethyl)piperazin-l-yl]-2-methyIpyrimidin-4-ylamino}thiazole-5-carboxamide (dasatinib, BMS-354825; J. Med. Chem., 2004, 47, 6658-
25 6661), and metalloproteinase inhibitors like marimastat, inhibitors of urokinase plasminogen activator receptor function or antibodies to Heparanase);
(iv) inhibitors of growth factor function: for example such inhibitors include growth factor antibodies and growth factor receptor antibodies (for example the anti erbB2 antibody trastuzumab [Herceptin™], the anti-EGFR. antibody panitumumab, the anti erbBl
30 antibody cetuximab [Erbitux, C225] and any growth factor or growth factor receptor antibodies disclosed by Stern et al. Critical reviews in oncology/haematology, 2005, Vol. 54, ppl 1-29); such inhibitors also include tyrosine kinase inhibitors, for example inhibitors of the

WO 2008/023161

PCT/GB2007/00317«J

epidermal growth factor family (for example EGFR family tyrosine kinase inhibitors such as
N"(3-chloro-4-fluorophenyl)-7-methoxy-6-(3-morpholinopropoxy)quinazolin-4-amine
(gefitinib, ZD1839), N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)quinazolin-4-amine
(erlotinib, OSI 774) and 6-acrylamido-Nw(3-chloro-4-fluorophenyl)-7-(3-
5 morpholinopropoxy)-quinazolin-4-amine (CI 1033), erbB2 tyrosine kinase inhibitors such as lapatinib, inhibitors of the hepatocyte growth factor family, inhibitors of the platelet-derived growth factor family such as imatinib, inhibitors of serine/threonine kinases (for example Ras/Raf signalling inhibitors such as farnesyl transferase inhibitors, for example sorafenib (BAY 43-9006)), inhibitors of cell signalling through MEK and/or AKT kinases, inhibitors of
10 the hepatocyte growth factor family, c-kit inhibitors, abl kinase inhibitors, IGF receptor (insulin-like growth factor) kinase inhibitors; aurora kinase inhibitors (for example AZD1152, PH739358, VX-680, MLN8054, R763, MP235, MP529, VX-528 AND AX39459) and cyclin dependent kinase inhibitors such as CDK2 and/or CDK4 inhibitors;
(v) antiangiogenic agents such as those which inhibit the effects of vascular
15 endothelial growth factor, [for example the anti vascular endothelial cell growth factor antibody bevacizumab (Avastin™) and VEGF receptor tyrosine kinase inhibitors such as 4-(4-bromo-2-fiuoroanilino)-6-methpxy-7-(l-methylpiperidin-4-ylmethoxy)quinazoline (ZD6474; Example 2 within WO 01/32651), 4-(4-fluoro-2-methylindol-5-yloxy)-6-methoxy-7-(3-pyrrolidin-l-ylpropoxy)quinazoline (AZD2171; Example 240 within WO 00/47212),
20 vatalanib (PTK787; WO 98/35985) and SUl 1248 (sunitinib; WO 01/60814), 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 avb3 function and angiostatin)];
(vi) vascular damaging agents such as Combtetastatin A4 and compounds
25 disclosed in International Patent Applications WO 99/02166, WO 00/40529, WO 00/41669, WO 01/92224, WO 02/04434 and WO 02/08213;
(vii) antisense therapies, for example those which are directed to the targets listed above, such as ISTS 2503, an anti-ras antisense;
(viii) gene therapy approaches, including for example approaches to replace aberrant
30 genes such as aberrant p53 or aberrant BRCAl or BRCA2, GDEPT (gene directed enzyme pro drug therapy) approaches such as those using cytosine deaminase, thymidine kinase or a

WO 20(18/023161

PCT/GB2007/003179

bacterial nitroreductase enzyme and approaches to increase patient tolerance to chemotherapy or radiotherapy such as multi drug resistance gene therapy; and
(ix) immunotherapy approaches, including for example ex vivo and in vivo approaches to increase the immunogenicity of patient tumour cells, such as transfection with 5 cytokines such as interleukin 2, interleukin 4 or granulocyte macrophage colony stimulating factor, approaches to decrease T cell anergy, approaches using transfected immune cells such as cytokine transfected dendritic cells, approaches using cytokine transfected tumour cell lines and approaches using anti idiotypic antibodies. Administration
10 The active compound or pharmaceutical composition comprising the active compound
may be administered to a subject by any convenient route of administration, whether systemically/ peripherally or at the site of desired action, including but not limited to, oral (e.g. by ingestion); topical (including e.g. transdermal, intranasal, ocular, buccal, and sublingual); pulmonary (e.g. by inhalation or insufflation therapy using, e.g. an aerosol, e.g.
15 through mouth or nose); rectal; vaginal; parenteral, for example, by injection, including subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, and intrasternal; by implant of a depot, for example, subcutaneously or intramuscularly.
20 The subject may be a eukaryote, an animal, a vertebrate animal, a mammal, a rodent
(e.g. a guinea pig, a hamster, a rat, a mouse), murine (e.g. a mouse), canine (e.g. a dog), feline (e.g. a cat), equine (e.g. a horse), a primate, simian (e.g. a monkey or ape), a monkey (e.g. marmoset, baboon), an ape (e.g. gorilla, chimpanzee, orangutang, gibbon), or a human. Formulations
25 While it is possible for the active compound to be administered alone, it is preferable
to present it as a pharmaceutical composition (e.g., formulation) comprising at least one active compound, as defined above, together with one or more pharmaceutically acceptable carriers, adjuvants, excipients, diluents, fillers, buffers, stabilisers, preservatives, lubricants, or other materials well known to those skilled in the art and optionally other therapeutic or
30 prophylactic agents.
Thus, the present invention further provides pharmaceutical compositions, as defined above, and methods of making a pharmaceutical composition comprising admixing at least

WO 2008/023161

PCT/GB2007/003179

one active compound, as defined above, together with one or more pharmaceutically acceptable carriers, excipients, buffers, adjuvants, stabilisers, or other materials, as described
herein.
A pharmaceutical composition of the present invention comprising a compound of formula (I) such as herein described, and a pharmaceutically acceptable carrier or diluent showed unexpected and surprising results. Therefore, the said composition is synergistic.
The term "pharmaceutically acceptable" as used herein pertains to compounds,
5 materials, compositions, and/or dosage forms which are, within the scope of sound medical
judgement, suitable for use in contact with the tissues of a subject (e.g. human) without
excessive toxicity, irritation, allergic response, or other problem or complication,
commensurate with a reasonable benefit/risk ratio. Each carrier, excipient, etc. must also be
"acceptable" in the sense of being compatible with the other ingredients of the formulation.
10 Suitable carriers, diluents, excipients, etc. can be found in standard pharmaceutical
texts. See, for example, refs. 27 to 29.
The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. Such methods include the step of bringing into association the active compound with the carrier which constitutes one or 15 more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active compound with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product.
Formulations may be in the form of liquids, solutions, suspensions, emulsions, elixirs, syrups, tablets, lozenges, granules, powders, capsules, cachets, pills, ampoules, suppositories, 20 pessaries, ointments, gels,. pastes, creams, sprays, mists, foams, lotions, oils, boluses, electuaries, or aerosols.
Formulations suitable for oral administration (e.g., by ingestion) may be presented as
discrete units such as capsules, cachets or tablets, each containing a predetermined amount of
the active compound; as a powder or granules; as a solution or suspension in an aqueous or
25 non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion; as
a bolus; as an electuary; or as a paste.
A tablet may be made by conventional means, e.g. compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active compound in a free-flowing form such as a powder or 30 granules, optionally mixed with one or more binders (e.g. povidone, gelatin, acacia, sorbitol, tragacanth, hydroxypropylmethyl cellulose); fillers or diluents (e.g. lactose, microcrystalline cellulose, calcium hydrogen phosphate); lubricants (e.g. magnesium stearate, talc, silica);

WO 2008/023161

PCT/GB2007/003179

disintegrants (e.g. sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose); surface-active or dispersing or wetting agents (e.g., sodium lauryl sulfate); and preservatives (e.g., methyl p-hydroxybenzoate, propyl p-hydroxybenzoate, sorbic acid). Molded tablets may be made by molding in a suitable machine a mixture of the 5 powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active compound therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile. Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.
10 Formulations suitable for topical administration (e.g. transdermal, intranasal, ocular,
buccal, and sublingual) may be formulated as an ointment, cream, suspension, lotion, powder, solution, past, gel, spray, aerosol, or oil. Alternatively, a formulation may comprise a patch or a dressing such as a bandage or adhesive plaster impregnated with active compounds and optionally one or more excipients or diluents.
15 Formulations suitable for topical administration in the mouth include losenges
comprising the active compound in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active compound in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active compound in a suitable liquid carrier.
20 Formulations suitable for topical administration to the eye also include eye drops
wherein the active compound is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active compound.
Formulations suitable foT nasal administration, wherein the carrier is a solid, include a coarse powder having a particle size, for example, in the range of about 20 to about 500
25 microns which is administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close up to the nose. Suitable formulations wherein the carrier is a liquid for administration as, for example, nasal spray, nasal drops, or by aerosol administration by nebuliser, include aqueous or oily solutions of the active compound.
30 Formulations suitable for administration by inhalation include those presented as an
aerosol spray from a pressurised pack, with the use of a suitable propellant, such as

WO 2008/023161

PCT/GB2007/003179

dichlorodifluoromethane, trichloroiluoromethane, dichoro-tetrafluoroethane, carbon dioxide, or other suitable gases.
Formulations suitable for topical administration via the skin include ointments, creams, and emulsions. When formulated in an ointment, the active compound may 5 optionally be employed with either a parafflnic or a water-miscible ointment base. Alternatively, the active compounds may be formulated in a cream with an oil-in-water cream base. If desired, the aqueous phase of the cream base may include, for example, at least about 30% w/w of a polyhydric alcohol, i.e., an alcohol having two or more hydroxyl groups such as propylene glycol, butane-1,3-diol, mannitol, sorbitol, glycerol and polyethylene glycol and
10 mixtures thereof. The topical formulations may desirably include a compound which enhances absorption or penetration of the active compound through the skin or other affected areas. Examples of such dermal penetration enhancers include dimethylsulfoxide and related analogues.
When formulated as a topical emulsion, the oily phase may optionally comprise
15 merely an emulsifier (otherwise known as an emulgent), or it may comprises a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil. Preferably, a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabiliser. It is also preferred to include both an oil and a fat. Together, the emulsifier(s) with or without stabiliser(s) make up the so-called emulsifying wax, and the wax together with the oil and/or
20 fat make up the so-called emulsifying ointment base which forms the oily dispersed phase of the cream formulations.
Suitable emulgents and emulsion stabilisers include Tween 60, Span 80, cetostearyl alcohol, myristyl alcohol, glyceryl monostearate and sodium lauryl sulphate. The choice of suitable oils or fats for the formulation is based on achieving the desired cosmetic properties,
25 since the solubility of the active compound in most oils likely to be used in pharmaceutical emulsion formulations may be very low. Thus the cream should preferably be a non-greasy, non-staining and washable product with suitable consistency to avoid leakage from tubes or other containers. Straight or branched chain, mono- or dibasic alkyl esters such as di-isoadipate, isocetyl stearate, propylene glycol diester of coconut fatty acids, isopropyl
30 myristate, decyl oleate, isopropyl palmitate, butyl stearate, 2-ethylhexyl palmitate or a blend of branched chain esters known as Crodamol CAP may be used, the last three being preferred esters. These may be used alone or in combination depending on the properties required.

WO 2008/023161

PCTYGB2
Alternatively, high melting point lipids such as white soft paraffin and/or liquid paraffin or other mineral oils can be used.
Formulations suitable for rectal administration may be presented as a suppository with
a suitable base comprising, for example, cocoa butter or a salicylate.
5 Formulations suitable for vaginal administration may be presented as pessaries,
tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active compound, such carriers as are known in the art to be appropriate.
Formulations suitable for parenteral administration (e.g., by injection, including cutaneous, subcutaneous, intramuscular, intravenous and intradermal), include aqueous and
10 non-aqueous isotonic, pyrogen-free, sterile injection solutions which may contain anti¬oxidants, buffers, preservatives, stabilisers, bacteriostats, and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents, and liposomes or other microparticulate systems which are designed to target the compound to
15 blood components or one or more organs. Examples of suitable isotonic vehicles for use in such formulations include Sodium Chloride Injection, Ringer's Solution, or Lactated Ringer's Injection. Typically, the concentration of the active compound in the solution is from about 1 ng/ml to about 10 fig/ml, for example from about 10 ng/ml to about 1 (ig/ml. The formulations may be presented in unit-dose or multi-dose sealed containers, for example,
20 ampoules and vials, and may be stored in a freeze-dried (iyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules, and tablets. Formulations may be in the form of liposomes or other microparticulate systems which are designed to target the active compound to blood
25 components or one or more organs. Dosage
It will be appreciated that appropriate dosages of the active compounds, and compositions comprising the active compounds, can vary from patient to patient. Determining the optimal dosage will generally involve the balancing of the level of
30 tlierapeutic benefit against any risk or deleterious side effects of the treatments of the present invention. The selected dosage level will depend on a variety of factors including, but not limited to, the activity of the particular compound, the route of administration, the time of

WO 2008/023161

PCT/GB2007/003179

administration, the rate of excretion of the compound, the duration of the treatment, other drugs, compounds, and/or materials used in combination, and the age, sex, weight, condition, general health, and prior medical history of the patient The amount of compound and route of administration will ultimately be at the discretion of the physician, although generally the 5 dosage will be to achieve local concentrations at the site of action which achieve the desired effect without causing substantial harmful or deleterious side-effects.
Administration in vivo can be effected in one dose, continuously or intermittently (e.g., in divided doses at appropriate intervals) throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those 10 of skill in the art and will vary with the formulation used for therapy, the purpose of the therapy, the target cell being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician.
In general, a suitable dose of the active compound is in the range of about 100 {ig to 15 about 250 mg per kilogram body weight of the subject per day. Where the active compound is a salt, an ester, prodrug, or the like, the amount administered is calculated on the basis of the parent compound and so the actual weight to be used is increased proportionately.
In addition to their use in therapeutic medicine, the compounds of formula (I) and their pharmaceutical^ acceptable salts are also useful as pharmacological tools in the 20 development and standardisation of in vitro and in vivo test systems for the evaluation of the effects of inhibitors of mTor in laboratory animals such as cats, dogs, rabbits, monkeys, rats and mice, as part of the search for new therapeutic agents.
In the above other pharmaceutical composition, process, method, use and medicament manufacture features, the alternative and preferred embodiments of the compounds of the 25 invention described herein also apply.

WO 2008/023161

PCT/GB2007/003179

Examples
General Experimental Methods
Thin Layer chromatography was carried out using Merck Kieselgel 60 F254 glass backed plates. The plates were visualized by the use of a UV lamp (254 nm). Silica gel 60 5 (particle sizes 40-63 Jim) supplied by E.M.Merck was employed for flash chromatography. 'H NMR spectra were recorded at 300 MHz on a Bruker DPX-300 instrument. Chemical shifts were referenced relative to tetramethylsilane. Purification of samples
The samples were purified on Gilson LC units. Mobile phase A - 0.1 % aqueous
10 TFA, mobile phase B - Acetonitrile; flow rate 6 ml/min; Gradient - typically starting at 90 %
A/10 % B for 1 minute, rising to 97 % after 15 minutes, holding for 2 minutes, then back to
the starting conditions. Column: Jones Chromatography Genesis 4jim, CI8 column, 10 mm
X 250 mm. Peak acquisition based on UV detection at 254 nm.
Identification of samples
15 QC Method QC2-AQ
Mass spectra were recorded on a Waters ZQ instrument in Electrospray ionisation mode. Mobile phase A - 0.1 % aqueous formic acid. Mobile phase B - 0.1 % Formic acid in acetonitrile; Flowrate 2 ml/min; Gradient - starting at 100 % A/0 % B for 1 minute, rising to 95 % B after 7 minutes and holding for 2 minutes before returning to the starting conditions. 20 Column: Varies, currently Genesis AQ 120A 4u 50mm X 4.6mm, Hiclirom Ltd. PDA detection Waters 996, scan range 210-400 nm.
QC Method QC2-Long
Mass spectra were recorded on a Waters ZQ instrument in Electrospray ionisation
25 mode. Mobile phase A - 0.1 % aqueous formic acid. Mobile phase B - 0.1 % Formic acid in
acetonitrile; Flowrate 2 ml/min; Gradient - starting at 95 % A/5 % B, rising to 95 % B after
20 minutes and holding for 3 minutes before returning to the starting conditions. Column:
Varies, but always C18 50 mm X 4.6 mm (currently Genesis C18 4u 50mm X 4.6mm,
Hichrom Ltd). PDA detection Waters 996, scan range 210-400 nm.
30
QC Method QC2-QC
Mass spectra were recorded on a Waters ZQ instrument in Electrospray ionisation
mode. Mobile phase A — 0.1 % aqueous formic acid. Mobile phase B - 0.1 % Formic acid in

WO 2008/023161

PCT/GB2007/003179

acetonitrile; Flowrate 2 ml/min; Gradient - starting at 95 % A/5 % B, rising to 95 % B after 5 minutes and holding for 5 minutes before returning to the starting conditions. Column: Varies, but always CI8 50 mm X 4.6 mm (currently Genesis C18 4urn 50 X 4.6 mm, Hichrom Ltd). PDA detection Waters 996, scan range 210-400 nm.
5
QC Method QC3-AQ-Long
Mass spectra were recorded on a Waters ZQ instrument in Electrospray ionisation
mode. Mobile phase A - 0.1 % aqueous formic acid. Mobile phase B - 0.1 % Formic acid in
acetonitrile; Flowrate 2 ml/min; Gradient - starting at 100 % A/0 % B for 1 minute, rising to
10 95 % B after 20 minutes and holding for 5 minutes before returning to the starting conditions.
Column: Varies, currently Genesis AQ 4pm 50mm X 4.6mm, Hichrom Ltd. PDA detection
Waters 996, scan range 210-400 nm.
Examples lu, 9a, 18bs, 18bv, 18bw, 18bx, 18by, 18bz, 18ca, 18cb, 18cc, 18cd, 18ce,
18cf, 18cg, 18ch, 18ci, 18cj, 18ck, 18cl, 18cm, I8dk, 18dl and 18dm were analysed using the
15 QC Method QC2-AQ.
Examples 12c, 12d, 13c, 13e, 13g, 14b, 15b, I8aa, 18ab, 18ac, ISad, 18ae, 18af, 18ag, 18ah, 18ai, 18aj, 18ak, 18al, 18am, 18an, 18ao, 18ap, 18aq, 18ar, 18as, 18at, 18au, 18az, 18bc, 18bl, 18bm, 18bt, 18bu, 18cn, 18co, 18cp, 18cq, 18cr, 18cs, I Set, 18cu, 18cv, 18cw,
20 18cx, 18cy, 18cz, 18da, 18db, 18dc, 18df, 18dj, 181, 18o, 18q, 18rs 18s, 18t, 18u, 18v, 18w, 18x, 18y, 18z, 19a, 19b, 19c, 19d, 19e, 19f, 19g, 19h, 19i, 19j, 19k, 191, 19m, 19n, 19o, la, laa, lab, lac, lad, lae, laf, lag, lah, lai, lak, las, lau, laz, Ibb, lcq, let Idg, lee, lg, li, lm, lw, lx, ly, Iz, 21a, 3a, 3ac, 3b, 3c, 3d, 3e, 3f, 3g, 3h, 3i, 3j, 3v, 3w, 3x, 3y, 3z, 4j, 4k, 41, 4m, 4n, 4o, 4p, 6a, comparative of Example lc, comparative of Example lj and comparative
25 of Example Ik were analysed using the QC Method QC2-Long,
Examples 10a, 11a, 12a, 12b, 12e, 13a, 13b, 13d, 13f, 14a, 15a, 15c, 16a, 17a, 18a, 18av, 18aw, 18ax, 18ay, 18b, 18ba, 18bb, 18bd, lSbe, 18bf, 18bg, lSbh, 18bi, 18bj, lSbk, 18bn, l8bo, 18bp, 18bq, 18br, 18c, 18d, 18dd, I8de, 18dg, 18dh, 18di, 18dn, 18do, 18e, 18f, 30 18g, 18h, 18i, 18j, 18k, 18m, 18n, 19p, 19q, 19r, 19s, 19t, 19u, 19v, 19w, 19x, laj, lal, lam, Ian, lao, lap, laq, lar, lat, lav, law, lax, lay, lb, Iba, lbc, lbe, lbf, lbg, lbh, Ibi, lbj, lbk, lbl, lbm, lbn, Ibo, Ibp, lbq, lbr, lbs, lbt, lbu, lbv, lbw, lbx, lby, Ibz, lc, lea, leb, lec, led, Ice, lef, leg, Ich, lei, lcj, Ick, lei, 1cm, len, lco, lep, lcr, les, leu, lev, lew, lex,

WO 2008/023161

PCT7GB2007/003179

ley, lcz, Id, Ida, Idb, Idc, ldd, lde, ldf, ldh, Idi, ldj, ldk, ldl, 1dm, ldn, ldo, ldp, ldq, ldr, Ids, ldt, ldu, ldv, ldw, ldx, ldy, ldz, le, lea, leb, led, lee, If, lh, lj, Ik, 11, In, lo, lp, Iq, lr, Is, It, lv, 20a, 20b, 20c, 2a, 2b, 2c, 2d, 2e, 2f, 2g, 2h, 2i, 2j, 3aa, 3ab, 3ad, 3k, 31, 3m, 3n, 3o, 3p, 3q, 3r, 3s, 3t, 3u, 4a, 4aa, 4ab, 4ac, 4ad, 4ae, 4af, 4ag, 4ah, 4ai, 4aj, 4ak, 4al, 5 4am, 4an, 4ao, 4ap, 4aq, 4ar, 4as, 4at, 4au, 4av, 4aw, 4ax, 4ay, 4az, 4b, 4ba, 4bb, 4bc, 4bd, 4be, 4bf, 4c, 4d, 4e, 4f, 4g, 4h, 4i, 4q, 4r, 4s, 4t, 4u, 4v, 4w, 4x, 4y, 4z, 5a, 5b, 5c, 5d, 5c, 5f, 5g, 5h, 5i, 5j, 5k, 51, 5m, 5n, 5o, 5p, 5q, 5r, 5s, 5t, 5u, 5v, 5w, 5x, 5y, 5z, 7a, 7b, 7c, 7d, 7e, 7f, 7g, 7h, 7i, 7j, 7k, 8a, 8b, 8c, 8d, comparative of Example la and comparative of Example lb, were analysed using the QC Method QC2-QC. 10
Examples 18p and lbd were analysed using the QC Method QC3-AQ-Long.
Microwave synthesis
Reactions were carried out using a Personal Chemistry™ Emrys Optimiser microwave 15 synthesis unit with robotic arm Power range between. 0-300 W at 2.45 GHz. Pressure range between 0-20 bar; temperature increase between 2-5°C/sec; temp range 60-250°C.
General procedure for the synthesis of 2,4,7-substituted pyridopyrimidine derivatives:


WO 2008/023161

PCT/GB2007/003179

*2-amino-6- chloronicotinic acid - X=N, Y=C, Z=C *3-amino-chloroisonicotinic acid - X=C, Y=N, Z=C *3-Amino-chloropyridine-2-carboxylic acid-X=C, Y=C, Z=N
5 a) NH3,14 bar; b) (i) SOCl2, THF, r.t, (ii) NH3 c) Oxalyl chloride, Toluene, A; d) DIPEA, POC^, Toluene or Anisole, A;e) Appropriate amine, diisopropylethylamine, CH2CI2 or Anisole; f) Appropriate amine, diiosopropylethyl amine, DMA, 70 °C;

To the appropriate amino acid (1 equiv) was added liquid ammonia (sufficient to make a 0.6M solution of substrate in ammonia). The suspension was sealed in a pressure vessel which was then heated slowly to 130 °C. It was noted that at this temperature a pressure of 18
15 bar was observed. This.temperature and pressure was maintained for a further 16 hours whereupon the mixture was cooled to room temperature. The pressure vessel was opened and the reaction poured into ice cold water (1 reaction volume). The resulting solution was acidified to pH 1-2 using concentrated HC1 which caused a precipitate to form. The acidic mixture was allowed to warm to room temperature and was stirred like this for a further 30
20 min The suspension was then extracted with diethyl ether (3 X 400 ml). The combined organic extracts were then filtered and the filtrate concentrated in vacuo to give a white solid which was dried further over P2O5 to give the title compound (typically 80-90 % yield and 90 %+ pure) in suitably pure form to be used without any further purification.
25 2-amino-6-chloronicotinic acid - X=N, Y=C, Z=C: (90 % yield, 96 % purity) m/z (LC-MS, ESP): 173 [M+H]+R/T = 3.63 min

WO 2008/023161

PCT/GB2007/003179


To a 0.3 M solution of amino acid (1 equiv) in anhydrous THF, under an inert atmosphere, was added thionyl chloride (3.3 equiv) in a dropwise fashion. The reaction mixture was 5 stirred at room temperature for 2 hours. After this time the reaction was concentrated in vacuo to give a crude yellow solid residue. The crude solid was dissolved in THF (equal to initial reaction volume) and concentrated in vacuo again to give a yellow solid residue. The residue was dissolved once more in THF and concentrated as before to give a solid residue which was then dissolved in THF (to give a solution of 0.3M) and ammonia gas bubbled through the 10 solution for 1 hour. The resultant precipitate was removed by filtration and the filtrate concentrated in vacuo to give a yellow precipitate which was triturated with water at 50 °C then dried to give the title compound (typically 90-95 %) yield and suitably clean enough to be used without any further purification.
15 2-Amino-6-chloronicotinamide - X=N, Y=C, Z=C: (92 % yield, 93 % purity) m/z (LC-MS, ESP): 172 [M+Hf R/T = 3.19min

20 To a stirred solution (0.06 M) of substrate (1 equiv) in anhydrous toluene under an inert atmosphere was added oxalyl chloride (1.2 equiv) in a dropwise manner. The resulting mixture was then heated to reflux (115 °C) for 4 hours whereupon it was cooled and stirred for a further 16 hours. The crude reaction mixture was then concentrated to half its volume in vacuo and filtered to give the desired product in suitably pure form to be used without any
25 further purification.

WO 2008/023161

PCT/GB2007/003179

7-Chloro-lH-pyrido[2,3-d]pyrimidine-2,4-dione- X=N, Y>C, Z=C: (95 % yield, 96 % purity) m/z (LC-MS, ESP): 196 [M-H]" R/T = 3.22 min

5
To a stirred 0.5 M suspension of the appropriate dione (1 equiv) in anhydrous toluene under an inert atmosphere was slowly added diisopropylethylamine (3 equiv). The reaction mixture was then heated to 70 °C for 30 minutes and then cooled to room temperature prior to the addition of POCI3 (3 equiv). The reaction was then heated to 100 °C for 2.5 hours before
10 being cooled and concentrated in vacuo to give a crude slurry which was then suspended in EtOAc and filtered through a thin pad of Celite™. The filtrate was concentrated in vacuo to give a brown, oil which was dissolved in CH2CI2 and stirred over silica gel for 30 minutes. After this time the silica was removed by filtration, the filtrate concentrated and the crude residue purified by flash chromatography (SiOz) to give the title compound in analytically
15 pure form.
2,4,7-Trichloro-pyrido[2s3-d]pyrimidine- X=N, Y=C, Z=C: (48 % yield, 96 % purity) m/z (LC-MS, ESP): 234 [M+H]+ R/T = 4.21 min

To a cooled (0-5 °C) stirred solution (0.1 M) of the appropriate trichloro-substrate (1 equiv) in CH2CI2 was added diisopropylethylamine (1 equiv) in a dropwise fashion. The appropriate amine (1 equiv) was then added to the reaction mixture portionwise over the period of 1 hour. 25 The solution was maintained at room temperature with stirring for a further 1 hour before the mixture was washed with water (2x1 reaction volume). The aqueous extracts were combined

WO 2008/023161

PCT/GB2007/003179

and extracted with CH2CI2 (2x1 reaction volume). The organic extracts were then combined, dried (sodium sulphate), filtered and concentrated in vacuo to give an oily residue which solidified upon prolonged drying. The solid was triturated with diethylether and then filtered and the cake washed with cold diethyl ether to leave the title compound in suitable clean form 5 to be used without any further purification.
2,7-Dichloro-4-morpholin-4-yl-pyrido[2,3-d]pyrimidine - Rl- morpholine, X=N, Y=C, Z=C: (92 % yield, 90 % purity) m/z (LC-MS, ESP): 285 [M+H]+ R/T = 3.90 min
10 2J-Dichloro-4-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidine - Rl= (S)-3-Methyl-morpholine, X=N, Y=C, Z=C: (87 % yield, 92 % purity) m/z (LC-MS, ESP): 301 [M*H]+ R/T = 4.13 min
2,7-Dichloro-4-((R)-3-methyl-morpholin-4-yl)-pyrido[233-d]pyrimidine - Rl= (R)-3-Metbyl-15 morpholine: (99 % yield, 94 % purity) m/z (LC-MS, ESP): 301 [M+H]+ R/T = 3.49 min
Alternatively, to a stirred 0.47 M suspension of the appropriate dione (1 equiv) in anhydrous anisole under an inert atmosphere was added POCI3 (2.6 equiv). The mixture was heated to 55 °C and then diisopropylethylamine (2.6 equiv) was slowly added. The reaction mixture
20 was then heated to 85-90 °C for 30 minutes. Water was added in portions (0.15 equiv), and the reaction mixture was held at 85-90 °C for a further 30 minutes. The reaction was cooled to 50 °C, and then 15% of the anisole solvent was removed by vacuum distillation. The mixtutre was then cooled to -5 °C and diisopropylethylamine (1.1 equiv) was added. A 4.9M solution of the appropriate amine (1.05 equiv) in anisole was then added to the reaction
25 mixture continuously over a period of 1 hour. The solution was then warmed to 30 °C and the reaction monitored by HPLC until reation completion.
One third of the resulting mixture from the above reaction was then added over lOmin to a stirred mixture of 1.95M aqueous potassium hydroxide (3.9 equiv) and i-butanol (6.9 equiv) at 60 °C. The stirring was stoped, the phases were allowed to separate, and the
30 aqueous phase was removed. Stirring was resumed, and 1.95M aqueous potassium hydroxide (3.9 equiv) was added to the retained organic phase. The second third of the resulting reaction mixture from the reaction above was then added over lOmin at 60 °C. Again, stirring

WO 2008/023161

PCT/GB2007/003179

was stoped, the phases were allowed to separate, and the aqueous phase was removed. Stirring was resumed, and 1.95M aqueous potassium hydroxide (3.9 equiv) was added to the retained organic phase. The remaining third of the resulting reaction mixture from the reaction above was then added over lOmin at 60 °C. Again, stirring was stoped, the phases 5 were allowed to separate, and the aqueous phase was removed. Water was then added to the organic phase with stirring, and the stirred mixture heated to 75 °C. Stirring was stoped, the phases were allowed to separate, and the aqueous phase was removed. The resulting organic phase was stirred and allowed to cool to 30 °C, and then as the mixture was heated to 60 °C heptane (11.5 equiv) was added over 20min when the mixture was around 40 °C. After being 10 heated to 60 °C, the mixture was cooled over 2.5h to 10 °C. After 30min, the resulting slurry was filtered off, washed with a 10:1 heptane:anisole mixture (2 x l,4equiv) and then washed with heptane (2 x 1.4equiv). The solid was then dried in a vacuum oven at 50 °C to leave the title compound in suitable clean form to be used without any further purification.

To a solution (0.2 M) of the appropriate dichloro-substrate (1 equiv) in anhydrous dimethyl acetamide under an inert atmosphere was added diisopropylethylamine (1 equiv) followed by the appropriate amine (1 equiv). The resulting mixture was heated for 48 hours at 70 °C
20 before being cooled to ambient temperature. The reaction was diluted with CH2CI2 (1 reaction volume) and then washed with water (3x1 reaction volumes). The organic extract was concentrated in vacuo to give a syrup which was dissolved in EtOAc (1 reaction volume) and washed with saturated brine solution before being dried (sodium sulphate) and concentrated in vacuo to give an oil. The crude residue was purified by flash chromatography
25 (Si02j eluted with EtOAcHex (7:3) going to (1:1)) to give the title compound as a yellow solid that was suitably clean to be used without any further purification.

WO 2008/023161

PCT/CB2007/003179

7-CMoro-2-((2S56R)-2J6-dimethyl-morpholin-4-yl)-4-morpholin-4-yl"pyrido[253~ d]pyrirnidine - Rl= morpholine, R2=cis-dimethylmorpholine, X=N, Y=C, Z=C: (45 % yield, 85 % purity) m/z (LC-MS, ESP): 348 [M+H]+ R/T = 4.16 min
5 7-Chloro-4-((S)-3-methyl-morpholin-4-yl)-2-((S)-3-me%l-moi^holiii-4-yl)-pyrido[2,3-djpyrimidine - Rl= (S)-3-Methyl-morpholine, R2=(S)-3-Methyl-morphoIine, X=N, Y=C, Z-C: (71 % yield, 90 % purity) m/z (LC-MS, ESP): 364 [M+H]+ R/T = 3.52 min
7-Chloro-2-(2-e1iiyl-piperidin-l-yl)-4-((S)-3-methyl-morpholm-4-yl)-pyrido[2)3"d]pyrimidiTie 10 - Rl = (S)-3-MethyI-morpholine, R2 = 2-Ethyl-piperidine, X=N, Y=C, Z=C: (51 % yield, 98 % purity) m/z (LC-MS, ESP): 376 [M+H]+ R/T = 3.88 min
7-Chloro-4-((S)-3-methyl-morpholin-4-yl)-2-morpholin-4-yl-pyrido[2,3-d]pyrimidine, - Rl = (S)-3-Methyl-morphoIine, R2 = morpholine, X=N, Y=C, Z=C: (72 % yield, 96 % purity) m/z 15 (LC-MS, ESP): 350 [M+H]+ R/T = 3.45 min
7-Chloro-2-((2S,6R)-2,6-diniethyl-morpholin-4-yl)-4-((S)-3-methyl-morpholin-4"yl" pyrido[2,3-d]pyrimidine - Rl= (S)-3-Methyl-morpholine, R2=cis-dimethylmorpholine: (33 % yield) m/z (LC-MS, ESP): 378 [M+H]+ R/T = 3.68 min 20
7-CWoro-4-((R)-3-methyl-moi^)holin-4-yl)-2-((R)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidine, - Rl = R2 = (R)-3-Methyl-morpholine: (48 % yield, 100 % purity) m/z (LC-MS, ESP): 364 [M+H]+ R/T = 2.80 min
25 To a 0.33 M solution of 2,7-dichldro-4-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-
djpyrimidine (1 equiv) in NjN-dimethylacetamide was added Hunig's base (1 equiv) followed by the appropriate amine (1.1 equiv). The reaction mixture was heated 40 °C for 1 hour. After this time the reaction was allowed to cool, diluted with EtOAc (1 reaction volume) and then washed with water (1 reaction volume). The aqueous fraction was removed and extracted
30 further with EtOAc (2X1 reaction volume). The combined organic extracts were dried (MgSCH), filtered and concentrated in vacuo to give a crude oily residue which was purified

WO 2008/023161

PCT/GB2007/003179

by flash chromatography (S1O2) using EtOAc/Hexanes as eluent which furnished the desired products in a suitably clean form.
7-Chloro-4-((S)-3-methyl-morpholin-4-yl)-2-thiomoipholin-4-yl-pyrido[233-d]pyrimidine: 5 (30 % yield, 100 % purity) m/z (LC-MS, ESP): 366.4[M+H]+ R/T = 3.00 min
7-Chloro-4-((S)-3-metliyl-morpholin-4-yl)-2-(4-methyl-piperazin-l-yl)-pyrido[2,3-d]pyrimidine:(32 % yield, 95 % purity) m/z (LC-MS, ESP): 363.4[M+H]+ R/T - 2.37 min

The appropriate chloro-substrate (1 equiv) was dissolved in a toluene/ethanol (1:1) solution (0.02 M). Sodium carbonate (2 equiv) and the appropriate pinacolate boron ester or boronic acid (1 equiv) were then added followed by tetrakis(triphenylphosphine) palladium0 (0.1 15 equiv). The reaction vessel was sealed and the mixture exposed to microwave radiation (140 °C, medium absorption setting) for 30 minutes.. Upon completion the samples were filtered through a silica cartridge, washed with EtOAc and then concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired products.
20 Example 1:
Preparation of 2,4,7-substituted pyridopyrimidine Intermediates:
Procedures for the synthesis of 2-Ch!oro-4-((S)-3-m ethyl-mo rpholin-4-yI)-7-ary 11-
pyrido[2,3-d]pyrimidine derivatives


WO 2008/023161

PCT/GB2007/003179

To a (0.1 M) solution of 257-dichloro-4-((S)-3.methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidine (1 equiv) in MeCN/HaO (1:1 mixture) was added the appropriate pinacolate boron ester or boronic acid (1.1 equiv) and potassium carbonate (3 equiv). The mixture was
5 degassed with nitrogen for 20 minutes before the addition of
tetrakis(triphenylphosphine)palladium° (0.05 equiv). The reaction was degassed for a further 5 minutes before being heated to reflux under an inert atmosphere for 3 hours. Whereupon, it was concentrated in vacuo and the crude residue partitioned between CH2CI2/H2O. The organic fraction was dried (MgS04), filtered and concentrated in vacuo to give an oil which
10 was further purified by flash chromatography (Si02) using 5 % MeOH in CH2CI2 as eluent.

3-[2-Chloro-4
5-[2-Chloro-4-((S)-3-methyl-morpholin4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-pyridin-2-20 ylamine: (93 % yield, 89 % purity) m/z (LC-MS,ESP):357 [M+H]+, R/T = 2.53 min)

WO 2008/023161

PCT/GB2007/003179


2-Chloro-7-(4-chloro-phenyI)-4-((S)-3-methyl-morpholin-4-yl)-pyrido[23-d]pyrimidine: (80 % yield, 85 % purity) m/z (LC-MS,ESP):357.5 [M+H]+; R/T * 4.26 min)
.O.

5 {5-[2-Chloro-4-((R)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidin-7-ylj-2-methoxy-phenyl}-methanol: (97 % yield, 93 % purity) m/z (LC-MS,ESP):401 [M+H]+, R/T = 3.42 min)
Procedures for the synthesis of boronic ester:

10
5-bromo-2-methoxybenzoic acid methyl ester (1 equiv) was dissolved in dioxane (0.1 M). Bis(pinacolato)diboron (1.1 equiv), potassium acetate (3.5 equiv) and dppf (0.05 equiv) were added and the mixture was degassed with nitrogen for 20 minutes. (1,1'-Bis(diphenylphosphino)ferrocene-dichloropalladium (0.05 equiv) was added and the mixture 15 was degassed for a further 5 minutes. The reaction mixture was heated to 120 °C for 2 hours under nitrogen. After cooling to room temperature, the reaction mixture was diluted with CH2CI2 and filtered through Celite™. The filtrate was concentrated in vacuo to give a dark oil. The residue was partitioned between EtOAc and saturated aqueous sodium bicarbonate

WO 2008/023161

PO7GB2007/003179

and the aqueous layer further extracted with EtOAc. The combined organic phases were dried (MgSCU), filtered and the filtrate was concentrated in vacuo to give a dark residue which was purified by flash column chromatography onto silica gel eluting with 0 to 30 % ethyl acetate in hexane.

2-Metlioxy~5-(4,4,5,5-tetramethyl-[l,332]dioxaboroIan-2-yI)-benzoic acid methyl ester: (77 % yield, 100 % purity) m/z (LC-MS, ESP): 293.5 [M+H]+ R/T = 4.24 min
Procedures for the synthesis of tetrazolyl boron ie acids:
10

The appropriate cyanophenylpinacolate boron ester or boronic acid (1 equiv) was dissolved in DMP (0.67 M). Sodium azide (6 equiv) and ammonium chloride (6 equiv) were added. The
15 reaction mixture was heated to 120°C for 2.5 hours. After cooling down, the reaction mixture was poured into a mixture of ice water and EtOAc. Sodium nitrite was added and the aqueous phase was acidified by 6N HC1 until pH 2. The mixture was allowed to stir at room temperature for 30 min and then was extracted with EtOAc and n-butanol. Organic fractions were collected, dried over sodium sulphate, filtered off and concentrated in vacuo, to yield a
20 crude residue which was further purified accordingly:

The crude residue was recrystallized from CH2CI2 / hexane, obtaining the desired product as a white solid.

WO 2008/023161

PCT/GB2O07/003379

[3-(lH-tetrazol-5-yl)phenyl]boronic acid: (15 % yield, 100 % purity) m/z (LC-MS, ESP): 191 [M+H]+R/T = 2.49min



5 The crude residue was recrystallized from CH2CI2 / hexane, to give the desired product as a white solid.
[4-(lH-tetrazol-5-yl)phenyl]boronic acid: (64 % yield, 100 % purity) m/z (LC-MS, ESP): 191 [M+H]+R/T = 2.49min


15

The residue was purified by reverse phase column using a gradient from 5 % to 20 % acetonitrile in 0.1 % formic acid/water solution, yielding the desired product. [4-fluoro-3-(lH-tetrazol-5-yl)phenyl]boronic acid: (18 % yield, 100 % purity) m/z (LC-MS, ESP): 207 [M-H]~ R/T = 2.51 min
Procedure for the synthesis of methanesulfonylamido boronic acid:


3-Amino-4-fluorophenylboronic acid (1 equiv) was dissolved in THF (0.1 M). Methane 20 sulphonyl chloride (10 equiv) and pyridine (1 equiv) were added. The reaction mixture was heated to 70°C for 30 minutes. After cooling down, the reaction mixture was concentrated in vacuo, to yield a crude residue which was used without further purification.

WO 2008/023161

PCT/GB20D7/003179


3~(Methanesulfony]ammo)-4-fluoro-phenylboronic acid: (51 % yield, 90 % purity) m/z (LC-MS, ESP): 232 [M-H]" R/T = 2.50 min
5 Procedure for the synthesis of 3-hydroxymethyl-5-(4,4,5,5-tetramethyl-[l,3,2]dioxaboroIan-2-yl)-pyridin-2-ol

10 To a 0.18 M solution of 5-bromo-2-hydroxybenzyl alcohol (1 equiv) in dioxane was added bis(pinacolato)diboron (1.2 equiv) and potassium acetate (3.5 equiv) followed by 1,1'-bis(diphenylphosphino)ferrocene (0.05 equiv). The mixture was degassed with nitrogen for 20 minutes. PdCl2(dppf) (0.05 equiv) was added and the misxture degassed for a further 5 minutes. The reaction was then heated to reflux under an inert atmosphere for 2 hours. Upon
15 completion, the reaction was cooled, filtered and concentrated in vacuo to give a crude residue which was purified by flash chromatography (Si02) using EtOAc/Hexanes - 1:1 as eluent to give the desired product.
3-Hydroxymethyl-5-(4,4,5,5-tetramethyl-[l)3)2]dioxaborolan-2-yl)-pyridin-2-ol6-Bromo-3H-20 pyrido[2,3-d]pyrimidin-4-one: (67 % yield, 94 % purity) m/z (LC-MS,ESP):251 [M-H]" R/T — 3.32 minutes)

WO 2008/023161

PCT/GB2007/003179

Procedure for the synthesis of 5-(4,4,5,5-Tetramethyl-[l,3,2]dioxaborolan-2-yl)-l,3-dihydro-pyrrolo[2,3-b]pyridin-2-one

H H
5
To a 0.05 M solution of 5-bromo-l,3-dihydro-pyrrolo[2,3-b]pyridin-2-one (1 equiv) in
dioxane was added bis(pinacolato)diboron (1.2 eqiv) and potassium acetate (1.5 equiv)
followed by 1 J'-bis(diphenylphospruno)ferrocene (0.05 equiv). The mixture was degassed
with nitrogen for 20 minutes. PdCl2(dppf) (0.05 equiv) was added and the mixture degassed
10 for a further 5 minutes. The reaction was then heated to 120 °C under an inert atmosphere for
8 hours. Upon completion, the reaction was cooled, filtered and concentrated in vacuo to give
a crude residue which was purified by flash chromatography (SiC^) using EtOAc/Hexanes -
4:1 as eluent to give the desired product.
15 5-(4,4,5)5-Tetramethyl-[l,3)2]dioxaborolan-2-yl)-l,3-dihydro-pyrrolo[2,3-b]pyridin-2-one: (6S % yield, 92 % purity) rn/z (LC-MS,ESP):260 [M-H]' R/T = 3.52 minutes)
Procedure for the synthesis of 6-(4,4,5,5-Tetramethyl-[l,3)2]dioxaborolan-2~yl)-3H-pyrido[2,3-dJpvrimidin-4-one boronic ester
20

To a 1.2 M solution of 5-bromoanthranilic acid (1 equiv) in N,N-dimethylformamide was 25 added formamidine acetate (1 equiv). The mixture was heated to reflux and stirred at this temperature for 16 hours. After this time, the reaction was cooled and NaHC03 solution (5 % in H2O) (3 volumes) were carefully added and the mixture stirred vigorously. The resulting

WO 2008/023161

PCT/GB2007/003179

precipitate was collected by filtration and then washed with water (2X1 volume) and then t-butyl methylether (2X1 volume) before being dried in a vacuum oven to give the desired product which required no further purification.
5 6-Bromo-3H-pyrido[2,3-d]pyrimidin-4-one: (91 % yield, insert) m/z (LC-MS,ESP):225 [M-H]-R/T = 2.31 minutes)
To a (0.35M) solution of 6-bromo-3H-pyrido[2,3-d]pyrimidin-4-one (1 equiv) in dioxane was added bis(pinacolato)diboron (1.2 eqiv) and potassium acetate (1.5 equiv) followed by 1,T-
10 bis(diphenylphosphino)ferrocene (0.05 equiv). The mixture was degassed with tutrogen for 20 minutes. PdCl2(dppf) (0.05 equiv) was added and the misxture degassed for a further 5 minutes. The reaction was then heated to reflux under an inert atmosphere for 16 hours. After this time, the mixture was cooled, filtered through Celite ™ and then partitioned between CH2Cl2/NaHC03(aq). The organic traction was removed, dried (MgS04), filtered and
15 concentrated in vacuo. The crude residue was purified by flash chromatography (S1O2) 1:1-Hexanes:EtOAc going to neat EtOAc. The purified material was then dissolved in the minimum volume of CH2CI2 and hexane added in order to precipitate the desired product as a whiter crystraliine solid
20 6-(4J4,5,5-Tetramethyl-[l,3,2]dioxaborolan-2-yl)-3H-pyrido[2,3--d]pyrimidin-4-one (15 % yield, 96 % purity) m/z (LC-MS,ESP):Mass ion not observable, R/T = 3.30 min)
Procedure for the synthesis of 7-(4,4,5,5-Tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-pyrido[2,3-b] [l,4]oxazin-2-one

To a 0.3 M solution of 7-bromo-lH-pyrido[2,3-b][l,43oxazin-2-one (1 equiv) were added bis(pinacolato)diboron(1.10 equiv), potassium acetate (3.5 equiv) and 1,1'-bis(diphenylphosphino)ferrocene (0.05 equiv). The mixture was degassed with nitrogen for 20 30 minutes before the addition of PdC^dppf) (0.05 equiv) and degassing for a rurthere 5

WO 2008/023161

PCT/GB2007/003179

minutes. A condenser was attached to the reaction vessel and the mixture heated to reflux under an inert atmosphere for 16 hours. After this time, the reaction was cooled, filtered through Celite ™. The cake was washed CH2CI2 and the filtrate concentrated in vacuo before being re-dissolved in EtOAc and washed with H20 and then saturated brine. The organic 5 fraction was separated, dried (MgSO,}) and concentrated in vacuo to give a crude residue which was further purified by flash chromatography (SiOi) using 1:1- EtOAc:Hexanes going to neat EtOAc as eluent to give the desired product.
7-(4J4>5,5-Tetramethyl41s3,2]dioxaborolan-2-yl)4H-pyrido[2J3-b][l,4]oxazin-2-one:(97% 10 yield, 90%purity)m/z(LC-MS,ESP):317 [M+H+MeCN]+,R/T = 3.72min)
Procedure for the synthesis of 2-Methoxynicotinonitrile-5-boronic acid




15
To a cooled (-78 °C ) solution (0.25 M) of 5-bromo-2-methoxybenzonitrile in THF was added
n-BuLi (1.10 equiv of a 2.5 M solution in hexanes) dropwise. The mixture was maintained at
this temperature with stirring for 45 minutes before the addition of triisopropylborate (1.25
equiv). The reaction was then warmed to -20 °C before the addition of IN HC1 (0.5 reaction
20 volumes). The mixture was allowed to warm to room temperature and stirred like this for a further 20 minutes. After this time the mixture was diluted with H2O and then extracted with Et20 (3X4 reaction volumes). The combined organic fractions were then dried (MgSCU), filtered and concentrated in vacuo to give an off white solid which corresponded to the title compound
25
2-Methoxynicotinonitrile-5-boronic acid: (44 % yield, 90 % purity) m/z (LC-MS.ESP): 177.0 [M+H]+)R/T = 2.87min)

WO 2008/023161 PCT/GB2007/003179
Procedure for the synthesis of 2-Ethoxynieotinonitrile-5-boi*onic acid




5 To a cooled (-78 °C ) solution (0.25 M) of 5-bromo-2-ethoxybenzonitrile in THF was added n-BuLi (1.10 equiv of a 2.5 M solution in hexanes) dropwise. The mixture was maintained at this temperature with stirring for 45 minutes before the addition of triisopropylborate (1.25 equiv). The reaction was then warmed to -20 °C before the addition of IN HC1 (0.5 reaction volumes). The mixture was allowed to warm to room temperature and stirred like this for a 10 further 20 minutes. After this time the mixture was diluted witfr H20 and then extracted with EtnO (3x4 reaction volumes). The combined organic fractions were then dried (MgSO-O, filtered and concentrated in vacuo to give an off white solid which corresponded to the title compound.
15 2-EthoxynicotinonitrJle-5-boronic acid: (23 % yield, 97 % purity) m/z (LC-MS,ESP):191.0 |M+H]+,R/T = 3.09min)
Procedure for the synthesis of 2-IsopropoxynicotinonitriIe-5-boronic acid


20
To a cooled (-78 °C) solution (0.25 M) of 5-bromo-2-isopropo#y-nicotinonitrile in THF was added n-BuLi (1.10 equiv of a 2.5 M solution in hexanes) dropwise. The mixture was maintained at this temperature with stirring for 45 minutes before the addition of
25 triisopropylborate (1.25 equiv). The reaction was then warmed to -20 °C before the addition of IN HC1 (0.5 reaction volumes). The mixture was allowed to warm to room temperature and stirred like this for a further 20 minutes. After this time the mixture was diluted with H20 and then extracted with Et20 (3x4 reaction volumes). The combined organic fractions were then

WO 2008/023161

PCT/GB20lt7/00317y



5

dried (MgS04), filtered and concentrated in vacuo to give an off white solid which which was triturated with CH2C12 to give the desired compound.
2-Isopropoxy-nicotinonitrile-5-boronic acid: (100 % yield, 97 % purity) m/z (LC-MS,ESP):204.2 [M+H]\ R/T = 3.25 min)
Procedure for the synthesis of 7-(4,4,5,5-Tetram ethyl-[1,3,2] dioxaborolan-2-yl)-2H-phthalazin-1-one






10 To a 3M solution of 5-bromo-2-formyl benzoic acid (1 equiv) in water was added hydrazine hydrate (5 equivs). The reaction was heated to 95 °C for 4 hours whereupon a white precipitate had formed in the mixture. The reaction was cooled, and filtered. The white solid material was washed with cold methanol and dried to give the desired product.
15 7-Bromo-2H-phthalazin-l-one: (73 % yield, 95 % purity) m/z (LC-MS,ESP):225.2 [M+H]+, R/T = 2.99 min)
Bis(pinacolato)diboron (1.1 equiv), potassium acetate (3.5 equiv) and 1,1'-bis(diphenylphosphino)ferrocene (0.05 equiv) were dissolved in dioxane. The mixture was
20 degassed with nitrogen for 20 minutes before the addition of PdCl2(dppf) (0.05 equiv). The mixture was degassed for a further 5 minutes. The mixture was heated to reflux for 16 hours and then allowed to cool to room temperature. Water was added to the mixture before it was extracted with EtOAc (2X2 reaction volumes). The combined organic fractions were dried (MgSO^), filtered aned concentrated in vacuo before being purified by flash chromatography
25 (S1O2) neat hexanes going to 1:1- Hexanes:EtOAc then neat EtOAc to give the desire product as a white crystalline solid.
7-(4,4J5,5-Tetramethyl-[l,3,23dioxaborolan-2-yl)-2H-phthalazin-l-one: (86 % yield, 92 % purity) m/z (LC-MS,ESP):191.3 [M+H]+, R/T = 2.29 min)

WO 2008/023161

PCT/GB2007/D03179

Procedure for the synthesis of 6-(4,4,5,5-Tetramethyl-[l,3»2]dioxaborolan-2-yl)-2,3-
dihydro-isoindol-l-one:

0 O
•Br \ J^ ^^ ^Br
5-Bromo-2-methylbenzoic acid (1 equiv) was dissolved in a 1:9 MeOH/toluene mixture (0.1 M). The reaction mixture was cooled to 0°C and a trimethylsilyldiazomethane (1.05 equiv) solution in diethylether (2M) was added slowly until a persistent yellow tinge was observed.
10 The reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was concentrated in vacuo. The resulting residue was sonicated in hexane, collected by vacuum filtration over a sintered funnel, dried and used without further purification. 5-Bromo-2-methyl-benzoic acid methyl ester: (99 % yield, 100 % purity) m/z (LC-MS, ESP): no ionisation R/T = 4.43 min
15

To a solution of 5-Bromo-2-methyl-benzoic acid methyl ester (1 equiv) in chloroform (0.1 M) were added N-bromosuccinimide (1.2 equiv) and benzoyl peroxide (0.05 equiv). The reaction mixture was stirred at reflux for 16 hours. It was then diluted with chloroform and a 20 precipitate was collected by vacuum filtration on a sintered funnel. The filtrate was concentrated in vacuo. The subsequent residue was purified by flash column chromatography

WO 2008/023161

PCT/GB2007/003179

onto silica gel eluting with DCM in hexane (0 to 20 %) to yield the desired product as a clear colourless oil.
5-Bromo-2-bromomethyl-benzoic acid methyl ester: 80 % yield, 100 % purity) m/z (LC-MS, ESP): no ionisation R/T = 4.40 min
5

A solution of 5-bromo-2-bromomethyl-benzoic acid methyl ester (1 equiv) in a 1:1 THF/MeOH mixture was treated by gentle bubbling of ammonia gas for 40 minutes at room temperature. The reaction mixture was concentrated in vacuo. The residue was sonicated in 10 CH2CI2 for 15 minutes then filtered to give the desired product as a white solid.
6-Bromo-2,3-dihydro-isoindol-l-one: (98 % yield, 90 % purity) m/z (LC-MS, ESP): 212.3/214.3 [M+Hf R/T = 2.98 min

15 To a solution of 6-bromo-2,3-dihydro-isoindol-l-one (1 equiv) in dry dioxan (0.1 M) were added bis(pinacolato)diboron (1.1 equiv), potassium acetate (3.5 equiv) and dppf (0.05 equiv). The reaction mixture was degassed with nitrogen for 20 minutes. PdCl2(dppf) (0.05 equiv) was added to the reaction mixture, which was degassed for a further 5 minutes. The reaction mixture was heated to 70°C for 2 hours under nitrogen then heated to 120°C for 16
20 hours. The reaction mixture was partitioned between EtOAc and water. The aqueous phase was further extracted with EtOAc and the combined organic phases dried (MgSO,*), filtered and concentrated in vacuo. The residue was sonicated in EtOAc, the suspension was filtered onto a sintered funnel and the collected grey solid was dried and used without further purification.
25 6-(4,4,5,5-Tetramethyl-[l,352]dioxaborolan-2-yl)-2,3-dihydro-isoindol-l-one: (82 % yield, 29 % purity, main impurity being the boronic acid 43 %) m/z (LC-MS, ESP): 519.5 [2M+H]+ JUT = 3.38 min

WO 2008/023161

PCT/GB2007/003179

Procedure for the synthesis of 7-(4,4,5,5-Tetramethyl-[l^,2]dioxaborolan-2-yl)-3,4-dihydro-lH-benzo[e][l,4]diazepine-2,5-dione:

To a solution of 5-bromoisatoic anhydride (1 equiv) in water (1 M) was added glycine (1,4 equiv) and triethylamine (1 equiv) at room temperature. The reaction mixture was stirred at room temperature for 4 hours to give a cloudy solution. The reaction mixture was concentrated in vacuo. Acetic acid was added and the reaction mixture was stirred at 140°C
10 for 4.5 hours. The reaction mixture was cooled down slowly to room temperature. A precipitate was formed. The reaction mixture was diluted with diethyl ether then filtered through a sintered funnel to yield the desired product.
7-Bromo-3,4-dihydro-lH-benzo[e][l34]diazepine-2,5-dione: (75 % yield, 100 % purity) m/z (LC-MS, ESP): 255.2/257.2 [M+H]+ R/T = 2.67 min
15

To a solution of 7-bromo-3,4-dihydro-lH-benzo[e][l,4]diazepine-2,5-dione (1 equiv) in dry dioxan (0.1 M) were added bis(pinacolato)diboion (1.1 equiv), potassium acetate (3.5 equiv) and dppf (0.05 equiv). The reaction mixture was degassed with nitrogen for 20 minutes. 20 PdCl2(dppf) (0.05 equiv) was added to the reaction mixture, which was degassed for a further 5 minutes. The reaction was heated to 120 °C for 16 hours under nitrogen. The reaction mixture was partitioned between CHiCk/MeOH and water. The aqueous phase was further extracted with CH2Cl2/MeOH. The combined organic phases were dried (MgS04), filtered

WO 2008/023161

PCT/CB2007/003179

and concentrated in vacuo. The residue was sonicated in hexane/ CH2CI2, filtered, sonicated in CH2CI2 and filtered to yield the desired product.
7-(4)455,5-Tetramethyl-[l,3,2]dioxaborolan-2-yl)-3)4-dihydro-lH-benzo[e][l,4]diazepine-2,5-dione: (63 % yield, 85 % purity main impurity being the boronic acid 15 %) m/z (LC-MS, 5 ESP): 303.4 [M+H]+R/T = 3.08 min

10 To a solution of 2-amino-4-bromobenzoic acid (1 equiv) in DMA (0.23 M), were added ammonium chloride (7 equiv), HBTU (1 equiv) and diisopropylethylamine (2 equiv). The reaction mixture was stirred for 24 hours at room temperature. DMA was evaporated and the residue was purified by flash column chromatography onto silica gel eluting with a gradient of TBME/hexane to yield the desired product as a white solid.
15 2-Amino-4-bromo-benzamide: 40 % yield, 100 % purity) m/z (LC-MS, ESP): 215 [M+H]+ R/T = 3.00 min

20
To a solution of 2-amino-4-bromo-benzamide (1 equiv) in DMA (0.14 M) were added triethyl orthoformate (10 equiv) and trifluoroacetic acid (1 equiv). The reaction vessel was sealed and exposed to microwave radiation (160 °C, medium absorption setting) for 30 minutes. The 25 reaction mixture was concentrated in vacuo and the residue was filtered through a silica pad with 10 % methanol in ethyl acetate yielding the required product as a pale yellow solid.

WO 2008/023161

PCT/GB2007/003179

7-Bromo-3H-quinazolin-4-one: (71 % yield, 100 % purity) m/z (LC-MS, ESP): 268 [M+H]+ R/T = 2.94min

5 To a solution of 7-bromo-3H-quinazolin-4-one (1 equiv) in dioxane (0.04 M) were added bispinacolato diboron (2.2 equiv), potassium acetate (1.5 equiv), dppf (0.1 equiv) and PdCl2(dppf) (0.1 equiv). The reaction mixture was degassed with nitrogen for 5 minutes, sonicated and stirred at 120°C for 3 houTs. The reaction mixture was concentrated in vacuo. The residue was filtered through a Celite pad topped with silica with ethyl acetate. The
10 mother liquor was concentrated in vacuo yielding a brown solid which was further purified by flash column chromatography onto silica gel eluting with a gradient of methanol/diethyl ether (0 to 5 %) to yield the desired product as a white solid.
7-(4,4,5,5-Tetramethyl-[l,3,2]dioxaborolan-2-yl)-3H-quinazolin-4-one: (53 % yield, 61 % purity main impurity being the boronic acid 39 %) m/z (LC-MS, ESP): [M+H]+ R/T = min
15

To a solution of 6-bromo-2-oxindole (1 equiv) in NMP (0.05 M) were added bispinacolato diboron (2.4 equiv), potassium acetate (1.5 equiv), dppf (0.05 equiv) and PdCbfdppf) (0.05 equiv). The reaction mixture was stirred at 130°C for 3 hours and then concentrated in vacuo.
20 The residue was partitioned between water and ethyl acetate. The organic phase was dried over anhydrous sodium sulphate, filtered and concentrated in vacuo. The crude residue was purified by flash column chromatography on silica gel eluting with EtOAc/hexane (9 / 1), yielding the desired product as a red solid. 6-(4A5,5-Tetramethyl-[l,3,2]dioxaborolan-2-yl>l,3- 25 purity main impurity being the boronic acid 28 %) m/z (LC-MS, ESP): 260 [M+H]+ R/T = 3.51 min

WO 2008/023161

PCT/GB2007/003179

Procedure for the synthesis of S-(4,4J5,5-TetramethyH13>2]dioxaborolan-2-yl)-2,3-dihydro-isoindol-1-one:

5 4-Bromo-2-bromomethyl-benzoic acid methyl ester was prepared according to literature. A solution of 4-bromo-2~bromomethyl-benzoic acid methyl ester (1 equiv) in a 1:1 THF/MeOH mixture was treated by gentle bubbling of ammonia gas for 4 hours at room temperature. The reaction mixture was concentrated in vacuo. The residue was sonicated in water, filtered, then sonicated in diethylether and filtered to give the desired product as a 10 white solid.
5-Bromo-2,3-dihydro-isoindol-l-one: (81 % yield, 100 % purity) m/z (LC-MS, ESP): 2123/214.3 [M+H]+ R/T = 3.06 min

O 15 To a solution of 5-bromo-2,3-dihydro-isoindol-l-one (1 equiv) in dry dioxan (0.1 M) were
added bis(pinacolato)diboron (1.1 equiv), potassium acetate (3.5 equiv) and dppf (0.05
equiv). The reaction mixture was degassed with nitrogen for 20 minutes. PdC^dppf) (0.05
equiv) was added to the reaction mixture, which was degassed for a further 5 minutes. The
reaction mixture was heated to 70QC for 2 hours under nitrogen then heated to 120QC for 16
20 hours. The reaction mixture was partitioned between EtOAc and water. The aqueous phase
was further extracted with EtOAc and the combined organic phases dried (MgSC^), filtered
and concentrated in vacuo. The residue was dissolved in CH2CI2 and hexane was added. The

WO 2008/023161

PCT/GB2007/00317y

resulting suspension was filtered and the collected brown powder was dried and used without further purification.
5-(4J4,5,5-Tetramethyl-[l)3,2]dioxaborolan-2-yl)-2,3-dihydro-isoindol-l-one: (94 % yield, 76 % purity, main impurity being the boronic acid 13 %) m/z (LC-MS, ESP): 260.4 [2M-FH]+ 5 R/T = 3.51min
Procedures for the preparation of Examples la to Idu
10

R4 = (S)-3-methyI-morpholine
R2 = (S)-3-methyl-morpholine or cis-dimethyhnorpholine or 2-Ethyl-piperidine or
morpholine or thiomorpholine or 4-methylpiperazine
15 R7 = aryl or heteroaryl
Procedures for the Suzuki coupling:
The synthesis of the appropriate chloro-substrate has been described in the present document as intermediates. The appropriate pinacolate boron ester or boronic acids were prepared according to synthesis described in the present document (as intermediates) or commercially 20 available, typically from the following suppliers:
Sigma-Aldrich, Lancaster, Frontier Scientific, Boron Molecular, Jnterchim, Asymchcm, Combi-blocks, Apollo Scientific, Fluorochem, ABCR, Digital Speciality Chemicals.
Conditions A:
25 The appropriate chloro-substrate (1 equiv) was dissolved in a toluene/ethanol (1:1) solution (0.02 M). Sodium carbonate (2 equiv) and the appropriate pinacolate boron ester or boronic acid (1 equiv) were then added followed by tetrakis(triphenylphosphine) palladium0 (0.1 equiv). The reaction vessel was sealed and the mixture exposed to microwave radiation (140 °C, medium absorption setting) for 30 minutes. Upon completion the samples were filtered
30 through a silica cartridge, washed with EtOAc and then concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired products.

WO 2008/023161

PCT/GB20n?/00317!>

Conditions B:
A mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (2.4 equiv), the appropriate pinacolate boron ester or boronic acid (1.1 equiv) and tetrakis(triphenylphosphine) palladium0 (0.05 equiv) in o-butanol (0.03 M of chloro-substrate) 5 was stirred at 120 °C for 2 hours. Upon completion the samples were filtered through a silica cartridge, washed through with CH2CI2 and then concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired products. Conditions C: To a mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (2.4 equiv).
10 and the appropriate pinacolate boron ester or boronic acid (1.1 equiv) in acetonitrile/water (1:1) (0.041 M of chloro-substrate) was added tetrakis(triphenylphosphine) palladium0 (0.05 equiv). The reaction vessel was sealed and exposed to microwave radiation (150 °C, medium absorption setting) for 30 minutes under nitrogen atmosphere. Upon completion the samples were filtered through a silica cartridge, washed with CH2CI2 and methanol and then
15 concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired products. Conditions D:
To a mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (1.2 equiv). and the appropriate pinacolate boron ester or boronic acid (1.2 equiv) in acetonitrile/water
20 (1:1) (0.083 M of chloro-substrate) was added tetrakis(triphenylphosphine) palladium0 (0.05 equiv). The reaction vessel was sealed and exposed to microwave radiation (130 °C, medium absorption setting) for 25 minutes under nitrogen atmosphere. Upon completion the sample was purified by column chromatography on silica gel using a gradient MeOH/ CH2C12 to afford the desired product which was recrystallised from diethyl ether.
25 Conditions E:
To a mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (2.4 equiv), and the appropriate pinacolate boron ester or boronic acid (1.3 equiv) in acetonitrile/water (1:1) (0.041 M of chloro-substrate) was added tetrakis(triphenylphosphine) palladium0 (0.05 equiv). The reaction vessel was sealed and heated at 95 °C for 16 hours. Upon completion the
30 reaction mixture was partitioned between aqueous HCl and CH2CI2 and washed with aqueous HCl. Combined aqueous phase were extracted with CH2CI2 (2 x), neutralised with aqueous NaOH (2N) to give a cloudy solution that was extracted with CH2C12. Combined organic

WO 2008/023161

PCT/GB2007/003179

phases were washed with brine, dried (MgSCO4), filtered and concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 0 to 4 % MeOH in CH2CI2 to give the desired product. Conditions F: 5 To a mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (2.0 equiv), and the appropriate pinacolate boron ester or boronic acid (1.5 equiv) in acetonitrile/water (1:1) (0.028 M of chloro-substrate) was added tetrakis(triphenylphosphine) palladium0 (0.05 equiv). The reaction vessel was sealed and heated at 120 °C for 2 hours under nitrogen atmosphere. Upon completion the reaction mixture was partitioned between water and CH2CI2
10 and extracted with CH2CI2. Combined organic phases were dried (MgSO^, filtered and concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 0 to 4 % MeOH in CH2CI2 to give the desired product which was recrystallised from hexane/diethyl ether. Conditions CJ:
15 To a mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (3.0 equiv), and the appropriate pinacolate boron ester or boronic acid (1.05 equiv) in acetonitrile/water (1:1) (0.068 M of chloro-substrate) was added tetrakis(triphenylphosphine) palladium0 (0.05 equiv). The reaction vessel was sealed and heated at 100 DC for 5 hours under nitrogen atmosphere. Upon completion the reaction mixture was partitioned between brine and CH2CI2
20 and extracted with CH2CI2. Combined organic phases were dried (MgS04), filtered and concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 0 to 4 % MeOH in CH2CI2 to give the desired products which were recrystallised fromhexane/ CH2C12. Conditions H:
25 A mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (3.0 equiv), the appropriate pinacolate boron ester or boronic acid (1.1 equiv) and tetrakis(triphenylphosphine) palladium0 (0.05 equiv) in acetonitrile/water (0.1 M of chloro-substrate) was stirred at 100 °C for 8 hours. Upon completion the sample was concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired product.
30 Conditions I:
Conditions I were similar to conditions H apart form the heating method: 100 °C for 2 hours.

WO 2008/023161

PCT/GB2007/003179

Conditions J:
A mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (1.2 equiv), the appropriate pinacolate boron ester or boronic acid (1.2 equiv) and 5 tetrakis(triphenylphosphine) palladium0 (0.05 equiv) in acetonitrile/water (0.03 M of chloro-substrate) was stirred at 100 °C for 2 hours. Upon completion the sample was concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired product. Conditions K: Conditions K were similar to conditions G apart form the heating method: 100 °C for 16
10 hours.
Conditions L:
To a mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (2.5 equiv), and the appropriate pinacolate boron ester or boronic acid (1.10 equiv) in acetonitrile/water (1:1} (0.041 M of chloro-substrate) was added tetrakis(triphenyiphosphine) palladium0 (0.05
15 equiv). The reaction vessel was sealed and exposed to microwave radiation (100 °C, medium absorption setting) for 90 minutes. Upon completion the reaction mixture was partly concentrated. The residue was partitioned between water and ethyl acetate and extracted with ethyl acetate and n-butanol. Combined organic phases were dried (MgS04), filtered -and concentrated in vacuo. The crude residue was purified by column chromatography on silica
20 gel eluting with 30 to 10 % hexane in ethyl acetate to give the desired product which was recrystallised from hexane/ CH2CI2. Conditions M:
A mixture of the appropriate chloro-substrate (1 equiv), cesium fluoride (3.0 equiv), the appropriate pinacolate boron ester or boronic acid (1.1 equiv) and
25 tetrakis(triphenylphosphine) palladium0 (0.05 equiv) in acetonitrile/water (0.09 M of chloro-substrate) was stirred at 115 °C for 48 hours. Upon completion the sample was concentrated in vacuo to half original volume. The residue was partitioned between water and CH2CI2. Organic phase was dried (MgSCU), filtered and concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 0 to 100 % ethyl acetate in
30 hexane to give the desired product.

WO 2008/023161

PCT/GB2007/IM3179

Conditions N:
A mixture of the appropriate chloro-substrate (1 equiv), tripotassium phosphate (1.5 equiv), the appropriate pinacolate boron ester or boronic acid (1.05 equiv) and bis(tri-t-butylphosphine) palladium (0.05 equiv) was suspended in dioxane (0.16 M of chloro-5 substrate). The reaction vessel was sealed and exposed to microwave radiation (170 °C, medium absorption setting) for 45 minutes. Upon completion the sample was concentrated in vacuo. The residue was partitioned between water and CH2CI2. The organic phase was dried (MgSO 10 desired product. Conditions Q:
A mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (2.5 equiv), the appropriate pinacolate boron ester or boronic acid (1.1 equiv) and tetralds(triphenylphosphine) palladium0 (0.05 equiv) in n-butanol (0.068 M of chloro-
15 substrate) was stirred at 95 °C for 15 minutes. Upon completion, the residue was partitioned between ethyl acetate and brine. Organic phase was dried (MgSCU), filtered and concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 30 to 100 % ethyl acetate in hexane to give the desired product which was recrystallised from ethyl acetate/hexane.
20 Conditions P:
To a mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (2.0 equiv), and the appropriate pinacolate boron ester or boronic acid (2.0 equiv) in acetonitrile/water (1:1) (0.041 M of chloro-substrate) was added tetrakis(triphenyIphosphine) palladium0 (0.05 equiv). The reaction vessel was sealed and exposed to microwave radiation (120 °C, medium
25 absorption setting) for 10 minutes under nitrogen atmosphere. Upon completion the samples were filtered through a silica cartridge, washed through with CH2CI2 and the concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired product. Conditions Q: A mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (2.5 equiv), the
30 appropriate pinacolate boron ester or boronic acid (1.1 equiv) . and tetrakis(triphenylphosphine) palladium0 (0.05 equiv) were dissolved in w-butanol (0.056 M of chloro-substrate). The reaction vessel was sealed and exposed to microwave radiation (150

WO 2008/023161

PCT/GB2007/003179

°C, medium absorption setting) for 30 minutes. Upon completion the samples were filtered through a silica cartridge, washed with CH2C12 and methanol and then concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with ethyl acetate and then 5 % MeOH in CH2CI2 to give the desired product. 5 Conditions R:
A mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (2.5 equiv), the appropriate pinacolate boron ester or boronic acid (1.2 equiv) and tetrakis(triphenyIphosphine) palladium0 (0.05 equiv) in acetonitrile/water (0.05 M of chloro-substrate) was stirred at 115 °C for 1.5 hours. Upon completion the crude reaction was filtered
10 and the filtrate was concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 5 to 20 % MeOH in CH2C12 to give the desired product. Conditions S: A mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (10.0 equiv), the
15 appropriate pinacolate boron ester or boronic acid (1.2 equiv) and tetrakis(triphenylphosphine) palladium0 (0.05 equiv) in acetonitrile/water (0.1 M of chloro-substrate) was stirred at 100 °C for 2 hours. Upon completion the reaction mixture was partitioned between water and CH2CI2 and extracted with CH2CI2. Combined organic phases were dried (MgS04),, filtered and concentrated in vacuo. The crude residue was purified by
20 column chromatography on silica gel eluting with 0 to 5 % MeOH in CH2CI2 to give the desired product which was recrystallised from hexane/ CH2CI2. Conditions T:
A mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (2.0 equiv), the appropriate pinacolate boron ester or boronic acid (2.0 equiv) and
25 tetrakis(triphenylphosphine) palladium0 (0.05 equiv) was dissolved in acetonitrile/water (0.02 M of chloro-substrate). The reaction vessel was sealed and exposed to microwave radiation (130 °C, medium absorption setting) for 30 minutes. Upon completion the sample was concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 0 to 5 % MeOH in CH2CI2 to give the desired product.
30 Conditions U:
A mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (3.0 equiv), the appropriate pinacolate boron ester or boronic acid (1.0 equiv) and

WO 2008/023161

PCT/GB20(I7/003179

tetrakis(triphenylphosphine) palladium0 (0.05 equiv) in acetonitrile/water (0.1 M of chloro-substrate) was stirred at 110 °C for 8 hours. Upon completion the reaction mixture was partitioned between water and CH2CI2 and extracted with CH2O2. Combined organic phases were washed with brine, dried (MgSC), filtered and concentrated in vacuo. The crude residue 5 was purified by column chromatography on silica gel eluting with 0 to 2 % MeOH in CH2CI2 to give the desired product which was recrystallised from hexane/ CH2CI2. Conditions V:
A mixture of the appropriate chloro-substrate (1 equiv), cesium fluoride (3.0 equiv), the appropriate pinacolate boron ester or boronic acid (1 equiv) and tetrakis(triphenylphosphine)
10 palladium0 (0.05 equiv) in acetonitrile/water (0.1 M of chloro-substrate) was stirred at 100 °C for 16 hours. The reaction mixture was partitioned between water and CH2CI2 and extracted with CH2CI2. The organic phase was dried (MgS04), filtered and concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 0 to 5 % MeOH in CH2CI2 to give the desired product which was recrystallised from hexane/CHaCb.
15 Conditions W:
A mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (2.5 equiv), the appropriate pinacolate boron ester or boronic acid (1 equiv) and tetrakis(triphenylphosphine) palladium0 (0.05 equiv) was dissolved in acetonitrile/water (0.04 M of chloro-substrate). The reaction vessel was sealed and exposed to microwave radiation (110 °C, medium absorption
20 setting) for 10 minutes. The crude residue was purified by column chromatography on silica gel eluting with 0 to 2 % MeOH in TBME to give the desired product.
Table 1;


WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB20n7/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB20O7/003179





WO 2008/023161

PCT7CB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB20O7/O03179





WO 2008/023161

PCT/GB2007/U03179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003I79





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179


Note:
The following examples were synthesized from the corresponding boronic acids: laa, lab,
lac, lad, lae, laf, lag, lah, lai, laj, lak, lal, lam, Ian, lao, lap, laq, las, lat, lau, lav,
5 law, lax, lay, laz, lba, lbb, Ibd, lbe, lbk, lbl, lbm, lbn, lbo, lbp, lbq, lbr, lbs, lbt, lbu,

►008/023161

PCT/GB2007/003179

lbv, lbw, lbx, lby, lbz, lea, leb, lec, led, lef, leg, lch, lei, lej, Id, le, If, lg, lh, li, Ij, Ik, In, lo, Ip, lr, It, lw, lx, ly len, lco, lep, les, lev and lz.
The following Examples were synthesized from the corresponding pinacolate boron esters: la-c, Ick, lcl, 1cm. lcq, lcr, let, leu, lar, lbf, Ice, lm, lq, Is, luand lv. 5
NMR data for Example In
lH NMR (300 MHz, CDCI3) 8 ppm 8.88 (ArH, d, J - 2.20 Hz, IH), 8.55 (ArH, dd, J = 8.70, 2.45 Hz, IH), 8.04 (ArH, d, / = 8.43 Hz, IH), 7.42 (ArH, d, J = 8.44 Hz, IH), 6.88 (ArH, d,./ = 8.70 Hz, IH), 5.01-4.90 (CH, m, IH), 4.65 (CH, d, J= 13.12 Hz, IH), 4.40 (CH, d, J =
10 6.68 Hz, IK), 4.04 (OCH3 + CH2, s, 5H), 3.96-1.69 (CH2, m, 7H), 3.6Q (CH, dt, J= 11.86, 11.60, 2.67 Hz, IH), 3.40 (CH, dt,J= 13.01,12.73, 3.60 Hz, IH), 1.50 (CH3, d, J~ 6.78 Hz, 3H), 1.39 (CH3, d, J= 6.81 Hz, 3H)
!3CNMR(75 MHz, CDCI3) Sppm 165.41,165.29,162.98,160.10,160.01, 146.58,138.51, 134.81,128.05,112.42,110.84, 104.75,71.29, 70.92, 67.26, 66.92, 53.75, 52.87, 46.94,
15 44.43, 39.33, 14.73 and 14.36.
NMR. data for Example lu
'H NMR (300 MHz, CDCI3) 8 ppm 8.80 (ArH, d, J = 1.91 Hz, IH), 8.39 (ArH, dd, J = 8.66,
2.39 Hz, IH), 7.96 (ArH, d, J = 8.48 Hz, IH), 7.35 (ArH, d, J = 8.49 Hz, IH), 6.59 (ArH, d, J 20 = 8.66 Hz, IH), 4.91 (CH, dd, J = 4.15, 1.62 Hz, IH), 4.78 (NH2, s, 2H), 4.67-4.55 (CH, m,
IH), 4.34 (CH, d, J = 6.88 Hz, IH), 4.04-3.91 (CH2, m, 2H), 3.90-3.64 (CH2, m, 7H), 3.62-
3.49(CH2)m, IH), 3.44-3.29 (CH2,m, IH), 1.45 (CH3, d,J= 6.77 Hz, 3H), 1.34(CH3, d,J =
6.82 Hz, 3H)
13CNMR(75MHz,CDCl3)5ppm 165.54,163.10,160.45,160.13, 159.45, 148.10, 137.82, 25 134.76, 125.17, 112.16, 108.45, 104.59, 71.44, 71.06,. 67.41, 67.07, 52.98, 47.05, 44.56,
36.46,14.84 and 14.75.
NMR data for Example lag
'H NMR (300 MHz, CDC13) 8 ppm 8.59 (ArH, dd, J = 7.33, 2.43 Hz, IH), 8.40 (ArH, ddd, J 30 = 8.53, 5.03,2.47 Hz, IH), 7.97 (ArH, d, J= 8.42 Hz, IH), 7.42 (ArH, d,./= 8.46 Hz, IH), 7.20-7.10 (ArH, m, IH), 4.84 (CH, dd, J = 3.67,2.96 Hz, IH), 4.53 (CH, d, J = 12.77 Hz, IH), 4.33 (CH2, d, J= 6.83 Hz, IH), 3.99-3.89 (CH2, m, 2H), 3.86-3.77 (CH2, m, 4H), 3.75-

WO 2008/023161

PCT/GB20II7/003179

3.65 (CH2) m, 5H), 3.67-3.32 (CH2j m, 3H), 3.57-3.45 (CH2, m, IH), 3.36-3.26 (CH2, m, IH), 1.42 (CH3, d, J = 6.78 Hz, 3H), 1.30 (CH3, d, J = 6.82 Hz, 3H)
13CNMR(75MHz,CDCl3) 8ppm 165.26,164.26,162.74,160.29,159.93,135.52,135.11, 133.47,133.34,130.89,116.84,116.51,113.11,105.11, 71.26, 70.91, 67.11, 66.91, 62.20, 5 52.79,47.02, 44.46, 43.02, 39.36,14.77 and 14.37.
NMR data for Example 1 aq
'H NMR (300 MHz, CDC13) 8 ppm 8.61 (ArH, t, J = 1.46,1.46 Hz, IH), 8.33 (ArH, d, J =
7.84 Hz, IH), 8.06 (ArH, d, J = 8.37 Hz, IH), 7.90 (ArH, s, IH), 7.62 (ArH, d, J= 7.84 Hz, 10 IH), 7.44 (ArH, d, J =8.38 Hz, IH), 5.30 (CH2, s, IH), 4.97-4.84 (CH2, m, IH), 4.64-4.52
(CH2, m, IH), 4.45-4.34 (CH2j m, JH), 4.06-3.94 (CH2, m, 2H), 3.93-3.64 (CH2, m, 8H),
3.61-3.51 (CH2, m, IH), 3.45-3.30 (CH2, m, IH), 3.19 (CH2, d, J= 4.84 Hz, 2H), 1.49 (CH3,
d, J = 6.78 Hz, 3H), 1.36 (CH3, d, J = 6.82 Hz, 3H).
i3CNMR(75 MHz, CDC13) 8 ppm 160.22,140.70,140.01, 134.25,131.82, 129.24, 128.02, 15 126.34, 113.12,105.41,104.65, 71.23, 70.87, 66.88, 61.03, 52.85,47.04,45.34, 44.42, 39.35,
14.78 and 14.38.
"NMR data for Example lar
lH NMR (300 MHz, CDC13) 8 ppm 8.81-8.73 (ArH, m, 2H), 8.14-7.99 (ArH, m, 3H), 7.48 20 (ArH, d, 7 =8.35 Hz, IH), 5.02-4.89 (CH,m, IH), 4.69-4.59 (CH2,m, IH),4.4I (CH,d,J =
6.84 Hz, IH), 4.08-3.96 (CH2, m, 2H), 3.82 (H2, dddd, J= 19.69, 14.05, 6.26, 3.77 Hz, 7H),
3.65-3.53 (CH2, m, IH), 3.48-3.31 (CH2, m, IH), 1.51 (CH3, d, J = 6.78 Hz, 3H), 1.38 (CH3,
d,J=6.83Hz,3H)
13CNMR(75MHz,CDCl3)5ppm 165.29,162.96,160.03,159.82, 150.36, 145.80, 135.20, 25 121.83,113.02, 105.93, 71.24, 70.87,67.21, 66.87,52.88, 46.99, 44.45, 39.35, 14.76 and
14.41.
NMR Data for Example las
lH NMR (300 MHz,CDCl3) 8 ppm 8.67 (ArH, t, J = 1.54,1.54 Hz, IH), 8.29 (ArH, dd, J -30 6.60,1.28 Hz, IH), 8.07 (ArH, d, J= 8.41 Hz, IH), 8.03-7.98 (ArH,m, 2H), 7.62-7.49 (ArH, m, 2H), 4.98-4.89 (CH, m, br, IH), 4.67-4.59 (CH, m, br, IH), 4.41 (CH, d, J = 6.78 Hz, IH),

WO 2008/023161

PCT/GB2007/003179

4.05-3.66 (CH2, m, 10H), 3.64-3.34 (CH2, m, 3H), 1.75 (s, 1.5H), 1.50 (CH3> d, J = 6.78 Hz, 3H), 1.38 (CH3s d, J= 6.82 Hz, 3H).
I3CNMR(75 MHz,CDCl3) 8ppm 168.86,165.34,162.90,161.21,160.01,138 97,135.00, 133.74,131.09,129.23,128.98,126.52,113.20, 105.20, 100.00, 71.23, 70.89, 67.22, 66.90, 5 52.82,46.97,44.45, 39.34,14.75 and 14.36
NMR data for Example lat
'H NMR (300 MHz, CDC13) 8 ppm 8.06 (ArH, ds ./= 2.05 Hz, IH), 7.98 (ArH, d,J= 8.41
Hz, IH), 7.86-7.79 (ArH, m, IH), 7.46-7.33 (ArH, m, 3H), 7.23 (NH, S, IH), 4.83 (CH, dd,./ 10 = 3.58,2.50 Hz, IH), 4.56-4.46 (CH, m, IH), 4.32 (CH, d, J= 6.74 Hz, IH), 3.93-3.89 (CH2,
m, 2H), 3.88-3.77 (CH2, m, 2H)3 3.76-3.58 (CH2, m, 5H), 3.49 (CH2, dt,J= 11.76,11.38,
2.76 Hz, IH), 3.35-3.20 (CH2, m, IH), 2.89 (SCH3, s, 3H), 1.42 (CH3, d, J= 6.78 Hz, 3H),
1.27 (CH3,d,y = 5.25, Hz, 3H)
,3CNMR(75MHz,CDCl3)6ppm 165.32,162.87,161.30, 159.96, 140.41, 137.57,135.01, 15 129.92, 124.55,122.25,120.57,113.32, 105.24, 71.25, 70.90, 67.22, 66.91, 52.86,46.99,
44.42, 39.40, 31.60, 22.66, 14.77 and 14.12.
NMR data for Example lax
'H NMR (300 MHz, CDCI3) 5 ppm 8.88 (ArH, t, J = 1.52,1.52 HZ} IH), 8.32-8.25 (ArH, m,
20 IH), 8.13-8.06 (ArH, m, IH), 7.99 (ArH, d, J= 8.42 Hz, IH), 7.53-7.39 (ArH, m, 2H), 4.90-4.80 (CH, m, IH), 4.58-4.48 (CH, m, IH), 4.33 (CH, d, J= 6.90 Hz, IH), 3.95-3.65 (CH2, + OH m,8H), 3.64 (CH2, d, J =2.85 Hz, 2H), 3.56-3.45 (CH,m, 1BX331 (CB,d,J = 3.67 Hz, IH), 1.42 (CH3, d,J= 6.79 Hz, 3H), 1.29 (CH3, d, J = 6.81 H%, 3H) 13CNMR(75MHz,CDCl3)5ppm 169.20,165.22,161.32,159.85,139.91,135.01, 131.39,
25 129.70, 128.71,113.31, 70.90, 67.IO, 52.80,47.07,44.42,39.36, U.77 and 14.37.
NMR data for Example laz
'H NMR (300 MHz, CDC13) 5 ppm 8.04-7.95 (ArH, m, 2H), 7.87 (ArH, d, J= 8.54 Hz, IH), 7.32 (ArH, d, J= 8.55 Hz, IH), 6.71-6.64 (ArH, m, 2H), 4.92-4.81 (CH, m, IH), 4.57 (CH, d, & frr, 2H), 4.29 (CU2, d, J= 7.10}JZy 2H), 3.91 (CH2, m, 2H)S 3.82-3.58 (CH2 + NH2, m, M), 3.48 (CH2,dd,J = 11.36,2.76 HZ; 1H), 3.33 (CH2, dd,J= 13.48,3.61 Hz, IH), 1.39 (CH3, d, J = 6.78 Hz, 3H), 1.28 (CH3, d, J = 6.82 Hz, 3H)

WO 2008/023161

PCT/GB2007/003179

13CNMR(75 MHz, CDC13) 5ppm 165.28,162.18,148.68,135.36,129.54,119.67, 114.75, 112.63, 104.43,104.00, 71.29, 70.94, 67.27,67.12, 66.95, 52.78,44.45,39.15, 14.74 and 14.37.
5 NMR data for Example lba
'H NMR (300 MHz, CDC13) 5 ppm 8.69 (ArH, t, J= 1.58,1.58 Hz, IK), 8.44-8.33 (ArH, m, IH), 8.11-8.03 (ArH, m, IH), 7.99 (ArH, d, J = 8.42 Hz, IH), 7.57-7.38 (ArH, m, 2H), 4.87 (CH2, dd, J = 4.84,0.43 Hz, IH), 4.57 (CH, d, J = 12.80 Hz, IH), 4.31 (CH2, t,J= 6.72,6.72 Hz, IH), 3.94 (CH2) dd, J = 11.15,3.26 Hz,2H), 3.90 (OCH3; d, J= 6.23 Hz, 3H), 3.83 -
10 3.62 (CH2j m, 7H), 3.57-3.45 (CH2, m, IH), 3.39-3.24 (CH2, m, IH), 1.42 (CH3, d, J = 6.78 Hz, 3H), 1.30 (CH3, d, J= 6.81 Hz, 3H)
,3CNMR(75MHz,CDCl3) Sppm 166.92,165.41,162.93,161.43,161.01,139.13,134.91, 132.51, 130.87, 130.57,128.87,113.26,105.16, 71.29, 70.91, 67.25, 66.91, 52,86, 52.18, 46.96,44.45, 14.77 and 14.37.
15
NMR data for Example Ibc
!H NMR (300 MHz, DMSO) 5 ppm 8.37 (ArH, dd, J = 7.40, 2.26 Hz, IH), 8.20 (ArH, d, J = 8.50 Hz, IH), 8.14-8.05 (ArH, m, IH), 7.62 (ArH, d, J= 8.51 Hz, IH), 7.29 (ArH, dd, J-9.77, 8.71 Hz, IH), 5.42 (CH, t,J= 5.76, 5.76Hz, IH), 4.77 (CH, dd, J= 6.57, 1.98 Hz, IH),
20 4.65 (CH2OH, d, J= 5.67 Hz, 2H), 4.51-4.37 (CH2) m, 2H), 3.98-3.83 (CH2, m, 3H), 3.80-3.70 (CH2, m, 2H), 3.69-3.56 (CH2, m, 4H), 3.45 (CH2, dt, J- 11.86, 11.77,2.75 Hz, IH), 3.30- 3.16 (CH2, m, 3H), 1.38 (CH3, d, J = 6.75 Hz, 3H), 1.25 (CH3, d, J =6.75 Hz, 3H) l3C NMR (75 MHz, DMSO) 5 ppm 164.91,162.60, 160.18,159.82, 136.10, 134.86, 130.19, 129.99, 128.61, 128.27, 128.15,115.85, 115.57, 113.00,104.80,70.89,70.66,66.84,66.67,
25 52.29,46.76, 44.34,14.84 and 14.34.
NMR data for Example lbd
lH NMR (300 MHz, DMSO) 5ppm 8.63 (ArH, t,J= 1.49, 1.49 Hz, IH), 8.26 (ArH, d,,/ -= 7.95 Hz, IH), 8.17 (ArH, d, J = 8.46 Hz, IH), 7.91-7.80 (ArH, m, IH), 7.62 (ArH, dd, J = 30 14.96, 8.10 Hz, 2H), 7.37 (NH2, s, 2H), 4.69 (CH, dd, /= 6.21,1.34 Hz, IH), 4.35 (CH2, d, J = 13.74 Hz, 2H), 3.91-3.74 (CH2, m, 3H), 3.73-3.46 (CH2, m, 6H), 3.36 (CH2, dt, J= 11.82,

WO 2008/023161

PCT/GB2007AJ03179

11.71,2.49 Hz, 1H),2.41 (CH2,td, J= 3.46,1.69,1.69 Hz, IH), 1.30(CH3)d, J-6.74 Hz, 3H), U7 (CHJf d, J= 6.75 Hz, 3H)
,3CNMR(75 MHz)8ppm 164.85,162.63,159.86,159.49,145.34,139.35,136.44,130.73, 129.98,127.34,124.96, 113.18,105.38,79.87,79.43, 78.99, 70.89, 70.67,66.85, 66.67, 5 52.29,46.79, 44.37, 14.88 and 14.41.
NMR data for Example lbk
'HNMR (300 MHz, CDC13) 8 ppm 8.19 (ArH, dd, J = 7.62,2.22 Hz, IH), 8.12 (ArH, ddd, J
= 8.54, 5.03,2.25 Hz, IH), 8.01 (ArH, d, J= 8.41 Hz, IH), 7.38 (ArH, d, J = 8.43 Hz, Ui), 10 7.24-7.19 (ArH, m, IH), 6.83 (NH, s, br, IH), 4.98-4.85 (CH, m, IH), 4.67-4.55 (CH, m, IH),
4.36 (CH2f d,J = 6.95 Hz, IH), 4.06-3.93 (CH2, m, 2H), 3.91-3.65 (CH2, m, IH), 3.53 (CH2,
dd, /=* 11,40, 2.69 Hz, IH), 3.44-3.28 (CH2, m, IH), 3.07 (SCH3, s, 3H)5 1.47 (CH3, d, J =
6.77 Hz, 3H), 1.34 (CH3, d, J = 6.81 Hz, 3H)
UCNMR(75 MHz,CDCl3) 8ppm 165.45,162.93,160.61,160.12,157.28, 153.99, 136.32, 15 135.15,123.51,116.31,116.05,113.26,105.18,71.39,71.01,67.36,67.01,53.00,47.07,
44.51,39.44,31.71,22.77 14.86, and 14.95.
Compounds were also synthesized according to the following procedures:
20 Procedure for the synthesis of 5-[2,4-bis-((S)-3-methyl-morphoIin-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-2-isopropoxy-benzamide (Example lew)

5-[2,4-Bis-((S)-3-memyl-morpholm--4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-2-isopropoxy-benzonitrile (1 equiv) was added portionwise to concentrated H2S04 (0.1 M substrate in 25 acid). The reaction was heated to 90 °C and maintained at this temperature until all starting material had dissolved to give a bright red solution. The mixture was cooled and water (2 reaction volumes) added dropwise, then the solution was neutralized by careful addition of

WO 2008/023161

PCT/GB2007/003179

solid NaOH until pH 4-5 was attained. The mixture was cooled and neutralised by addition of 2N NaOH and then extracted using EtOAc (2X10 reaction volumes). The combined extracts were dried (MgSO4, filtered and concentrated in vacuo to give a crude residue which was purified by flash chromatography (S1O2) using MeOH/DCM - 0:100 going to 5:95 as eluent 5 to give the desire product as a yellow powder.
5-[2J4-Bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2J3-d]pyrimidin-7-yl]-2-isopropoxy-benzamide: (53 % yield, 100 % purity) m/z (LC-MS,ESP):507.5 [M+H]\ R/T = 3.01 min)
10 Procedure for the synthesis of 5-[2,4-bis-((S)-3-methyI-morphoIin-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-2-hydroxy-benzamide (Example lex)

5-[2,4-Bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidin-7-yI]-2-isopropoxy-
15 benzonitrile(l equiv) was added portionwise to concentrated H2S04 (0.1 M substrate in acid). The reaction was heated to 90 °C and maintained at this temperature until all starting material had dissolved to give a bright red solution. The mixture was cooled and water (2 reaction volumes) added dropwise, then the solution was neutralized by careful addition of solid NaOH until pH 4-5 was attained. The mixture was cooled and neutralised by addition of
20 2N NaOH and then extracted using EtOAc (2 X 10 reaction volumes). The combined extracts were dried (MgSO4 filtered and concentrated in vacuo to give a crude residue which was purified by flash chromatography (Si02) using MeOH/DCM - 0:100 going to 5:95 as eluent to give the desire product as a yellow powder.
25 5-[2J4-Bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2)3-d]pyrimidin-7-yl]-2-hydroxy-
benzamide: (44 % yield, 100 % purity) m/z (LC-MS,ESP):465.4 [M+H]", R/T = 2.70 min)

WO 2008/023161 PCT/GB2007/003179
Procedure for the synthesis of 5-[2,4-bis-((S)-3-m ethyl-mo rpholin-4-yl)-pyrido [2,3-d]pyrimidin-7-yIl-pyridine-2-carboxyIic acid amide (Example ley)

Synthesis of 5~[2,4-bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2)3-d]pyrimidin-7-yl]-pyridine-
2-carbonitrile was carried out as follows:-
To the appropriate chloro-substrate (1 equiv), potassium carbonate (3 equiv) and the
10 appropriate boronic acid or pinacolate boron ester (1.1 equiv) and
tetrakis(triphenylphosphine) palladium0 (0.05 equiv) which were dissolved inN,N-dimethylacetamide (0.17 M of chloro-substrate). The mixture was degassed with nitrogen, sealed and exposed to microwave radiation (130 °C, medium absorption setting) for 15 minutes. The mixture was concentrated in vacuo and then suspended in t-butylmethyl ether,
15 filtered and dried to give the desired product.
5-[2>4-Bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-pyridine-2-carbonitrile: (84 % yield, 93 % purity) m/z (LC-MS,ESP):191.3 [M+Hf, R/T = 2.29 min)

WO 2008/023161 PCT/GB2007/003179
To a suspension of 5-[2,4-bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-pyridine-2-carbonitriIe (1 equiv) in concentrated H2SO4. The mixture was heated to 90 °C until a pale brown solution formed. The mixture was allowed to cool and then basified with 50 % w/w NaOH solution. The aqueous mixture was extracted using EtOAc (3X2 reaction 5 volumes). The combined organic fractions were dried (MgSCU), filtered and concentrated in vacuo to give a pale yellow solid which was triturated with EtOAc to give the desire product.
5 -[2,4-Bis-((S)-3 -methyl-morpholin-4-y l)-pyrido [2,3 - d]pyrimidin-7-y l]-pyridine-2-carboxy lie acid amide: 93 % yield, 96 % purity) m/z (LC-MS,ESP):450.4 [M+H]+, R7T = 3.72 min) 0
Procedure for the synthesis of 4-[2,4-bis-((S)-3-methyl-morphoUn-4-yl)-pyrido[2,3-d]pyrimidin-7-yIJ-pyridia-2-ylanime (Example lcz)

15 To a 1.2 M solution of compound lau (1 equiv) in THF was added hydrazine hydrate (9 reaction volumes). The reaction vessel was sealed and exposed to microwave radiation (115 °C, medium absorption setting) for 2 hours. Upon completion, the reaction mixture was extracted with EtOAc (2X1 reaction volume). The organic fractions were combined, dried (MgSCU), filtered and concentrated in vacuo to give the desired product in suitably clean form
20 for use in subsequent reactions.
{442,4-Bis^(S)-3-memyl-moipholin-4-yl)-pyrido[23-d]pyrmiidm-7-yll-pyridin-2-yl}-hydrazine 7-(494,5,5-Tetramethyl-[l,3,2]dioxaborolan-2-yl)-2H-phthalazin-l-one: (77 % yield, 84 % purity) m/z (LC-MS,ESP):437.4 [M+Hf, TUT = 2.23 min) 25
A 0.12 M solution of {4-[2,4-bis-((S)-3-methyl-morpholin-4-yl)-pyrido[233-d]pyrimidin-7-y l]-pyridin-2-y 1} -hydrazine 7-(4,4,5,5-TetramethyI-[1 ,3,2]dioxaborolan-2-yl)-2H-phrhalazin-i-one (1 equiv) in EtOH was added to a a glass lined autoclave which contained activated Ra-Ni. The reaction was maintained under 5 bar H2 for 30 hours. Upon completion, the mixture

WO 2008/023161

PCT/GB2007/003179

was filtered through a pad of Celite™ and the filtrate concentrated in vacuo. The resulting crude residue was purified by reverse phase flash chromatography using 5:95 -0.1 % TFA/MeCN:0.1 % TFA/H2O as eluent to give the desire product as a yellow powder.
5 4-[2,4-Bis-((S)-3-memyl-morpholin^-yl)-pyrido[23-d]pyrimidin-7-yl]-pyridm-2-ylamine: (70 % yield, 100 % purity) m/z (LC-MS,ESP):422 [M+Hf, R/T = 2.25 min)
Procedure for the synthesis of 4-[2,4-bis-((S)-3-methyl-morphoUn-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-2-fluoro-benzamide (Example Ida).
10


To the appropriate chloro-substrate (1 equiv), potassium carbonate (2.5 equiv) and the appropriate boronic acid or pinacolate boron ester (1.1 equiv) and tetrakis(triphenylphosphine) palladium0 (0.05 equiv) which were dissolved in MeCN/H20 15 (0.03 M of chloro-substrate). The mixture was degassed with nitrogen, sealed and exposed to microwave radiation (110 °C, medium absorption setting) for 25 minutes. The mixture filtered and the precipitate collected, and recrystallised from MeCN/IfcO to give the desired product.
4-[2,4-Bis-((S)-3-memyl-morpholin-4-yl)-pyrido[233-d]pyrimidin-7-yI]-2-fluoro-benzonitiile: 20 (49 % yield, 87 % purity) m/z (LC-MS,ESP):449 [M+H]+, R/T = 2.93 min)

WO 2008/023161

PCT/GB2007/003179

4-[2,4-Bis-((S)0-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-2-fluoro-benzomtrile (1 equiv) was dissolved concentrated sulfuric acid (0.15 M substrate in acid). The reaction was heated rapidly to 90 °C for 5 minutes before cooling the mixture and quenched, carefully, with solid NaOH until the solution was basic. The mixture was extracted with EtOAc/nBuOH 5 (2X1 reaction volume —1:1 ratio). The organic extracts were combined, dried (MgSCO4), filtered and concentrated in vacuo to give a residue which was further purified using flash chromatography (Si02) with TBME going to TBME/MeOH (95:5) as eluent, the give the title compound as a yellow solid.
10 4-[2,4-Bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2)3-d]pyrirrudm-7-yl]-2-fluoro-benzarnide Synthesis: (71 % yield, 99 % purity) m/z (LC-MS3ESP):467 [M+H]+, R/T = 2.60 min)
Procedure for the synthesis of 5-[2,4-bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-lH-pyridin-2-one (Example ldb).


To a 0.2 M solution of compound lah (1 equiv) in DMA was added a 1.6 M aqueous solution of sodium hydroxide (5 equiv). The reaction vessel was sealed and exposed to microwave radiation (110°C, medium absorption setting) for 10 minutes. The reaction mixture was 20 concentrated in vacuo. The residue was suspended in water and sonicated to give a turbid solution, washed with TBME then cooled and neutralised with 2M HC1. forming a yellow precipitate. The precipitate was filtered and washed with water and TBME and dried to give the desired product.
25 5-[2)4-Bis-((S)-3-memyl-morpholm-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-lH-pyridin-2-one: (69 % yield, 96 % purity) m/z (LC-MS,ESP):423 [M+H]+, R/T =* 3.60 min)

WO 2008/023161

PCT/GB2007/003179

Procedure for the synthesis of 5-[2,4-bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-dlpyrimidin-7-yI]-lH-pyridin-2-one (Example ldc).

5 To the compound lah (1 equiv) was added a solution of 40 % methylamine in methanol (100 equiv). The reaction vessel was sealed and exposed to microwave radiation (115°C, medium absorption setting) for 30 minutes. The solution was concentrated in vacuo to yield a yellow solid. The crude residue was then purified by preparative HPLC to give the desired product. {5-[2,4-Bis-(3-memyl-moiphoUn-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-pyridin-2-yl}-methyl-10 amine: (61 % yield, 99 % purity) m/z (LC-MS,ESP):436 [M+Hf, R/T = 3.34 min)
NMR data for Example ldc
lH NMR (300 MHz, CDCI3.6 ppm 8.69 (ArH, d, J = 2.06 Hz, IH), 8.56 (ArH, dd, J = 9.02,
2.32 Hz, IH), 7.97 (ArH, d, J = 8.47 Hz, IH), 7.33 (ArH, d, J = 8.48 Hz, IH), 6.59 (ArH, d, 15 J = 9.03 Hz, IH), 5.92 (NH, s, br, IH), 4.90 (CH2, dd, J = 5.85,0.41 Hz, IH), 4.59 (CH2, d, J
= 12.53 Hz, IH), 4.41-4.29 (CH2, m, IH), 4.05-3.93 (CH2, m, 2H), 3.90-3.62 (CH2, m, 8H);
3.62-3.50 (CH2, m, IH), 3.43-3.31 (CH2,m, IH), 3.00 (NCH3, s, 3H), 1.47 (CH3, d,J = 6.7S
Hz, 3H), 1.35 (CHj, d, J = 6.82 Hz, 3H)
l3CNMR(75 MHz, CDClj 5ppm 167.59,165.30,162.90,159.99,158.84, 144.22,139.19, 20 134.85, 123.35, 111.65, 106.51, 104.57,71.28,70.91,67.24,66.92,52.83,46.96,44.42,
39.34,29.05,14.73 and 14.34.
25

WO 2008/023161

PCT/GB2007/003179

Procedure for the synthesis of {5-[2,4-bis-((S)-3-methyl-morphohn-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-pyridin-2-yI}-dimethyl-amine (Example ldd).


F N

To a solution of compound lah (1 equiv) in THF (0.05 M) was added a solution of 33 % dimethylamine in ethanol (200 equiv). The reaction vessel was sealed and exposed to microwave radiation (130°C, medium absorption setting) for 40 minutes. The solution was concentrated in vacuo to yield a yellow solid. The crude residue was then purified by 10 preparative HPLC to give the desired product. {5-[2,4-Bis-((S)-3-memyl-morpholin^-yl)^ dimethyl-amine: (54 % yield, 97 % purity) m/z (LC-MS,ESP):450 [M+H]+, R/T == 3.52 min)
Procedure for the synthesis of 8-[2,4-bis-((S)-3-methyI-morpholin-4-yI)-pyrido[2,3-15 d]pyrimidin-7-yl]-l,2,3,4-tetrahydro-benzo[e] [l,4Jdiazepin-5-one (Example lde).

The appropriate chloro-substrate (1 equiv), potassium carbonate (2.5 equiv), 3-methoxy-4-methoxycarbonylphenylboronic acid, pinacol ester (1.1 equiv) were suspended in (1:1) 20 acetonitrile/water (0.1 M of chloro-substrate). The mixture was sonicated and degassed for 15 minutes with nitrogen. Tetrakistriphenylphosphine (0.05 equiv) was then added and the mixture was sonicated for a further 5 minutes with nitrogen. The mixture was heated to 100°C for 3 hours under nitrogen. The reaction was cooled and the insoluble residue was filtered off.

WO 2008/023161

PCT/GB2007/M3179

The filtrate was concentrated to half the original volume and the remaining water mixture was extracted with CH2CI2. The organic layers were washed with water and brine, combined and dried with magnesium sulphate, filtered and concentrated in vacuo to yield an oil which was purified by flash column chromatography eluting with 50 % to 100 % EtOAc/Hexane. 5 4-[2J4-Bis-((S)-3-memyl-moipholm^-yl)-pyrido[2,3-d]pyrimidin-7-yl]-2-methoxy-benzoic acid methyl ester: (67 % yield, 100 % purity) m/z (LC-MSSESP):494 [M+H]+, R/T = 2.86 min)
A solution of 4-[2,4-bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2;3-d]pyrimidin-7-yi]-2-
10 methoxy-benzoic acid metliyl ester (1 equiv) in ethylenediamine (0.35 M) was stirred at room temperature for 24 hours. DMA was added to the solution (ethylenediamine/DMA 1:1.25). The reaction vessel was sealed and exposed to microwave radiation (180°C, medium absorption setting) for 1 hour. The reaction mixture was diluted with CH2CI2 and extracted with water and washed with brine. The organic layer was dried with magnesium sulphate,
15 filtered and concentrated in vacuo to yield a yellow solid which was then purified by preparative HPLC to give the desired product.
8-[2,4-Bis-((S)-3-methyl-morpholin-4-yl)-pyiido[23-d]pyrimidin-7-yl]-l!2,3,4-tetrahydro-benzo[e][l,4]diazepin-5-one: (49 % yield, 99 % purity) m/z (LC-MS,ESP):490 [M+H]4", R/T *= 3.52 min)
20
NMR data for Example 1 de
'H NMR (300 MHz, CDClj 5 ppm 8.65 (NH, s, or, 1H), 8.01 (ArH, &,J= 8.38 Hz, 1H), 7.78 (ArH, s, 1H), 7.68 (ArH, s, 1H), 7.44 (ArH, dd, J = 18.50, 8.20 Hz, 2H), 4.93-4.77 (CH2, m, 1H), 4.50 (CH2, s, 1H), 4.46-4.32 (CH2, m, 1H), 4.05-3.61 (CH2, m, 14H), 3.53 (CH2, d, 7 =
25 2.04 Hz, 1H), 3.41-3.26 (CH2) m, 1H), 1.47 (CH3, d, J = 6.76 Hz, 3H), 1.33 (CH3, d;./ = 6.78 Hz, 3H).
13CNMR(75MHz>CDCl38ppm 165.11,165.07,165.00,163.41,162.64,161.07, 159.90, 144.87,135.18,129.50,118.02,116.80,113.87,109.20,105.45,71.20, 70.89,67.14, 66.89, 52.77, 47.04, 44.76, 44.40, 39.33, 14.78 and 13.32.

WO 2008/023161

PCT/GB2007/003179

Procedure for the synthesis of 7-[2,4-bis-((S)-3-methyl-morphoIin-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-l,2^,4-tetrahydro-benzo[e][l,4]diazepin-5-one (Example ldf).

A solution of the compound Ibg (1 equiv) in ethylenediamine (0.35 M) was stirred at room temperature for 24 hours. DMA was added to the solution (ethylenediamine/DMA 1:1.25). The reaction vessel was sealed and exposed to microwave radiation (180°C, medium absorption setting) for 1 hour. The reaction mixture was diluted with ethyl acetate and
10 extracted with water. The organic layer was dried with magnesium sulphate, filtered and concentrated in vacuo to yield a residue which was then purified by flash column chromatography eluting with 0 % to 20 % MeOH/ CH2C12.
8-[2,4-Bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2J3-d]pyrimidin-7-yl]-lJ2;3,4-tetrahydro-benzo[e][l,4]diazepin-5-one: (40 % yield, 100 % purity) m/z (LC-MS,ESP):490 [M+H]+, R/T
15 =3.49min)
NMR data for Example ldf
lH NMR (300 MHz, CDCfe 5 ppm 8.60 (ArH, d, J = 2.23 Hz, IH), 7.93-7.83 (ArH, m, 2H), 7.34 (ArH, d, J = 8.56 Hz, IH), 6.89 (ArH, d, J= 8.97 Hz, IH), 4.82-4.71 (CH2, m, IH), 4.47 20 (CH2, dd, J = 7.28, 6.58 Hz, IH), 4.30 (CH2= d,J = 6.93 Hz, IH), 3.95-3.55 (CH2, m, 13H), 3.55-3.42 (CH2, m, IH), 3.35-3.21 (CH2, m, IH), 1.40 (CH3, d, J= 6.77 Hz, 3H), 1.26 (CH3, d,y = 6.80Hz,3H).


WO 2008/023161

PCT/GB2007/003179

Procedure for the synthesis of 5-[2,4-bis-((S)-3-methyl-morpholin-4-yI)-pyrido[2,3-d]pyrimidin-7-yl]-2-difluoromethoxy-benzamide (Example ldg).




5 To a solution of 5-bromo-2-difluoromethoxy-benzoic acid (1 equiv)inTHF (0.1 M) was added dropwise thionyl chloride (5 equiv) at room temperature. The reaction mixture was stirred at 40°C for 2 hours. The reaction mixture was concentrated in vacuo. The residue was suspended in dry THF (0.04 M) and ammonia gas was slowly bubbled into the reaction mixture for 45 minutes. The reaction mixture was concentrated in vacuo. The residue was 10 dissolved in minimum CH2CI2 and hexane was added to give a white precipitate that was collected by vacuum filtration in suitably clean form for use in subsequent reactions. 5-Bromo-2-difluoromethoxy-benzamide: (45 % yield, 73 % purity) m/z (LC-MS, ESP):266/268 [M+H]+, BJT = 3.42 min)
15 To a solution of 5-bromo-2-difluoromethoxy-benzamide (1 equiv) in dioxan (0.1 M) were added bis(pinacolato)diboron (l.l equiv), potassium acetate (3.5 equiv) and dppf (0.05 equiv). The reaction mixture was degassed with nitrogen for 15 minutes. PdCl2(dppf) (0.05 equiv) was added to the reaction mixture, which was degassed for a further 5 minutes. The reaction mixture was stirred at 110°C for 12 hours under nitrogen. The reaction mixture was
20 partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc and the combined organic phases were washed with water, dried with magnesium sulphate, filtered and concentrated in vacuo to give the desired product for use in subsequent reactions

WO 2008/023161

PCT/GB2007/003179

2-Difluoromethoxy-5-(434,5J5-tetramethyl-[ls3,2]dioxaborolan-2-yl)-benzamide: (71 % yield, crude taken forward without further analysis)
A mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (3.0 equiv), 2-5 difluoromethoxy-5-(4,435,5-tetramethyl-[l,3,2]dioxaborolan"2-yl)-benzamide (1.1 equiv) and tetrakis(triphenylphosphine) palladium0 (0.05 equiv) in acetonitrile/water (0.1 M of chloro-substrate) was stirred at 100 °C for 4 hours. Upon completion the reaction mixture was partitioned between water and CH2CI2 and extracted with CH2CI2. Combined organic phases were washed with brine, dried (MgS04), filtered and concentrated in vacuo. The crude residue 10 was purified by preparative HPLC to give the desired product.
5"[2,4-Bis-((S)-3-memyl-morpholin^-yl)-pyrido[23-d]pyrimidin-7-yl]-2-difluoromethoxy-benzamide: (14 % yield, 100 % purity) m/z (LC-MS, ESP): 515 [M+H]\ R/T - 7.40 min
Procedure for the synthesis of 5-[2,4-bis-((S)-3-methyl-morpholui-4-yl)-pyrido[2,3-15 d]pyrimidin-7-yl]-2-difluoromethoxy-N-methyl-benzamide (Example ldh).

o
To a solution of 5-bromo-2-difluoromethoxy-benzoic acid(l equiv) in DMF (0.1 M) was added triethylamine (4 equiv). The reaction mixture was cooled to 0°C and HBTU (1.2 equiv) was added. The reaction mixture was allowed to reach room temperature over 1 hour and 20 methylamine hydrochloride (2 equiv) was added. The reaction mixture was stirred at room temperature for 2 hours. The reaction mixture was partitioned between EtOAc and water and the aqueous phase was further extracted with EtOAc. The combined organic phases were

WO 2008/023161

PCT/GB2007/003I79

washed with water, dried with magnesium sulphate, filtered and concentrated under in vacuo to give the desired product in suitably clean form for use in subsequent reactions. 5-Bromo-2-difluoromemoxy-N-methyl-benzamide: (100 % yield, 75 % purity) m/z (LC-MS, ESP): 280/282 [M+H]+, R/T = 3.55 min) 5
2-Difiuoromethoxy-N-memyI-5-(4!4,5,5-tetramethyl-[13f2]dioxaborolan-2-yl)-beri2arnide was prepared in a similar way as 2-Difluoromethoxy-5-(4,4:5,5-tetramethyl-[l,3,2]dioxaboiolan-2-yl)-beiizarriide using 5-bromo-2-difluoromethoxy- N-methyl-benzamide as the starting material. 10 2-Difluoromethoxy-N-methyl-5-(4,4,53-tetramethyl-[l,3,2]dioxaborolan--2-yl)-ben2arnide: (100 % yield, crude taken forward without further analysis)
A mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (3.0 equiv), 2-difluoromethoxy-5-(4,4,5,5-tetramethyI-[l33,2]dioxaboroIan-2-yl)--benzamide (1.1 equiv) and
15 tetrakis(triphenylphosphine) palladium0 (0.05 equiv) in acetonitrile/water (0.1 M of chloro-substrate) was stirred at 100 °C for 2 hours. Upon completion the reaction mixture was partitioned between water and CH2G2 and extracted with CH2CI2. Combined organic phases were washed with brine, dried (MgSO4), filtered and concentrated in vacuo. The crude residue was purified by preparative HPLC to give the desired product.
20 5-[2)4-Bis~((S)-3-methyl-moipholm~4-yl)-pyrido[2,3-d]p>Timidm-7-yl]-2-dirluoromethoxy-N-methyl-benzamide: (53 % yield, 87 % purity) m/z (LC-MS, ESP): 421 [M+H]+, R/T = 4.06 min)

WO 2008/(123161

PCT/GB2007/003179


Procedure for the synthesis of 4-[2,4-bis-((S)-3-methyl-morpholin-4-yI)-pyrido[2,3-d]pyrimidin-7-yI]-2-methoxy-benzamide (Example ldi).
5 442,4-Bis-((S)-3-methyl-morpholin-4-yl)-pyrido[23-d]pyrimidin-7-yl]-2-methoxy-benzoic acid methyl ester (1 equiv) was dissolved in methanol (0.2 M). 1M Sodium hydroxide aqueous solution (5.0 equiv) was added. The reaction mixture was stirred at room temperature for 3 hours. Upon completion the reaction mixture was neutralised with 1M aqueous HC1 and concentrated in vacuo. The crude residue was purified by column chromatography on silica
10 gel eluting with 0 to 10 % MeOH in CH2C12 to give the desired product.
4_[2;4"Bis-((S)-3-methyl-morpholin^-yl)-pyrido[233-d]pyrmiidin-7-yl]-2-methoxy-henzoic acid: (100 % yield, 100 % purity) m/z (LC-MS, ESP): 480 [M+Hf, R/T = 2.69 min)
\5 4[2!4-Bis-((S)0-methyl-moiphoVm 4-y})
acid (1 equiv) was suspended in THF (0.05 M). Thionyl chloride was added dropwise at 40°C. The reaction mixture was then heated for an hour at 40°C. Ammonia gas was then slowly bubbled into the reaction mixture. THF was then added for further dilution (0.025 M) and the reaction mixture was heated for an hour at 40°C. Upon completion the reaction
20 mixture was cooled down and concentrated in vacuo. The residue was partitioned between water and CH2C12. The aqueous phase was extracted with CH2CI2. Combined organic phases were dried (MgSCU), filtered and concentrated in vacuo. The crude residue was purified by column chromatography on sijica gel eluting with 0 to 5 °/Q MeOH in CH2C12 to give the desired product

WO 2008/023161

PCT/GB2007/003179

4-[2,4-Bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2J3-d]pyrimidin-7-yl]-2-metrioxy-benzamide: (88 % yield, 99 % purity) m/z (LC-MS, ESP): 479 [M+H]+, R/T = 3.92 min)
5 NMR data for Example ldi
lH NMR (300 MHz, CDC13 6 ppm 8.30 (ArH, d, J = 8.17 Hz, IH), 8.04 (ArH, dd, J = 6.21, 4.98 Hz, 2H), 7.80 (NH: br, s, IH), 7.67 (ArH, dd, J= 8.21,1.49 Hz, IH), 7.49 (ArH, d, J = 8.44 Hz, IH), 5.96 (NH, s, br, IH), 4.98-4.85 (CH2, m, IH), 4.61 (CH2, d, J~ 12.90 Hz, IH), 4.39 (CH2, d= J = 6.89 Hz, IH), 4.13 (OCH3; s, 3H), 4.05-3.64 (CH2, m, 9H), 3.64-3.51 (CH2, 10 m, IH), 3.41 (CH2, dd, J= 13.34, 3.62 Hz, IH), 1.49 (CH3) d, J = 6.79 Hz, 3H), 1.36 (CH3, d, ./=6.82Hz,3H).
,3CNMR(75 MHz, CDClj) 5 ppm 166.78,165.32,162.81, 160.99, 160.02, 158.15,143.57, 134,98, 132.76,121.80, 120.15,113.62,111.30,105.44, 71.27,70.89, 67.23, 66.90, 56.42, 52.88,47.01, 44.41, 39.36, 14.77 and 14.40.
Procedure for the synthesis of 4-[2,4-bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-2-methoxy-N-methyI-benzamide (Example ldj).

4-[2,4-Bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-2-methoxy-benzoic 20 acid (1 equiv) was dissolved in THF (0. 1 M) and HBTU (1.5 equiv) was added. Methylamine in THF (15 equiv) was added dropwise followed by triethylamine (1.5 equiv) and the reaction mixture was stirred at room temperature for 2 hours. The reaction mixture was concentrated in vacuo. The residue was partitioned between water and CH2CI2. The aqueous phase was extracted with CH2CI2. Combined organic phases were dried (MgS04), 25 filtered and concentrated in vacuo. The crude residue was purified by preparative HPLC to give the desired product.

WO 2008/023161 PCT/GB2007/003179
4-[2J4-Bis-((S)-3-methyl-moipholin-4-yl)-pyrido[233~d3pyriiTudin-7-yl]-2-methoxy-N-methyl-benzamide: (56 % yield, 96 % purity) m/z (LC-MS, ESP): 493 [M+HJ+, R/T = 4.00 min)
NMR data for Example Idi 5 !H NMR (300 MHz, CDClj 5 ppm 8.26 (ArH, d, J = 8.16 Hz, 1H), 7.98 (ArH, dd, ./= 8.74, 4.91 Hz, 2H), 7.91-7.81 (NH, ra, br, 1H), 7.60 (ArH, dd,J = 8.21,1.52 Hz, 1H), 7.43 (ArH, d, J - 8.45 Hz, 1H), 4.93-4.81 (CH2, m, 1H), 4.62-4.51 (CH2> m, 1H), 4.39-4.28 (CH2, m, 1H), 4.07 (OCH3, s, 3H), 4.00-3.58 (CH2, m, 9H), 3.57-3.45 (CH2, m, 1H), 3.40-3.27 (CH2, ms 1H), 2.99 (NHCH3) d, J = 4.82 Hz, 3H), 1.43 (CH3, d, J => 6.78 Hz, 3H), 1.31 (CH3, d, J = 10 6.81 Hz, 3H).
Procedure for the synthesis of 2-methoxy-N-methyl-5-[4-((S)-3-methyl-morpholin-4-yl)-2-morpholin-4-yl-pyrido[2,3-d]pyrimidin-7-yl]-benzamide (Example ldk).

15 To a mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (3.0 equiv), and 2-methoxy-5-(4,4J5,5-tetramethyl-[l,3,2]dioxaborolan-2'yl)-benzoic acid methyl ester (1.05 equiv) in acetonitrile/water (1:1) (0.028 M of chloro-substrate) was added tetrakis(triphenylphosphine) palladium0 (0.05 equiv). The reaction vessel was sealed and exposed to microwave radiation (130°C, medium absorption setting) for 20 minutes. Upon
20 completion the reaction mixture was partitioned between water and CH2CI2 and extracted with CH2CI2. Combined organic phases were dried (MgSO4, filtered and concentrated in

WO 2008/023161

PCT/CB2007/003179

vacuo. The crude residue was purified by column chromatography on silica gel eluting with 0 to 20 % MeOH in CH2C12 to give the desired product.
2-Methoxy~5-[4-((S)-3-methyl-morpholin-4-yl)-2-morpholin-4-yl-pyrido[2,3-d]pyrimidin-7-yl]-benzoic acid: (91 % yield, 100 % purity) m/z (LC-MS, ESP): 466.4 [M+H]4", R/T = 2.68 5 min)
2-Methoxy-5-[4-((S)-3-methyI~morpholin-4-yl)-2-moiphoIin-4-yl-pyrido[2,3-d]pyrimidin-7-yl]-benzoic acid (1 equiv) was dissolved in DMF (0.1 M) and DIPEA (8 equiv) was added. HBTU (1.2 equiv) was added at 0°C and the reaction mixture was stirred for 30 minutes. Methylamine hydrochloride (5 equiv) was added and the reaction mixture was stirred 0°C for
10 30 minutes and at room temperature for 1 hour. The reaction mixture was partitioned between water and ethyl acetate. The aqueous phase was extracted with ethyl acetate. Combined organic phases were washed with water and brine, dried (MgS04), filtered and concentrated in vacuo. The crude residue was purified by column chromatography to give the desired product. 4-[2,4-Bis-((S)-3-memyl-moipholm-4-yl>pyrido[2,3:d]pyrimidin-7-yl]-2-methoxy-N~methyl-
15 benzamide: (73 % yield, 97 % purity) m/z (LC-MS, ESP): 479.2 [M+H]+, R/T = 3.97 min)
NMR data for Example Idk
'HNMR(300 MHz, CDCb 5ppm 8.71 (ArH, d, J = 2.45 Hz, IH), 8.45 (ArH, dd, J = 8.75,
2.48 Hz, IH), 7.97 (ArH, d, J = 8.52 Hz, 1H)S 7.78 (NH, s, br, IH), 7.51 (ArH, d, J = 8.56 20 Hz, IH), 7.01 (ArH, d, J = 8.84 Hz, IH), 4.39 (CH2, d, J= 6.69 Hz, IH), 3.96 (OCH3, s, 3H),
3.95-3.77 (CH2, m, 7H), 3.76-3.58 (CH2, m, IK), 2.98 (NCH3, d, J= 4.81 Hz, 3H), 1.42
(CH3,d,./=6.78Hz,3H).
I3CNMR(75 MHz, CDCb)5ppm 165.74, 164.71, 161.22, 160.99, 159.11, 159.04, 135.09,
132.93, 131.23, 131.16, 121.32, 119.02, 113.63, 111.84, 104.61,70.90,66.90,56.27,52.70, 25 44.70, 44.48, 26.70 and 14.85.

WO 2008/023161

PCT/GB2007/003179

Procedure for the synthesis of 6-[2,4-bis-((S)-3-methyl-morpholin-4-yI)-pyrido[2,3-d]pyrimidin-7-yI]-lH-indazoI-3-yIaniine (Example ldl).

5 To a mixture of 7-chloro-4-((S)-3-methyl-morpholin-4-yl)-2-C(S)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidine (1 equiv), potassium carbonate (2.5 equiv), and 4-cyano-3-fluorophenylboronic acid (1,2 equiv) in acetonitrile/water (1:1) (0.03 M of chloro-substrate) was added tetrakis(triphenylphosphine) palladium0 (0.05 equiv). The reaction vessel was sealed and exposed to microwave radiation (110 °C, medium absorption setting) for 25 10 minutes under nitrogen atmosphere. Upon .completion the precipitate was collected by vacuum filtration, which was in suitably pure form to be used with no further purification.

4-[2,4-Bis-((S)-3-memyl-morphoun-4-yl)-pyrido[2,3-d]pyrimidiri-7-yl]-2-iluoro-benzonitrile: (49 % yield, 96 % purity) m/z (LC-MS, ESP): 449.2 [M+Hf R/T = 2.93 min
15
To a 0.2 M solution of 4-[2}4-bis-((S)-3-methyI-morpholin-4-yl)-pyrido[2t3-d]pyrimidin-7-yi]~2-fluoro-benzonitrile (1 equiv) in n-BuOH was added 0.2 reaction volumes of hydrazine hydrate. A reflux condenser was attached to the mixture which was then heated to 140 °C for 2 hours whereupon it was cooled, and concentrated in vacuo to give an orange residue which
20 was purified by flash chromatography (Si02) using Et20:MeOH - 94:6 as eluent which
allowed a yellow solid which was then recrystallised from CH2CL2exanes to furnish the title compound as a yellow solid,

WO 2008/023161

PCT/GB2007/003179


6-[234-Bis-((S)-3-methyl-morpholin-4-yl)-pyrido[253-d]pyrim^din-7-yl]-lH-indazol-3-ylamine: (90 % yield, 97 % purity) m/z (LC-MS, ESP): 461.2 [M+Hf R/T = 3.77 min NMR data for Example ldl 5 ('HNMR (300MHz, CD3SOCD3 8ppm 11.6 (IH, s, formate), 8.31-8.01 (ArH. m, 2H), 7.74 (ArH, ddd, J= 18.90, 15.23, 8.49 Hz, 3H), 5.42 (NH2, s, 2H), 4.88-4.70 (NH, m, IH), 4.44
(CH2, d, J = 10.93 Hz, 2H), 3.91 (CH2, m, 3H), 3.81-3.54 (Ctfi, m, 6H), 3.46 (CH2, dt, J =
11.82, 11.67, 2.52 Hz, IH), 3.38-3.13 (CH2, m, IH), 2.51 (CH, td, J = 3.52, 1.73,1.73 Hz,
IH), 1.38 (CH3, d, J = 6.75 Hz, 3H), 1.26 (CH3, d, J = 6.79 Hz, 3H). 10 13CNMR(75 MHz, CD3SOCD3) 5 ppm 164.43,162.05,161-20,159.29,149.19,141.79,
136.12,135.32,120.42,116.81,114.78,113.13,108.47,104.30, 70.39, 70.15, 66.34, 66.15,
51.81, 46.24,43.81, 30.89, 22.0,14.31 and 13.89.
Procedure for the synthesis of N-{4-[2,4-bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-15 d]pyrimidin-7-yl]-pyridin-2-yl}-acetamide (Example 1dm)-

To a 0.1 M solution of 4-[2,4-bis-((S)-3-methyl-morpholin-4'yl)-pyrido[2,3-d]pyrimidin-7-yl]-pyridin-2-ylamine (example lcz) (1 equiv) in pyridine was added acetic anhydride (3 equiv). A reflux condenser was attached to the reaction vessel which was then heated to 70 °C

WO 2008/023161

PCT/GB2007/003179

for 2 days. Upon completion, the reaction was purified, in its crude for by preparative HPLC to give the title compound as a white solid.
N44-[2,4-Bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-pyridin-2-yl}-acetamide :(95 % yield, 99 % purity) m/z (LC-MS, ESP): 464.1 [M+H]+ R/T = 3.77 min 5 NMR data for Example 1 dm
'H NMR (300 MHz, CDC13.5 ppm 8.70 (NH, s, IH), 8.37-8.29 (ArH, m, 2H), 8.01 (ArH, d, J = 8.36 Hz, IH), 7.94 (ArH, dd, J = 5.26,1.54 Hz, IH), 7.49 (ArH, d, J = 8.39 Hz, IH), 4.87 (CH2, ddd, J= 2.90,1.56, 0.64 Hz, IH), 4.56 (CH2, d, J = 13.43 Hz, IH), 4.33 (CH2, d, / = 6.86 Hz, IH), 3.99-3.58 (CH2) m, 10H), 3.57-3.45 (CH2, m, IH), 3.39-3.25 (CH2, m, IH), 10 2.19 (CH3, s, 3H), 1.43 (CH3j d, J = 6.78 Hz, 3H), 1.31 (CH3) d, J= 6.82 Hz, 3H)
,3CNMR(75 MHz,CDClj 8 ppm 168.79,165.32,162.82,160.00,159.87, 151.94, 148.64, 148.15,135.18,118.86,113.66,111.94,106.03,71.27,70.89,67.23,66.89,52.89,46.98, 44.46, 39.35,24.81, 14.77 and 14.41.
15 Procedure for the synthesis of Examples ldn to Idp

The appropriate 7-chloropyridopyrimidine was reacted with 2-methoxy-5-(4,4,5,5-
tetramethyl-[l,3,2]dioxaborolan-2-yl)-benzoic acid methyl ester according to conditions E to 20 give2-methoxy-5-[4-((S)-3-methyl-morpholin-4-yI)-2-thiomorpholin"4-yl-pyrido[2,3-
d]pyrimidin-7-yl]-benzoic acid methyl ester as the desired product (1 equiv) which was then diluted in MeOH to give a 0.03M solution. NaOH (5 equiv of 1 M solution) was then added and the resultant mixture stirred at room temperature for 5 days. After this time the reaction was filtered and neutralized with IM HCl before being concentrated in vacuo to give a crude

WO 2008/023161

PCT/GB2007/003179

yellow residue which was diluted in CH2C12. The mixture was filtered and the resulting filtrate concentrated to give the desired product as an oil.

2-Methoxy-544-((S)-3-methyl-morpholin-4-yl)-2-tMornorpholin-4-yl-pyrido[2,3-d]pyrirrudin-7-yl]-benzoic acid: (99 % yield, 95 % purity) m/z (LC-MS, ESP): 482.2[M+H]+ R/T = 2.78 min

2-Methoxy-5-[4-((S)-3-methyl-morphoan^-yl)-2-(4-methyl-r?iperazin-l-yl)"pyrido[233-d]pyrimidin-7-yl]-benzoic acid: (88 % yield, 96 % purity) m/2 (LC-MS, ESP): 479.5[M+H]" R/T = 2.26 min

WO 2008/023161

PCT/GB200 7/003179


2-Memoxy-5~[4-((S)-3-methyl-morphoHn^-yl)-2-moi^holin-4-yl-pyrido[23-d]pyrirnidin-7--yl]-benzoic acid: (91 % yield, 100 % purity) m/z (LC-MS, ESP):466.4 [M+H]+ R/T = 2.68
5 min
To a wanned (40 °C) 0.06 M solution of the appropriate benzoic acid derivative(l equiv) in anhydrous THF was added thionyl chloride (2.5 equiv) in a dropwise fashion. The reaction was maintained at this temperature and stirred for a further I hour. After this time the mixture
10 was evaporated to give a brown oil, which was diluted in dry THF (sufficient to make 0.06 M solution) before ammonia gas was bubble through the mixture, which was accompanied by an exotherm. Upon completion, addition of ammonia was stopped and the mixture concentrated in vacuo to give a a yellow oily residue which was dissolved in CH2CI2 (1 reaction volume) and washed with water (2X1 reaction volume). The organic extract was removed, dried
15 (MgSOit), filtered and concentrated in vacuo to give the title compound.


WO 2008/023161

PCT/GB2007/003179

2-Methoxy-5-[4-((S)-3-methyl-morpholm-4-yl)-2-thiomorpholin-4-yl-pyrido[2,3-d]pyrimidin-7-yl]-benzaraide: (30 % yield, 97 % purity) m/z (LC-MS, ESP): 481.1[M+H]+ R/T = 4.02 min
5 NMR data for Example ldn
'H NMR (300 MHz, CDCIj 5 ppm 8.83 (ArH, d, J= 2.46 Hz, IH), 8.61 (ArH, dd, J = 8.75, 2.48 Hz, IH), 8.00 (ArH, d, J = 8.47 Hz, IH), 7.72 (NH, d, J = 0.76 Hz, IH), 7.56 (ArH, d,./ = 8.50 Hz, IH), 7.13 (ArH, d, J = 8.82 Hz, IH), 5.88 (NH, d,./= 0.98 Hz, IH), 4.42-4.23 (CH2, m,4), 4.05 (CH30, s, 3H), 4.03-3.94 (CH2) m, IH), 3.85 (CH2, ddd, J = 14.51, 8.58, 10 5.82 Hz, 2H), 3.78-3.62 (CH2, m, 3H), 2.75-2.65 (CH2, m, 3H), 1.46 (CH3, d, J= 6.16 Hz, 3H).
13CNMR(75MHz,CDCb 5ppm 166.60,165.41, 162.87, 161.09, 159.89, 159.23, 134.73, 133.71, 131.82,131.68,120.56, 113.16, 111.89,104.63, 70.95, 66.91, 56.29, 52.81,46.70, 44.54, 27.45 and 14.70.

2-Methoxy-5-[4-((S)-3-methyl-morpholin-4-yl)-2-(4-methyI-piperazin-l-yl)-pyrido[2;3-20 d]pyrimidm-7-yl]-benzamide: (12 % yield, 98 % purity) m/z (LC-MS, ESP): 481.1 [M+H]+
R/T = 43.28 min
NMR data for Example ldo
'H NMR(300 MHz, CDCI3 8 ppm 8.78 (ArH, d,J= 2.48 Hz, IH), 8.57 (ArH, dd, J = 8.76,
2.52 Hz, IH), 8.28 (NH, s, br, IH), 7.96 (ArH, d, J = 8.50 Hz, IH), 7.68 (NH, s, br, IH), 7.54 25 (ArH, d, J= 8.55 Hz, IH), 7.08 (ArH, dyJ= 8.84 Hz, IH), 4.42-4.28 (CH2, m, IH), 4.09

WO 2008/023161

PCT/GB2007/003179

(CH2, s, br, 2H), 4.01 (OCH3, s, 3H), 3.77 (CH2, ddd, J - 36.04,19.80,10.87 Hz, 9H), 2.76 (CH2, t, J = 5.05, 5.05 Hz, 4H), 2.47 (NCH3, s, 3H), 1.42 (CH3, d, J = 6.77 Hz, 3H).

2-Methoxy-5-[4-((S)-3-methyl-morpholin-4-yl)-2-morpholin-4-yl-pyrido[2,3-d]pyrimidin-7-yl]-benzamide: (61 % yield, 97 % purity) m/z (LC-MS, ESP): 465.4 [M+H]+ R/T = 2.69 min (NMR data for Example ldp
10 lH NMR (300 MHz,, CDClj 8 ppm 8.77 (ArH, d, J = 2.44 Hz, 1H), 8.58 (ArH, dd, J = 8.76, 2.47 Hz, 1H), 7.94 (ArH, d, J = 8.48 Hz, 1H), 7.65 (NH, s, br, 1H), 7.51 (ArH, d, J = 8.53 Hz, 1H), 7.06 (ArH, d, J - 8.84 Hz, 1H), 5.91 (NH, s, br, 1H), 4.32 (CH2, d, 7 = 6.79 Hz, 1H), 3.98 (OCH3, s, 3H), 3.95-3.86 (CH& m, 5H), 3.84-3.55 (CH2, m, 9H), 1.40 (CHj, d, J = 6.77 Hz, 3H)
15 I3CNMR(75 MHz, CDCI3 6 ppm 166.63, 165.31,162.78, 160.96, 160.31, 159,29, 134.76, 133.68, 131.69,131.60,120.56,113.09,111.88,104.76,70.94, 67.04,66.91, 56.28, 52.76, 44.58, 44.45 and 14.75.
Procedure for the synthesis of Example ldq


WO 2008/023161

PCT/GB2007/IW3179

To a (0.1 M) solution of example lat (1 equiv) in CHCI3 was added m-CPBA (5.5 equiv). A reflux condenser was added to the apparatus and the mixture heated to 60 °C for 17 hours. After this time the reaction was concentrated in vacuo and purified by flash chromatography (SIO2) using CH2Cl2:MeOH - 95:5 as eluent to furnish the desired product.

N-{3-[2J4-Bis-((S)^-methyl-moipholin4-yl)-8-oxy-pyrido[2,3-d]pyrimidin-7-yl]-phenyl}-methanesulfonamide: (39 % yield, 100 % purity) m/z (LC-MS, ESP): 515.5[M+H]+ R7T = 2.95min.
10
NMR data for Example ldq
lHNMR(300 MHz, CDCb Sppm 10.04 (NH, s, br, 1H), 8.42 (ArH, s, 1H)5 7.55-7.25 (ArH,
m, 4H), 6.96 (ArH, d, J = 8.67 Hz, 1H), 4.80 (CH2, s, br, 1H), 4.51 (CH2) s, br, 1H), 4.31 15 (CH2, d, J = 6,71 Hz, 1H), 4.00-3.51 (CH2, m, 9H), 3.49-3.34 (CH2, m, IK), 3.24 (CH2, dd, J
= 13.22, 3.30 Hz, 1H), 2.80 (SCH3, s, 3H), 1.42 (CH3, &,J= 6.78 Hz, 1H), 1.19 (CH3, d, J =
6.69 Hz, 3H).
13CNMR(75 MHz,CDCl3 5ppm 165.58,159.69, 158.28,149.96, 138.59, 134.47, 129.53,
125.86,123.35, 123.30, 116.17, 107.52,71.33,71.11,67.32,67.10,53.39,47.62,44.87, 20 39.79, 38.68, 31.90, 22.97 and 15.16.

WO 2008/023161

PCT/GB2007/003179

Procedure for the synthesis of Example ldr



7-CliIoro-2,4-bis-((S)-3-methyl-morphoUn-4-yl)-pyrido[2)3-d]pyrimidine was couple with 3-nitrobenzoic acid usine Suzuki conditions D to eive the desire product as a yellow Dowder.

2,4-Bis-((S)-3-methyl-morpholin-4-yl)-7-(3-nitro-phenyl)-pyrido[2,3-d]pyrimidine:(90% yield, 100 % purity) m/z (LC-MS, ESP): 451.6[M+H]+ R/T = 3.41min
To a 0.1M solution of 2,4-bis-((S)-3-metIiyl-morpholin-4-yl)-7-(3-nitro-phenyl)-pyrido[2.3-10 d]pyrimidine (1 equiv) in EtOH/tfeO-l:! was added ammonium chloride (8 equiv) and iron powder (8 equiv). The reaction mixture was heated to 100 °C for 1 hour before cooling and filtering through a thin CeHte ™ pad. The cake was washed with EtOH (1 reaction volume). The filtrate was concentrated in vacuo and then partitioned between water and CH2Q2 0 reaction volume of each). The organic phase was removed, dried (MgSO*) , filtered and 15 concentrated in vacuo and then purified by flash chromatography (SiO2) using MeOH:CH2Cl2 (0:100 - 5:95 - 10-90) as eluent to give the title compound as a yellow solid.



WO 2008/023161

PCT/GB2007/003179

3-[2,4-Bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-phenylamine: (88% yield, 98 % purity) nVz (LC-MS, ESP): 421.1 [M+H]+ R/T = 3.76min
NMR data for Example ldr 5 'HNMR (300 MHz, CDClj 5ppm 7.93 (AiH, d,/= 8.45 Hz, IH), 7.62-7.55 (ArH, m, IH), 7.41-7.32 (m, IH), 7-20 (ArH, d, J= 7.32 Hz, 2H), 6.71 (ArH, ddd, J = 7.88,2.40, 0.86 Hz, IH), 4.87 (ArH, dd, / = 3.54, 1.66 Hz, IH), 4.57 (NH, d, J = 13.25 Hz, IH), 4.30 (NH, s, br, IH), 3.98-3.56 (CH2, m, 11H), 3.56-3.44 (CH2, m, IH), 3.37-3.24 (CH2, m, IH), 1.40 (CH3, dt J= 6.77 Hz, 3H), 1.29 (CH3, &,J= 6.81 Hz, 3H) 10 [3CNMR(75 MHz, CDCb 5ppm 165.46,162.87, 162.60,159.96, 146.80, 139.75, 134.48, 129.35,117.99, 116.69,114.74, 113.48, 104.92, 71.32, 70.93, 67.28, 66.94, 52.80,46.90, 44.49,39.33,14.71 and 14.33.
Procedure for the synthesis of Example Ids

To a0.3M solution of 5-[2,4-bis-((S)-3-methyI-morpholin-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-2-fluoro-benzonitrile (example lav) (1 equiv) in EtOH was added hydrazine hydrate (5
20 equiv). The mixture was refluxed tor 90 minutes wherepon it was cooled and partitioned between CH2CI2 and water (1 reaction volume of each). The organic extract was removed. The aqueous phase was further extracted with CH2CI2 (2 X 1 reaction volume). The combined organic extracts were then dried (MgSCU), filtered and concentrated in vacuo to give a yellow slurry which was further purified by flash chromatography (SiO2) using EtOAC/Hexanes as
25 eluent to give the title compound as a yellow powder.

WO 2008/023161

PCT/GB2007/003179


5-[2,4-Bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidin-7--yl]-lH-indazol-3-5 ylamine: (52% yield, 100 % purity)m/z(LC-MS, ESP): 461.6[M+H]+R/T = 2.85min
NMR data for Example Ids
10 'HNMR(300 MHz, CDClj 5ppm 8.52 (ArH, s, IH), 8.06 (ArH, dd, J = 8.84,1.50Hz, IH), 7.94 (ArH, d: J = 8.49 Hz, IH), 7.42 (ArH, d, J = 8.50 Hz, IH), 7.29 (ArH, d, J = 8.79 Hz, 2H), 4.87 (CH2, dd, J= 3.99,1.99 Hz, IH), 4.60 (CH2, s, br, IH), 4.32 (CH2, d, J= 6.78 Hz, IH), 3.98-3.58 (CH2) m, 9H), 3.51 (CH2, dt,J = 11.78,11.46,2.71 Hz, IH), 3.39-3.25 (CH2, m, 1H), 1.42 (CH3, d, J = 6.77 Hz, 3H), 1.29 (CH3, d, J = 6.81 Hz, 3H) (NH's not clearly
15 seen)
l3C NMR (75 MHz, CDClj 5 ppm 165.43, 162.97,162.33, 160.01, 142.86, 134.62,130.11, 127.06,120.17,115.21,112.98, 109.71,104.51, 71.32,70.94, 67.28, 66.95, 52.80,46.95, 44.48, 39.36, 27.01, 14.79 and 14.33.

WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179


Note:
The following examples were synthesized from the corresponding boronic acids: ldu, ldv, ldzand lee.
The following examples were synthesized from the corresponding pinacolate boron esters: 5 ldw, ldx. lea, leb and lee.
The following Examples were synthesized from a mixture of the corresponding boronic acids and pinacolate boron esters: ldt, ldy, and led.
NMR data for Example lee 10 'H NMR (300 MHz, CDClj 5 ppm 8.15 (ArH, d, J= 7.5 Hz, 2H), 7.97 (ArH, d, 7- 8.46 Hz,
IH), 7.42 (ArH, d, 7= 8.46 Hz, IH), 6.98 (ArH, d,7- 9.24 Hz, IH), 4.91 (CH2, d, 7= 5.55
Hz, IH), 4.77 (CH2OH, s, 2H), 4.61 (CH2) d, 7= 12.42 Hz, IH), 4.36 - 4.34 (CH2, m, IH),
4.00- 3.70 (OCH3 + CH2, m, 9H), 3.69-3.51 (CH2, m, IH), 3.41 -3.31 (CH2, m, IH), 1.46
(CH3, d, 7= 6.69 Hz, 3H), 1.35 (CH3, d, 7= 6.87 Hz, 3H). 15 13C NMR (75 MHz, CDClj) 8 ppm 165.42,162.88,161.87,159.95, 159.16, 134.54, 131.13,
129.25,128.89,128.44,112.85,110.27,104.49, 7130, 70.92, 67.26, 66.93, 61.98, 55.56,
52.78, 46.91,44.45, 39.32,14.69 and 14.31.
NMR data for Example led
20 ]H NMR (300 MHz, CDClj 8 ppm 8.34 (ArH, s, IH), 8.11 (ArH, d,J= 8.02 Hz, IH), 8.00 (ArH, d, 7 = 8.41 Hz, IH), 7.90 (ArH, d, J = 7.98 Hz, IH), 7.43 (ArH, d, 7 = 8.42 Hz, IH), 7.10 (NH, br, s, IH), 4.95-4.81 (CH2, m, IH), 4.57 (CH2, d, 7 = 13.37 Hz, IH), 4.47 (NHCH2, s, 2H), 4.33 (CH2, d, 7 = 6.68 Hz, IH), 3.99-3.58 (CH2, m, 9H), 3.51 (CH2, dt, J= 11.81, 11.45,2.72 Hz, 1H),3.31 (CH2,dt,7 = 12.91, 12.52, 3.57 Hz, IH), 1.42 (CH3, d, 7 = 6.78 Hz,
25 3H), 1.30 (CH3, d, J=6.81 Hz, 3H).

WO 2008/023161

PCT/GB2D07/003179

13CNMR(75 MHZ,CDC1J) 5ppm 171.32,165.36, 162.94,161.42,160.04, 144.06, 142.31, 135.01,133.20,127.63, 123.83,123.08,113.49,105.35,71.27, 70.91, 67.24, 66.91, 52.85, 46.96,45.70,44.48, 39,35,14.76 and 14.39.
5 NMR data for Example 1 ef
'HNMR (300 MHz, CDC13.8 ppm 8.08 (ArH, d, J= 1.95 Hz, 1H), 8.01-7.94 (ArH, m, 1H), 7.82 (ArH, td,./ - 6.63,1.80, 1.80 Hz, 1H), 7.48 (NH, br, s, 1H), 7.39 (ArH, dd,./ = 12.99, 5.20 Hz, 3H), 4.34 (CH2, q, J= 6.63,6.56, 6.56 Hz, 1H), 3.97-3.76 (CH2, m, 7H), 3.75-3.57 (CH2, m, 7H), 2.87 (S02CH3, s, 3H), 1.42 (CH3, d, J = 6.78 Hz, 3H). 10 13CNMR(75 MHz, CDCI3) §ppm 165.21,162.77,161.34,160.28,140.30,137.69,135.07, 129.91,124.53,122.37,120.57,113.44,105.22, 70.91, 66.97, 66.89, 52.84,44.58,44.39, 39.32 and 14.79.
NMR data for Example ldz
15 'HNMR(300 MHz, CDClj 8ppm 8.11-8.03 (ArH,m, 2H), 7.96 (ArH, d, J= 8.44 Hz, IH), 73S-7.3L (ArH, m, f H), 7.32-7.24 (ArH, m, 2H), 4.85 (CH2, d,J= 5.45 Hz, 1H), 4.54 (CH2, d, J= 12.83 Hz, 1H), 4.32 (CH2, d, J= 6.78 Hz, 1H), 3.97-3.57 (CH2, tn, 9H), 3.50 (CH2, dt, J = 11.75, 11.35, 2.73 Hz, 1H), 3.37-3.24 (CH2, m, 1H), 2.95 (S02CH3, s, 3H), 1.42 (CH3, d, J = 6.78 Hz, 1H), 1.29 (CHZ, d,./ = 6.81 Hz, 3H) (NH not seen).
20 13CNMR(75 MHz, CDCySppm 165.36,162.93,161.33,160.00,138.73, 135.29,134.86,
129.34, 119.66,112.95, 104.90,71.27,70.92,67.24,66.93,52.82,46.97,44.45,39.58,33.35, 14.75 and 14.36.
NMR data for Example lea
25 lHNMR(300 MHz, CDC13.5 ppm 7.87 (ArH, d, J = 8.55 Hz, IH), 7.81 (ArH, d, J = 1.80 Hz, 1H), 7.47 (ArH, dd, J = 8.17,1.85 Hz, 1H), 7.35 (ArH, d, J = 8.57 Hz, 1H), 6.69 (ArH, d,./ = 8.14 Hz, 1H), 4.85 (CH2, d, J= 5.96 Hz, IH), 4.62-4.52 (CH2, m, 1H), 4.28 (CH2, d, J = 6.77 Hz, IH), 4.02 (NH2, s, br, 2H), 3.95 (d, J = 6.54 Hz, IH), 3.93 (CH3, s, 3H), 3.92-3.57 (CH2, in, 9H), 3.55-3.45 (CH2,m, IH), 3.38-3.25 (CH2, m, IH), 1.39 (CH3) d,y = 6.77Hz! 3H),
30 1.29 (CH3,d,/= 6.81 Hz, 3H). '

WO 2008/023161

PCT/GB2007/003179

13CNMR(75 MHz, CDCU) 5 ppm 165.48,162.91,162.45,159.98, 147.22,138.70,134.14, 128.92, 121.24, 113.97, 112.74, 110.15, 104.11,71.35,70.95,67.32,66.96,55.83,52.79, 46.89,44.44, 39.31,31.60,22.66 and 14.30.
5 NMR data for Example leb
'H NMR (300 MHz, CD3COCD3 5 ppm 11.83 (ArH, s, IH), 9.06 (ArH, d, J - 2.07 Hz, IH), 8.75 (ArH,d,/= 2.09 Hz, IH), 8.30-8.10 (ArH, m, IH), 7.72 (ArH,d, J= 8.55 Hz, IH), 7.54 (ArH,s, IH), 6.59 (NH. s, IH), 4.77 (CH2, dd, .7-6.66,1.89Hz, IH), 4.49-4.34 (CH3, m, 2H), 4.03-3.83 (CH2, m, 3H), 3.81-3.55 (CH2, m, 6H), 3.54-3.38 (CH2, m, IH), 3.23 (CH2, dd, 10 J = 13.19,3.46 Hz, IH), 1.37 (CH3, d, J = 6.74 Hz, 3H), 1.25 (CH3, d, .7 = 6.75 Hz, 3H). UCNMR(75 MHz, CD3COCD3) 5ppm 165.41,163.09, 161.32,160.22,150.21, 143.45, 136.25, 128.14,128.09, 126.99, 120.44,113.54, 104.90, 101.82, 71.32, 71.09, 67.27, 67.09, 52.78,47.17, 44.79 and 15.25.
15 NMR data for Example Idv
IH NMR (300 MHz, CD3SOCDj 5 ppm 8.40 (ArH, d, J= 1.37 Hz, IH), 8.33 (ArH, dd, J = 8.38, 1.63 Hz, IH), 8.25 (ArH, d,J= 8.45 Hz, 2H), 8.17 (ArH, s, IH), 7.79 (ArH, d,/= 8.48 Hz, IH), 4.84-4.73 (CH, m, IH), 4.45 (CH2, d, J = 13.67 Hz, 2H), 4.00-3.84 (CH2, m, 3H), 3.81-3.57 (CH2, m, 6H), 3.46 (CH2, dt, J= 11.84,11.73,2.61 Hz, IH), 3.23 (CH2, dt,J =
20 13.16,12.92, 3.65 Hz, IH), 1.39 (CH3, d, J= 6.75 Hz, 3H), 1.26 (CH3, d, J = 6.75 Hz, 3H) 13CNMR(75 MHz, CD3SOCD3 6 ppm 164.30,162.06,160.45,159.32, 159.03,149.10, 145.91,143.53,135.86,126.40, 125.73,125.30, 123.33, 113.30, 105.06, 70.35, 70.14, 66.31, 66.14, 51.79,46.27, 43.81, 30.38,14.35 and 13.89.
NMR data for Example ldv
'HNMR (300 MHz, CD3SOCD3 5ppm 8.39 (ArH, dd, J = 5.45, 3.65 Hz, IH), 8.23 (ArH, d, J = 8.47 Hz, IH), 8.14-8.03 (ArH, m, 2H), 7.81-7.69 (ArH + NH, m, 2H), 4.77 (CH2, dd, J = 6.52, 2.00 Hz, IH), 4.43 (CH2, d, J= 13.75 Hz, 2H), 3.99-3.83 (CH2, m, 2H), 3.80-3.56 (CH2, 30 m, 6H), 3.52-3.15 (CH2j m, 5H), 2.50 (CH2, td, J= 3.67, 1.83,1.83 Hz, 2H), 1.38 (CH3, d, J = 6.75 Hz, 3H), 1.25(CH3,d, .7=6.75 Hz, 3H), 1.19-1.10 (CH3, m, 3H).

WO 2008/023161

PCT/GB2007/003179

l3CNMR (75 MHz, CD3SOCD38ppm 164.80, 163.51,162.52,159.84,158.73,158.27, 142.53, 142.43, 136.38, 131.02,125.84,123.52,123.48,115.08, 114.76,113.45,105.57, 70.87, 70.64,66.83, 66.65, 52-32,46.79,44.32, 34.59,15.10,14.87 and 14.42.
5 NMR data for Example ldv
'H NMR (300 MHz,, CD3SOCD3 6 ppm 10.52 (NH, s, IH), 8.19 (ArH, d, J = 8.50 Hz, IH), 7.79-7.68 (ArH, m, 2H), 7.61 (ArH, d,./- 8.52 Hz, IH), 7.35 (ArH, d,J = 7.66 Hz, IH), 4.84-4.69 (CH2, m, IH), 4.42 (CH2f dd; J = 7.38, 5.30 Hz, 2H), 3.91 (CH2) dd, J = 14.30,7.97 Hz, 3H), 3.82-3.52 (CH2, m, $H), 3.45 (CH2, d, J = 2.42 Hz, IH), 3.26 - 3.15 (CH2] m, IH), 10 1.37 (CH3) d,J= 6.74 Hz, 3H), 1.25 (CH3, d,J = 6.76 Hz, 3H).
13CNMR(75 MHz, CD3SOCDj 6ppm 176.84,164.89,162.55,160.83,159.82,144.90, 138.19, 135.98,128.66, 125.02, 120.94,113.09,108.05,104.92, 70.90, 70.67, 66.84, 66.67, 52.32, 46.76, 44.30, 36.29,14-85 and 14.35.
15 NMR data for Example 1 dt
lH NMR (300 MHz, CDC13.5 ppm 8.58 (ArH, d, J =7.97 Hz, IH), 8.49 (ArH, d, J = 45.95 Hz, IH), 7.99 (ArH, d, J= 8.42 Hz, IH), 7.50 (ArH, dd, J = 17.31, 8.21 Hz, IH), 7.24 (ArH, d, J= 17.82 Hz, IH), 5.01-4.26 (CH2, s, br, IH), 4.65-4.39 (CH2, m, 3H), 4.33 (CH2, d,J = 6.25 Hz, IH), 4.04-3.58 (CH2, m, 8H), 3.49 (CH2, d, J = 11.36 Hz, IH), 3.31 (CH2, d, J -
20 2.99 Hz, IH), 1.41 (CH3, d,J= 6.72 Hz, 3H), 1.29 (CH3, d, J= 6.76 Hz, 3H) (I proton missing, lots of overlap seen, NH not seen either)
t3CNMR(75 MHz, CDCb 5ppm 171.52,165.39,162.90,161.33,160.01,145.14,139.06, 135.02,132.17,123.57,122.56,113.20, 105.21,71.29,70.92,67.25,66.93,52.78,46.95, 45.68, 44.51, 39.34,27.00,14-74 and 14.35.
25
NMR data for Example ldu
!H NMR (300 MHz, CDClj 2 ppm 8.12 (ArH, d, J = 8.32 Hz, 2H), 8.03 (ArH, d,./ = 8.43 Hz, IH), 7.51-7.39 (ArH, m, 3H), 5.08 (CH2, br, s, IH), 4.89 (CH2, d, J= 4.91 Hz, IH), 4.58 30 (CH2) d,J = 12.59 Hz, IH), 4.40 (CH2NH, br, s, 2H), 4.22 (NH, br, s, IH), 4.04-3.64 (CH2, m, 9H), 3.56 (CH2, dt, ./ = 11.80, H.45, 2.75 Hz, IH), 3.44-3.30 (CH2, m, IH), 2.87 (S02CH3s, 3H), 1.48 (CH3, d,J = 6.78 Hz, 3H), 1.35 (CH3: d,./= 6.81 Hz, 3H)

WO 2008/023161

PCT/GB2007/003179

l3CNMR (75 MHz, CDC3 5 ppm 165.16, 163.70, 162.51, 159.71, 138.71, 138.33, 134.99, 128.40,128.15, 113.45,105.20, 71.23, 70.89, 67.18, 66.90, 52.82, 47.07, 46.90, 44.21,41.25, 39.41, 14.78 and 14.38.
5 Tested in the Biological Assay; Ex. (lb) 0.00185^iM; Ex. (lc) 0.00184uM Ex. (Id)
0.00245(iM;Ex. (laz) 0.006865^M.
Tested in Alternative Enzyme Assay: Ex. (la) 0.0089jiM; Ex. (le) 0.0044nM; Ex. (If)
0.005nM; Ex. (Ig) 0-01 l^M; Ex. (lh) 0.0021 jiM; Ex. (li) 0.0056^iM; Ex. (lj) 0.035fiM; Ex.
(Ik) 0.015uM; Ex. (U) 0.0057^iM; Ex. (lm) 0.31^M; Ex. (In) 0.085uM; Ex. (lo) 0.14uM; 10 Ex. (lp) 0.038}iM; 0x. (lq) 0.39uM; Ex. (Ir) 0.23^M; Ex. (Is) 0.028nM; Ex. (It) 0.34nM;
Ex. (lu) 0.015jiM; fa- (lv) 0.18^M; Ex. (lw) 0.26nM; Ex. (Ix) 0.53nM; Ex. (ly) 0.33^M;
Ex. (lz) 0.37|iM; Ex (laa) 0.025jaM; Ex. (lab) 0.029nM; Ex. (lac) 0.14(iM; Ex. (lad)
0.0069^M; Ex. (lae) 0.38uM; Ex. (laf) 0.054uM; Ex. (lag) 0.029uM; Ex. (lah) 0.012uM;
Ex. (lai) 1.1 jiM; Ex- (laj) 0.49(iM; Ex. (lak) 0.017fiM; Ex. (lai) 0.23 ^M; Ex. (lam) 15 0.21nM; Ex. (Ian) 0-14^M; Ex. (lao) 0.0083uM; Ex. (lap) 0.02^M; Ex. (laq) 0.084uM; Ex.
(lar) 0.006nM; Ex. (las) 0.013|iM; Ex. (lat) 0.031^iM; Ex. (lau) 0.09jiM; Ex. (lav) 0.29|iM;
Ex. (law) 0.062nM; Ex. (lax) 0.0092uM;Ex. (lay) 0.15^M; Ex. (lba) 0.44uM; Ex. (lbb)
0.14uM; Ex. (lbc) 0-083^M; Ex. (lbd) 0.01 ljiM; Ex. (lbe) 0.18^M; Ex. (lbf) 0.06nM; Ex.
(lbg) 0.17jiM; Ex. (lbh) O.OHjaM; Ex. (lbi) 0.032^M; Ex. (lbj) 0.035nM; Ex. (lbk) 20 0.039p.M; Ex. (Ibl) 0.0027uM; Ex. (Ibm) O.055uM; Ex. (Ibn) 0.04^M; Ex. (Ibo) 0.018uM;
Ex. (lbp) O.lluM; Ex (lbq) O.H^M; Ex. (lbr) 0.056uM; Ex. (lbs) 0.039^iM; Ex. (lbt)
0.1 l|iM; Ex. (lbu) 0.O16nM; Ex. (lbv) 0.0051nM; Ex. (lbw) 0.036^M; Ex. (Ibx) 0.038MM;
0.0064nM; Ex. (led) 0.46nM; Ex. (Ice) 0.091nM; Ex. (lef) 0.073^M; Ex. (leg) 0.00026MM; 25 Ex. (lch) 0.22^M; Ex (lei) 0.15jaM; Ex. (lcj) 0.091HM; Ex. (lck) 0.065^M; Ex. (lcl)
0.2uM; EX. (1cm) QA6[iM; Ex. (len) 0.31jiM; Ex. (lco)2.5-M; Ex. (lep) luM; Ex. (lcq)
0.25MM; Ex. (lcr) 0-69jaM; Ex. (les) 7.5^M; Ex. (let) 0.024^M; Ex. (leu) 0.042^iM; Ex.
(lev) 0.3uM; Ex. (lew) 0.49^M; Ex. (lex) 0.12^iM; Ex. (Icy) 0.72^M; Ex. (Icz) 0.066)iM;
Ex. (Ida) 1.8JJM; Ejt- Odb) 0.03uM; Ex. (ldc) 0.02nM; Ex. (ldd) 0.073|iM; Ex. (lde) 30 0.0049(iM; Ex. (ldg) 0.014uM; Ex. (Idh) 0.041^M; Ex. (Idi) 0.23nM; Ex. (ldj) 0.25^M; Ex.
(Idk) 0.02(iM; Ex. (Idi) 0.018uM; Ex. (1dm) 0.0075uM; Ex. (ldn) 0.0O55[iM; Ex. (Ido)
0.03uM; Ex. (ldp) 0.OO67fiM; Ex. (ldq) 0.037^M; Ex. (Idt) 0.0026jiM; Ex. (ldu)

WO 2008/023161

PCT/GB2007/003179

0.00039uM; Ex. (ldv) 0.72uM; Ex. (ldw) 0.021uM; Ex. (ldx) 0.035uM; Ex. (ldy) 0.0035uM; Ex. (ldz) 0.099uM; Ex. (lea) 0.057uM; Ex. (leb) 0.17uM; Ex. (lee) 0.013uM; Ex. (led) 0.016^M; Ex. (lee) 0.0048yM.
Tested in phospho-Ser473 Akt assay: Ex. (Idf) 0.3813 uM; Ex. (Idr) 0.01415uM; Ex. (Ids) 5 0.06066uM.
Example 2

To a solution (0.2 M) of the appropriate chloro-substrate (1 equiv) in dioxane was added 15 diisopropylethylamine (2 equiv). To this mixture was then added the appropriate amine (2 equiv). The reaction was then heated under the influence of microwave radiation (120 °C, medium absorption setting) for 10 minutes. Upon completion the sample was concentrated in vacuo and the resulting residue dissolved in CH2CI2 and washed with H2O. The organic fraction was removed, dried (MgSO4). The crude residue was purified by flash 20 chromatography (S1O2) to give the desired products.


WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179


Tested in Alteraative Enzyme Assay: Ex. (2a) 0.7uM; Ex. (2b) 0.56uM; Ex. (2c) 0.6uM; Ex. (2d) 0.27uM: Ex. (2e) 0.35|iM; Ex. (2f) 0.17(iM; Ex. (2g) 0.064uM; Ex. (2h) 0.29|iM; Ex. 5 (2i) 0.64uM; Ex. (2j)0.2uM.

WO 2008/023161

PCT/GB2007/003179

Example 3:
(Compounds 3a to 3ab) R1 = (S)-3-methyI-morpholine R2 = (S)-3-methyI-morphoIine 5 Ar = aryl

Carboxy-substrates are reported in Example 1,
Method: Amide Formation
10 Conditions A:
The appropriate carboxy-substrate (1 equiv) was dissolved in DMF (0.067 M). HBTU (1.2 equiv) and appropriate amines (1.05 equiv) were added along with 3 drops of triethylamine at 0°C. The reaction vessels were sealed and the mixtures were stirred between 1 and 12 hours at room temperature. Upon completion the samples were concentrated in vacuo. The crude
15 residues were then purified by preparative HPLC to give the desired products.
Table 3:


WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB20U7/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB 200 7/003179


Tested in Alternative Enzyme Assay: Ex. (3a) 0.048uM; Ex. (3b) 0.32uM; Ex. (3c) 0.09uM; Ex. (3d) 0.28uM; Ex. (3e) 0.0047uM; Ex. (3f) 0.28^M; Ex. (3g) 0.0052uM; Ex. (3h) 0.18uM; Ex. (3i) 0.14uM; Ex. (3j) 0.17uM; Ex. (3k) 0.23uM; Ex. (31) 0.044uM; Ex. (3m) 5 0.32|jM; Ex. (3n) 0.23 uM; Ex. (3o) 0.37uM; Ex. (3p) 0.56uM; Ex. (3q) 0.12uM; Ex. (3r)

VO 2008/023161

PCT/GB2007/003179

0.5uM; Ex. (3s) 0.38uM; Ex. (3t) 0.042uM; Ex. (3u) 0.13uM; Ex. (3v) 0.16uM; Ex. (3w) 0.5uM; Ex. (3x) 0.24uM; Ex. (3y) 0.74uM; Ex. (3z) 0.34uM; Ex. (3aa) 0.026uM; Ex. (3ab) 0.14uM; Ex. (3ac) 1.6uM; Ex. (3ad) 0.066uM.
5 Example 4



Benzyl alcohol substrates are reported in Example 1.
10 The appropriate benzyl alcohol (1 equiv) was dissolved in CH2CI2 (0.08 M). Triethylamine (1 equiv) was added at room temperature, followed by the addition of thionyl chloride (2 equiv). The reaction mixture was stirred at 30°C for 45 minutes. Upon completion the reaction mixture was partitioned between brine and CH2CI2 and extracted with CH2CI2. Combined organic phases were dried (MgSO4), filtered and concentrated in vacuo. The crude residue
15 was purified by column chromatography on silica gel eluting with 10 to 70 % ethyl acetate in
hexane.
,0.



20

7-(3-Chloromethyl-phenyl)-2,4-bis-((S)-3-methyl-morpholiri-4-yl)-pyrido[2,3-d]pyrimidine: (72 % yield, 90 % purity) m/z (LC-MS, ESP): 454 [M+H]+ R/T = 3.15 min

WO 2008/023161 PCT/GB 20ft 7/003179
The appropriate benzyl alcohol (1 equiv) was dissolved in CH2CI2 (0.052 M). Thionyl chloride (3.3 equiv) was added. The reaction mixture was heated up to 55°C and a solution of triethylamine (1.7 equiv) in CH2CI2 (0.044 M) was added dropwise over 10 minutes. The reaction mixture was allowed to stir at 30°C for 10 minutes. Upon completion the reaction 5 mixture was partitioned between brine and CH2CI2 and extracted with CH2CI2. Combined organic phases were dried (MgS04), filtered and concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 10 to 50 % ethyl acetate in hexane.
7-(4-Chloromethyl-phenyl)-2,4-bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2)3-d]pyrimidine: 10 (65 % yield, 90 % purity) m/z (LC-MS, ESP): 454 [M+H]+ R/T = 3.15 min

The appropriate benzyl alcohol (1 equiv) was dissolved in CH2CI2 (0.044 M). Thionyl chloride (3.3 equiv) was added. The reaction mixture was heated up to 55°C and a solution of triethylarnine (1.7 equiv) in CH2CI2 (0.044 M) was added dropwise over 10 minutes. The
15 reaction mixture was allowed to stir at 30°C for 30 minutes. Upon completion the reaction mixture was partitioned between brine and CH2CI2 and extracted with CH2CL2. Combined organic phases were dried (MgSO,)), filtered and concentrated in vacuo. The crude residue was used without further purification. 7-(3-Chloromethyl-4-fluoro-phenyl)-2,4-bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-
20 djpyrimidine: (96 % yield, 90 % purity) m/z (LC-MS, ESP): 472 [M+H]+ R/T = 3.96 min



WO 2008/023161

PCT/GB2007/003179

The appropriate benzyl alcohol (1 equiv) was dissolved in CH2CI2 (0.086 M). Triethylamine (2.5 equiv) and thionyl chloride (2.5 equiv) were added. The reaction mixture was heated up to 45°C afor 3 hours. Upon completion the reaction mixture was partitioned between water and CH2CI2 and extracted with CH2CI2. Combined organic phases were dried (MgSO,f), 5 filtered and concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 10 to 50 % ethyl acetate in hexane.
7-(3-Chloromethyl-4-methoxy-phenyl)-2,4-bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidine: (37 % yield, 90 % purity) m/z (LC-MS, ESP): 484 [M+H]+ R/T = 3.21 min



10

(Compounds 4a to 4ak) R4 = (S)-3-methyJ-morpholine R2 = (S)-3-methyJ-morpholhie Ar = aryl

Method: Benzvlamines. benzvlethers and bcnzylsulfones formation
Conditions A:
The appropriate chlorobenzyl-substrate (1 equiv) was dissolved in THF (0.067 M). The
appropriate amine (80 equiv) as well as triethylamine (1 equiv) was added. The reaction 20 vessels were sealed and the mixtures were stirred for 3 to 5 hours at 95°C. Upon completion
the samples were concentrated in vacuo. The crude residues were then purified by preparative
HPLC to give the desired products.
Conditions B:
The appropriate chlorobenzyl-substrate (1 equiv) was dissolved in an aqueous ammonia/n-25 butanol (1.5:1) solution (0.011 M). The reaction vessel was sealed and the mixture was stirred
for 10 minutes at 140°C. Upon completion the sample was concentrated in vacuo. The crude
residue was then purified by preparative HPLC to give the desired products.
Conditions C:
The appropriate chlorobenzyl-substrate (1 equiv) and sodium hydroxide (1 equiv) were 30 dissolved in ethanol (0.011 M). The reaction vessel was sealed and the mixture was stirred for
3 hours at 50°C Upon completion the sample was concentrated in vacuo. The crude residue
was then purified by preparative HPLC to give the desired products.

WO 2008/023161

PCT7GB2007/003179

Conditions D:
The appropriate chlorobenzyl-substrate (1 equiv) was dissolved in DMF (0.022 M). Imidazole (3 equiv) and potassium re/-£-butoxide (3 equiv) were added. The reaction vessel was sealed and the mixture was stirred for 2 hours at room temperature. Upon completion the sample was 5 concentrated in vacuo. The crude residue was then purified by prepai-ative HPLC to give the desired product. Conditions E:
The appropriate chlorobenzyl-substrate (1 equiv) was dissolved in DMF (0.066 M). Sodium sulfonate (1.3 equiv) was added. The mixture was stirred for 2 hours at 125°C. Upon
10 completion the sample was concentrated in vacuo. The crude residue was then purified b}' preparative HPLC to give the desired product. Conditions F:
The appropriate chlorobenzyl-substrate (1 equiv), potassium carbonate (2.6 equiv) triethylamine (1 equiv) and the appropriate amine (1.1 equiv) were suspended in DMF (0.028
15 M). The reaction vessel was sealed and the mixture was stirred for 16 hours at 40°C. Upon completion the sample was filtered through a silica cartridge, washed with CH2CI2 and then concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired products.
20 Table 4:


WO 2008/023161

PCT/GB2007/003I79





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/W03179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161 PCT/GB2007/003179


WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/00317!)


NMR data for example 4h
fH NMR (300 MHz, DMSO) 5 ppm 8-21 (ArH, d,J = 8.39 Hz, 2H), 8.08-8.01 (ArH, m, IH), 7.65 (ArH; d, J = 8.49 Hz, IH), 7.49 (ArH, d, J = 4.85 Hz, 2H), 4.82-4.72 (CH2, m, IH), 4.45 5 (CH2,+ NHm, 3H)3 3.99-3.82 (CH2; m, 7H), 3.69 (CH2, ddd, 7= 19.97, 8.86, 5.32 Hz, 8H), 3.53 (CH2, t, J - 5.65, 5.65 Hz, 2H), 3-29-3.15 (CH2, m, 2H), 1.38 (CH3, d, J= 6.75 Hz, 3H), 1.25 (CH3,d, .7=6.75 Hz, 3H)
NMR data for Example 4r
10 'HNMR (300 MHz, CDC13 8 ppm 8.19 (ArH, s, IH), 8.03 (ArH, ddd, J = 8.43, 5.31, 3.28 Hz, 2H), 7.54-7.37 (ArH, m, 3H), 5.00-4.85 (CH, m, IH), 4.68-4.56 (CH2, m, IH), 4.36 (CH2, ddd, J = 6.83, 4.79,2.16 Hz, 2H), 4.07-3.92 (CH2,m, 2H), 3.91-3.66 (CH2, m, 11H), 3.63-3.49 (CH2, m, IH), 3.39 (CH2) dd, J = 13.37, 3.58 Hz, IH), 3.04-2.92 (CH2j m, IH), 2.80 (CH2,d)t/= 10.30 Hz, m),2.65(CB2,dd,J=: 10.23,4.92 Hz, IH), 2.52-2.39 (CH2, m, IH),
15 2.21 (CH2, d, J= 7.02 Hz, IH), 1.89-1.73 (CH2, m, IH), 1.46 (CH3) d, J= 6.77 Hz, 3H), 1.35 (CH3,d,J=6.81Hz,3H)
,3CNMR(75 MHz, CDC13) 5ppm 165.57,163.03,162.50,160.11,139.06,134.83,130.62, 128.81, 127.10,113.70, 105.06,71.44, 71.06,67.41, 67.25,67.07, 62.87, 60.08, 52.98, 52.49, 47.07,44.58, 39.47, 35.02, 14.86 and 14.90.
20 •

WO 2008/023161

PCT/GB2007/003179

NMR data for Example 4s
lH NMR (300 MHz, CDC13) 5 ppm 8.22 (ArH, s, IH), 8.11-7.96 (ArH, m, 2H), 7.48 (ArH, dd, J= 10.85, 7.98 Hz, 3H), 4.99-4.86 (CH, m, IH), 4.68-4.55 (CH, m, IH), 4.44-4.30 (CH2, m, 2H), 4.06-3.92 (CH2, m, 2H), 3.93-3.65 (CH2, m, 10H), 3.62-3.50 (CH2, m, IH), 3.39 5 (CH2,dd,y= 13.39, 3.57 Hz, IH), 3.14-3.01 (CH2,m, IH), 2.88 (CH2, &,J= 10.59 Hz, IH), 2.77-2.67 (CH2, m, IH), 2.63-2.43 (CH2] m, IH), 2.31-2.14 (CH2, m, IH), 1.92-1.79 (CH2, m, IH), 1.47 (CH3, d, J = 6.77 Hz, 3H), 1.35 (CH3, A, J = 6.81 Hz, 3H)
13CNMR(75 MHz, CDC13)) 5 ppm 165.56,163.03,162.34,160.12, 139.16,134.89,130.80, 128.94, 128.91, 127.39, 113.69,105.11, 71.44, 71.21, 71.06,67.40, 67.06, 62.61, 59.93, 10 52.98, 52.42,47.08, 44.58, 39.47, 34.88, 31.73,22.80,14.86 and 14.91.
Tested in the Biological Assay: Ex. (4f) 0.001967uM.
Tested in Alternative Enzyme Assay: Ex. (4a) 0.0016uM; Ex. (4b) 0.025uM; Ex. (4c)
0.093^iM; Ex. (4d) 0.013pM; Ex. (4e) 0.0019uM; Ex. (4f) 15 (4h) 0.031nM; Ex. (4i) 0.027uM; Ex. (4j) 0.054nM; Ex. (4k) 0.016uM; Ex. (41) 0.0091^iM; Ex. (4m) 0.015uM; Ex. (4n) 0.0071pM; Ex. (4o) 0.021 pM; Ex. (4p) 0.17jiM; Ex. (4q) 0.13uM; Ex. (4r) 0.04uM; Ex. (4s) 0.029uM; Ex. (4t) 0.09uM; Ex. (4u) 0.027uM; Ex. (4v) 0.14uM; Ex. (4w) 0.028uM; Ex. (4x) 0.12uM; Ex. (4y) 0.13uM; Ex. (4z) 0.13uM; Ex. (4aa) 0.2 luM; Ex. (4ab) UfiM; Ex. (4ac) 0.0S7uM; Ex. (4ad) 0.081uM; Ex. (4ae) 0.16(iM; Ex.
20 (4af) 0.58uM; Ex. (4ag) 0.54uM; Ex. (4ah) 0.2nM; Ex. (4ai) 0.22uM; Ex. (4aj) 0.46^iM; Ex. (4ak) 0.015fiM; Ex. (4al) 0.064uM; Ex. (4am) 0.024\iU; Ex. (4an) 0.095uM; Ex. (4ao) 0.064uM; Ex. (4ap) 0.1 luM; Ex. (4aq) 0.012uM; EX. (4ar) 0.06uM; Ex. (4as) 0.091uM; Ex. (4at) O.U^M; Ex. (4au) 0.096nM; Ex. (4av) 0.003&^vM; Ex. (4aw)G.UyiM;Ex. 25 0.076uM; Ex. (4bd) 0.12uM; Ex. (4be) 0.049uM; Ex. (4bf) 0.059uM.
Example 5

30 Benzyl chloride substrates are reported in Example 4.

WO 2008/023161

PCT/GB2007/003179

The appropriate benzyl chloride (1 equiv) was dissolved in an ammonium hydroxide and n-butanol (1.5:1) solution (0.01 M). The reaction vessel was sealed and the mixture exposed to microwave radiation (140 °C, medium absorption setting) for 10 minutes. Upon completion the reaction mixture was partitioned between brine and ethyl acetate and extracted with ethyl 5 acetate. Combined organic phases were dried (MgSO.4), filtered and concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 0 to 5 % methanol in CH2CI2.

3-[2)4-Bis-((S)-3-memyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidin-7-yIj-benzylamine: (81 % 10 yield, 100 % purity) m/z (LC-MS, ESP): 435 [M+H]+ R/T = 2.44 min

542,4-Bis-((S)-3-memyl-moirhoUn-4-yl)-pyrido[23-d]pyrimidm-7-yl]-2-fluoro-benzylamine: (85 % yield, 98 % purity) m/z (LC-MS, ESP): 453 [M+H]+ R/T = 3.21 min
15

4-[2;4-Bis-(3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-benzylamirLe:(95 % yield, 97 % purity) m/z (LC-MS, ESP): 435 [M+H]+ R/T = 2.36 min

WO 2008/023161

PCT/GB2007/003179


The appropriate benzyl chloride (1 equiv) was dissolved in a 2 M solution of methylamine in THF (80 equiv). Triethylamine (1 equiv) was added. The reaction mixture was stirred at 95°C for 2.5 hours. Upon completion the reaction mixture was concentrated in vacuo and the 5 residue was diluted with ethyl acetate and n-tmtanol and the organic phase was washed with brine, dried (MgSC4), filtered and concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 0 to 7 % methanol in CH2CI2.



10 {3-[2,4-Bis-((S)-3-methyl-morpholin~4-yl)-pyrido[2>3-d]pyrirnidin-7-yl]-benzyl}-metliyl-
amine: (77 % yield, 94 % purity) m/z (LC-MS, ESP): 449 [M+H]+ R/T = 2.44 min ,0,



15

{4-[2,4-Bis-(3-methyl-morpholm-4-yl)-py^^
(93 % yield, 87 % purity) m/z (LC-MS, ESP): 449 [M+H]+ R/T = 2.40 min
Procedures for the synthesis of Examples 5a to 5z
R4 = (S)-3-raethyI-morpholine R2 = CSV3-methvl-mnmhnline

' /

WO 2008/(123161

PCT/GB2007/003179

Conditions A:
The appropriate aminobenzyl-substrate (1 equiv) was dissolved in CH2CI2 (0.035 M). The appropriate acyl chloride or acid anhydride (2 equiv) as well as triethylamine (1 equiv) was then added. The mixtures were stirred for 2 hours at room temperature. Upon completion the 5 samples were concentrated in vacuo. The crude residues were then purified by preparative HPLC to give the desired products.
Conditions B:
10 The appropriate methylaminobenzyl-substrate (1 equiv) was dissolved in CH2CI2 (0.035 M). The appropriate acyl chloride or acid anhydride (2 equiv) as well as triethylamine (1 equiv) were added. The mixtures were stirred for 12 hours at 95°C. Upon completion the samples were concentrated in vacuo. The crude residues were then purified by preparative HPLC to give the desired products.
15
Table 5:


WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/W>3179





WO 2008/023161

PCT/GB2007/003179

Tested in Alternative Enzyme Assay: Ex. (5a) 0.023uM; Ex. (5b) 0.054uM; Ex. (5c) 0.12uM; Ex. (5d) 0.12uM; Ex. (5e) 0.12uM; Ex. (5f) 0.37uM; Ex. (5g) 0.12uM; Ex. (5h) 0.19uM; Ex. (5i) 0.2uM; Ex. (5j) 0.31uM; Ex. (5k) 0.89uM; Ex. (51) 0.049uM; Ex. (5m) 5 1.4uM; Ex. (5n) 0.64uM; Ex. (5o) 0.12jiM; Ex. (5p) 0.5uM; Ex. (5q) 0.091uM; Ex. (5r) 0.56uM; Ex. (5s) 0.67uM; Ex. (5t) 0.057uM; Ex. (5u) 0.16uM; Ex. (5v) 0.14}iM; Ex. (5w) 0.16uM; Ex. (5x) 0.29uM; Ex. (5y) 0.44uM; Ex. (5z) 1.4uM.
Example 6

10
The chloro-substrate was reported in Example 1.
The appropriate chloro-substrate (1 equiv) was dissolved in n-butanol (0.055 M). 2-
formylfuran-3-boronic acid (1.0 equiv), potassium carbonate (1.2 equiv), and
tetrakis(triphenylphosphine)palladium0 (0.05 equiv) were added. The reaction vessel was
15 sealed and exposed to microwave radiation (110 °C, medium absorption setting) for 15
minutes. Upon completion the reaction mixture was filtered through a silica cartridge and the
filtrate was concentrated in vacuo. The crude residue was purified by column chromatography
on silica gel eluting with 40 % ethyl acetate in hexane to give the desired product.

20


3-[234-Bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidin-7-yl]-furan-2-carbaldehyde: (26 % yield, 90 % purity) m/z (LC-MS, ESP): 424 [M+H]+ R/T = 2.81 min

WO 2008/023161

PCT/GB2007/003179

(Compound 6a)



The above product was dissolved in THF (0.018 M) and sodium borohydride (2 equiv) was added. This mixture was allowed to stir at room temperature for 5 minutes. Upon completion 5 the reaction mixture was filtered through a silica cartridge and the filtrate was concentrated in vacuo. The crude residue was purified by preparative HPLC to give the desired product.
Table 6:

Purity (%) Retention time (min) m/z [M+H]+ Example Structure
6a 96 6.89 426.3 H3
10 Tested in Alternative Enzyme Assay: Ex. (6a) 0.0l3juM Example 7

15 The chioro-substrate was reported in Example I.

WO 2008/023161

PCT/GB2007/003179

The appropriate chloto-substrate (1 equiv) was dissolved in dioxane (0.16 M). 5-formyl-2-furylboronic acid (1.05 equiv), tripotassium phosphate (1.5 equiv) and bis(tri-t-butylphosphine)palladium (0.05 equiv) were added. The reaction vessel was sealed and 5 exposed to microwave radiation (170 °C, medium absorption setting) for 45 minutes. Upon completion the reaction mixture was partitioned between water and CH2CI2 and extracted with CH2CI2. Combined organic phases were dried (MgS04)3 filtered and concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 40 to 100 % ethyl acetate in hexane to give the desired product.

5-[2,4-Bis-(3-methyl-morphoLin-4-yI)-pyrido[2}3-d]pyrimidin-7-yl]-ftiran-2-carboxaldehyde: (100 % yield, 100 % purity) m/z (LC-MS, ESP): 424 [M+H]+ R/T = 2.75 min
(Compounds 7a to 7k)

The appropriate formylfuran-substrate (1 equiv) was dissolved in a THF/CH2CI2 (1:1) solution (0.036 M). The appropriate amines (2.2 equiv) sodium borohydride (2.4 equiv) and acetic acid (0.03 equiv) were added. The reaction mixture was stirred at room temperature for 24 hours. Upon completion the samples were filtered through a silica cartridge, washed with 20 methanol and then concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired products.
Table 7:

WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/00317!)


Tested in Alternative Enzyme Assay: Ex. (7a) 0.59uM; Ex. (7b) 0.13uM; Ex. (7c) O.091^M; Ex. (7d) 0.097uM; Ex. (7e) O.lSuM; Ex. (7f) 0.12uM; Ex. (7g) 0.17uM; Ex. (7h) 0.33uM; Ex. (7i) 0.079uM; Ex. (7j) 0.12uM; Ex. (7k) 0.14uM.

WO 2008/023161

PCT/GB2007/003179

Example 8 (Compounds 8a to 8b)



5 The methylbenzoic ester substrates were reported in Example 1.
Conditions A:
Example Iba (1 equiv) was dissolved in dioxane (0.16 M). Ethanolainine (51.0 equiv) was
added. The reaction vessel was sealed and exposed to microwave radiation (130 °C, medium
10 absorption setting) for 50 minutes. Upon completion the reaction mixture was concentrated in vacuo. The crude residue was then purified by column chromatography on silica gel using a gradient 0 to 5 % MeOH in CH2CI2 to afford the desired product. Conditions B: Example lbg (1 equiv) was dissolved in dioxane (0.05 M). Ethanolamine (2.0 equiv) was
15 added. The reaction vessel was sealed and exposed to microwave radiation (130 °C3 medium
absorption setting) for 2 X 20 minutes. Upon completion the reaction mixture was
concentrated in vacuo. The reaction mixture was partitioned between water and CH2CI2 and
extracted with CH2CI2. Combined organic phases were dried (MgSCU), filtered and
concentrated in vacuo. The crude residue was then purified by column chromatography on
20 silica gel using a gradient 0 to 5 % MeOH in CH2G2 to afford the desired product.
,0. .(X

Conditions C:
To a solution of the appropriate carboxylic acid derivative (1 equiv) suspended in CH2CI2 was
added HBTU (1.3 equiv) followed by diisopropylethylamine (3 equiv). The mixture was

WO 2008/023161

PCT/GB2007/003179

cooled (-78 °C) and the appropriate amine added (1.1 equiv). The mixture was stirred for 3 hrs before being concentrated to dryness and purified by preparative HPLC to give the desire products.
5 Table 8:

NMR data for Example 8a
lHNMR(300 MHz, CDC13) 6ppm 8.55 (ArH, s, IH), 8.09 (ArH, d, J = 7.85 Hz, IH), 7.95 (ArH, dsy= 8.42 Hz, IH), 7.86 (ArH, d,J = 7.86 Hz, IH), 7.49-7.33 (ArH, m, 2H), 4.89-4.75 10 (CH, m, IH), 4.56-4.46 (CH, m, IH), 4.38-4.26 (CH2, m, IH), 3.97-3.87 (CH2, m, 2H), 3.85-

WO 2008/023161

PCT/GB2007/003179

3.75 (CH2, m,4H), 3.72-3.55 (CH2, m, 7H), 3.53-3.44 (CH2, m, 1H), 3.34-3.24 (CH2, ra, 1H),
1.41 (CH3j d, J = 6.17 Hz, 3H), 1.28 (CH3, d, J = 6.82 Hz, 3H).
Tested in Alternative Enzyme Assay: Ex. (8a) 0.028p.M; Ex. (8b) 0.079uM; Ex. (8c)
0.13uM;Ex.(8d)2uM.

10 The benzyl alcohol substrate was reported in Example 1.
20 Tested in Alternative Enzyme Assay: Ex. (9a) 0.088uM.
Example Ibc (1 equiv) was dissolved in THF (0.022 M). Sodium terf-butoxide (3.0 equiv) and iodomethane (10.0 equiv) were added. The reaction vessel was stirred at room temperature for 48 hours. Upon completion the sample was filtered through a silica cartridge, 15 washed with EtOAc and then concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired product.


WO 2008/023161

PCT7GB2007/003179

Example 10 (Compound 10a)



5 The pyridinone substrate was reported in Example 13.
Example 13c (1 equiv) was dissolved in DMF (0.1 M). Potassium carbonate (1.1 equiv) and iodomethane (1.1 equiv) were added. The reaction vessel was stirred at 100°C for 2 hours. Upon completion the sample was concentrated in vacuo. The crude residue was then purified 10 by preparative HPLC to give the desired product.
Table 10:

15

WO 2008/023161

PCT/GB2007/003179


Example 11 (Compound 11a)
5 The sulfonamide substrate was reported in Example 1.
Example lat (1 equiv) was dissolved in DMF (0.1 M). Potassium carbonate (2.0 equiv) and iodomethane (1.5 equiv) were added. The reaction vessel was heated at 100°C for 2 hours. Upon completion the sample was concentrated in vacuo. The crude residue was then pui'ified 10 by preparative HPLC to give the desired product.
Tested in Alternative Enzyme Assay: Ex. (11a) 0.37uM.
Table 11;

15

WO 2008/023161

PCT/GB2007/003179

Example 12

To a solution of the appropriate 7-substituted-lH-pteridine-2,4-dione (1 equiv) in anhydrous 5 toluene (sufficient to make a 0.1 M solution) was added Hunig's base (3 equiv), A reflux condenser was attached to the reaction vessel and the mixture heated, under an inert atmosphere) to 70 °C for 30 minutes. After this time, the reaction was cooled to 40 DC whereupon POCl3 (3 equiv) was added. The mixture was then heated, with stirring, to 110 °C for 3 hrs. Upon completion, the reaction was cooled and concentrated in vacuo to give a tany 10 residue which was dissolved in the minimum volume of CH2CI2 and filtered through a thick silica pad. The resulting filtrate was concentrated in vacuo to give the desired 2,4-dichloro-7-substituted-pteridine product (typically 65-99 % yield) in suitably pure form to be used without any further purification.
15 2)4-Dichloro-7-p-tolyl-pteridine;R7=toluyl, R2= CI, R4=C1, X=N, Y=C, Z=N: (61 % yield, 99 % purity) m/z (LC-MS, ESP): Did not ionize, R/T = 3.27 min
2,4-Dicworo-7-phenyl-pteridine;R7=phenyl, R2= CI, R4=C1, X=N, Y=C, Z=N: (66 % yield, 99 % purity) m/z (LC-MS, ESP): Did not ionize, R/T = 3.10 min

To a cooled (-5 °C) solution of the appropriate amine (1 equiv = R4) in N,N-dimethylacetamide (sufficient to make 0.2 M solutiion) was added the appropriate 2,4-25 dichloro-7-substituted-pteridine (1 equiv added as a 0.04 M solution in N,N-
dimethylacetamide). After approx 10 minutes Hunig's base was added (1 equiv) and the resultant mixture stirred at -5 °C for 30 minutes. After this time, the reaction was allowed to

WO 2008/023161

PCT/GB20D7/003179

warm to room temperature, whereupon the appropriate amine (1 equiv = R2) and Hunig's base (1 equiv) were then added. The resultant mixture was heated to 60 °C and maintained at this temperature, with stirring, for 16 hours. Upon completion, the mixture was allowed to cool to room temperature before being purified by preparative HPLC to give the desired 5 product. Table 12


WO 2008/023161

PCT/GB2007/003179

Tested in Alternative Enzyme Assay: Ex. (12a) 0.02669uM; Ex. (12b) 0.2147uM; Ex. (12c) 0,04872uM; Ex. (12d) 0.0263|iM; Ex. (12e) 0.5414uM.
Example 13
5 (Compounds 13a to 13f)

The pyridine substrates were reported in Example 1. Conditions A:
10 Example Iw (1 equiv) was dissolved in a dry THF/methanol (1:1) solution (0.057 M). Sodium hydride (4.5 equiv) was added. The reaction mixture was stirred at room temperature for 15 minutes under nitrogen. The reaction vessel was sealed and the mixture exposed to microwave radiation (130 °C, medium absorption setting) for 40 minutes. Upon completion the sample was concentrated in vacuo. The crude residue was then purified by preparative
15 HPLC to give the desired product. Conditions B:
Example lw (1 equiv) was dissolved in dry THF (0.057 M). Dimethylethanolamine (10.0 equiv) and sodium hydride (5.0 equiv) were added. The reaction mixture was stirred at room temperature for 15 minutes under nitrogen. The reaction vessel was sealed and the mixture
20 exposed to microwave radiation (130 °C, medium absorption setting) for 20 minutes. Upon completion the sample was concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired product. Conditions C: Example lau (1 equiv) was dissolved in DMSO (0.59 M). 8N aqueous sodium hydroxide
25 solution (50.0 equiv) was added. The reaction vessel was sealed and the mixture exposed to microwave radiation (130 °C, medium absorption setting) for 20 minutes. Upon completion concentrated aqueous HC1 was added carefully. The mixture was neutralized with 2N aqueous sodium hydroxide solution. The suspension was diluted with methanol then filtered through a

WO 2008/023161

PCT/GB200T/003179

sintered funnel. The filtrate was concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired products. Conditions D:
Example lau (1 equiv) was dissolved in NMP (0.1 M). Potassium cyanide (20.0 equiv) was 5 added. The reaction vessel was sealed and the mixture exposed to microwave radiation (130 °C, medium absoiption setting) for 46 hours. Upon completion the reaction mixture was partitioned between water and CH2CI2. The aqueous phase was extracted with CH2C12. Combined organic phases were dried (MgSO-iX filtered and concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 50 to 100 % ethyl
10 acetate in hexane to give the desired product. Conditions E:
Example lau (1 equiv) was dissolved in NMP (0.1 M). Potassium cyanide (20.0 equiv) was added. The reaction vessel was sealed and the mixture exposed to microwave radiation (130 °C, medium absorption setting) for 46 hours. Upon completion the reaction mixture was
15 partitioned between water and CH2CI2. The aqueous phase was extracted with CH2CI2. Combined organic phases were dried (MgSO*)), filtered and concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 50 to 100 % ethyl acetate in hexane first, then eluting with 10 % methanol in CH2CI2. The crude fractions were then further purified by preparative HPLC to give the desired product.
20 Conditions F:
Example lah (1 equiv) was dissolved in NMP (0.1 M). Potassium cyanide (8.0 equiv) was added. The reaction vessel was sealed and the mixture exposed to microwave radiation (180 °C, medium absorption setting) for 40 minutes. Upon completion the sample was filtered through a silica cartridge, washed with EtOAc and then concentrated in vacuo. The crude
25 residue was then purified by preparative HPLC to give the desired products.

WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2l)in/0l>317!>


Tested in Alternative Enzyme Assay: Ex. (13a) 0.2uM; Ex. (13b) 0.33uM; Ex. (13c) 0.14uM; Ex. (13d) 0.48uM; Ex. (13e) 0.19uM; Ex. (13f) 0.16uM; Ex. (13g) 0.1 luM.
5 Example 14
(Compounds 14a-14b)

The ester substrate was reported in Example I.
10
Ester hydrolysis: Conditions A
Example lbg (1 equiv) was dissolved in methanol (0.2 M). IM Sodium hydroxide aqueous
solution (5.0 equiv) was added. The reaction mixture was stirred at room temperature for 3
15 hours. Upon completion the reaction mixture was neutralised with IM aqueous HCl and

WO 2008/023161

PCT/GB2007/003179

concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 0 to 10 % MeOH in CH2CI2 to give the desired product. Amide formation: Conditions B 5 Example lbg (1 equiv) was suspended in THF (0.05 M). Thionyl chloride (2.5 equiv) was added dropwise at 40°C. The reaction mixture was then heated for an hour at 40°C. Ammonia gas was then slowly bubbled into the reaction mixture. THF was then added for further dilution (0.025 M) and the reaction mixture was heated for an hour at 40°C. Upon completion the reaction mixture was cooled down and concentrated in vacuo. The residue was partitioned 10 between water and CH2CI2. The aqueous phase was extracted with CH2CI2. Combined organic phases were dried (MgSCU), filtered and concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 0 to 5 % MeOH in CH2CI2 to give the desired product.
15 Table 14:

NMR data for Example 14a
*HNMR (300 MHz, CDC13) 5ppm 8.82-8.69 (ArH, m, IH), 8.68-8.56 (ArH, m, IH), 8.03-7.90 (ArH, m, IH), 7.52-7.39 (ArH, m, IH), 7.18-7.05 (ArH, m, IH), 4.92^.80 (CH, m5 IH), 20 4.61-4.47 (CH, m, IH), 4.37-4.27 (CHa> m, IH), 4.07 (OCH3, s, 3H), 4.00-3.87 (CH2j m, 2H),

WO 2008/023161

PCT/GB2007/003179

3.85-3.60 (CH2, m, 6H)3 3.57-3.24 (CH2, m, 3H), 1.41 (CH3, d, J = 6.65 Hz, 3H), 1.30 (CH3, d,J = 6.74Hz,3H)
l3CNMR(75 MHz, CDC13) 5ppm 165.33,134.98,134.96,132.64,132.61,132.58,119.77, 112.83,112.11,100.01, 71.29, 70.90,67.24,66.91, 52.80,46.96,44.44, 39.34 and 14.74. 5
NMR data for Example 14b
'H NMR(300MHz, CDCh) 5ppm 8.83 (ArH, d, J = 2.46 Hz, IH), 8.64 (ArH, dd, J = 8.76, 2.49 Hz, IH), 8.01 (ArH, d, J = 8.47 Hz, IH), 7.71 (NH, s, br, IH), 7.57 (ArH, d, J = 8.50 Hz, IH), 7.13 (ArH, d, J- 8.83 Hz, IH), 5.79 (NH, s, br, IH), 5.00-4.84 (CH, m, IH), 4.62
10 (CH, dd, J = 13.82,0.70 Hz, IH), 4.37 (CH2, d, J = 6.77 Hz, IH), 4.05 (OCH3, s, 3H), 4.03-3.94 (CH2, m,2H), 3.91-3.79 (CH2, m, 3H), 3.79-3.63 (CH2, m, 4H), 3.64-3.51 (CH2, m, IH), 3.44-3.30 (CH2,m, IH), 1.47 (CH3j d, J= 6.78 Hz, 3H), 1.35 (CH3, d, J= 6.81 Hz, 3H) 13CNMR(75 MHz, CDC13) 5 ppm 166.58, 165.45,162.87,159.99,159.22,134.71,133.75, 131.84,131.65,120.52,113.07,111.87,104.80,102.94, 71.33, 70.94, 67.29, 66.94, 56.28,
15 52.80,46.93, 44.49, 39.33,14.72 and 14.34.
Tested in Alternative Enzyme Assay: Ex. (14a) 0.00Ol5uM; Ex. (14b) 0.0032(iM.
Example 15

The chloro-substrate was reported in Example 1.
To a mixture of 7-chloro-4-((S)-3-methyl-morpholin-4-yl)-2-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrimidine (1 equiv), potassium carbonate (1.2 equiv), and 3-BOC-25 aminophenylboronic acid (1.2 equiv) in acetonitrile/water (1:1) (0.08 M of chloro-substrate) was added tetrakis(triphenylphosphine) palladium0 (0.05 equiv). The reaction vessel was sealed and exposed to microwave radiation (130 °C, medium absorption setting) for 10 minutes under nitrogen atmosphere. Upon completion the samples were filtered through a

WO 2008/023161

PCT/GB20O7/O03179

silica cartridge, washed with ethyl acetate and then concentrated in vacuo. The crude residue was used as such in the next reaction.

{3-[2)4-Bis-((S)-3-methyI-morpholin^-yl)-pyrido[23-d]pyrimidin-7-yl]-phenyl}-carbarnic 5 acid tert-butyl ester: (95 % yield, 100 % purity) m/z (LC-MS, ESP): 520.9 [M+H]+ R/f = 3.23 min
The above product (1 equiv) was dissolved in a TFA/ CH2CI2 solution (1:20) (0.018 M). The reaction mixture was stirred at room temperature for 15 hours. The reaction mixture was then 10 concentrated in vacuo. The residue was partitioned between water and CH2CI2. The aqueous phase was neutralized with IN aqueous sodium hydroxide. Combined organic phases were dried (MgSO,*), filtered and concentrated in vacuo. The crude residue was used as such in the next reaction.
c°i

15 3-[2,4-Bis-((S)-3-methyl-morpholm-4-yJ)-pyrido[2,3-d]pyrimidin-7-yl]-phenylamine: (100 % yield, 100 % purity) m/z (LC-MS, ESP): 520.9 [M+H]+ R/T = 2.72 min
(Compound 15a)


WO 2008/023161

PCT/GB2007/003179

The above product (1 equiv) was dissolved in THF (0.013 M). Chloroethanesulfonyl chloride (3.5 equiv) was gently added to the reaction mixture at 0°C and the reaction mixture was stirred at room temperature for 15 hours. 8N Aqueous sodium hydroxide (50 equiv) was then added and the reaction mixture was heated at 40°C for 12 hours. The reaction mixture was 5 concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 0 to 5 % MeOH in CH2CI2 to give the desired product.

3-[2,4-Bis-((S)-3-methyl-morpholin^-yl)-pyrido[2,3-d]pyrirnidin-7--yll-phenylamme (1
10 equiv) was dissolved in THF (0.1 M). Pyridine (10 equiv) and isopropylsulfonyl chloride (10 equiv) were added to the reaction mixture at room temperature. The reaction mixture was then stirred at 90°C for 4 hours. The reaction mixture was partitioned between CH2Q2 and water. Organic phase was dried (MgSCU), filtered and concentrated in vacuo. The crude residue was purified by column chromatography on silica gel eluting with 0 to 60 % EtOAc in hexane to
15 give the desired product.

3-[2)4-Bis-((S)-3-methyl-morpholin-4-yl)-pyrido[2,3-d]pyrirnidin-7-yl]-phenylamine (1
equiv) was dissolved in CH2CI2(0.24 M). Tetrahydro-2-furoic acid (1.1 equiv), HBTU (2.0 20 equiv) and triethylamine (2 equiv) were added and the reaction mixture was then stirred at room temperature for 3 hours. The reaction mixture was partitioned between CH2CI2 and water. Organic phase was dried (MgSO,4), filtered and concentrated in vacuo. The crude

WO 2008/023161

PCT/GB2
residue was purified by column chromatography on silica gel eluting with 0 to 4 % MeOH in TBME to give the desired product.
Table 15:

NMR data for Example 15b
'HNMR(300 MHz), CDC13 §ppm 8.00-7.94 (ArH, m, 2H), 7.81 (ArH, td, J = 7.12,1.52,
1.52 Hz, IH), 7.45-7.32 (ArH, m, 3H), 6.84 (NH, s, br, IH), 4.93-4.80 (CH2, m, IH), 4.55 10 (CH2, d, J = 12.97 Hz, IH), 4.38-4.25 (CH2, m, IH), 4.01-3.57 (CH2, m, 9H), 3.57-3.45 (CH2,
m, IH), 3.36 -3.32 (CH2, m, 2H), 1.42 (CH3,d, J = 6.78 Hz, 3H), 1.37-1.26(3 XCH3,m5
9H)
13CNMR(75 MHz, CDC13) 5ppm 165.36,162.85,161.36,159.98, 140.27,137.89, 134.92,
129.80, 124.03,121.31,119.64, 113.30,105.19,71.28,70.91, 67.25, 66.91, 52.89, 52.87, 15 44.42, 39.33, 31.60, 22.66, 16.60, 14.75 and 14.36.

WO 2008/023161

PCT/GB2007/003179

NMR data for Example 15c 5 'H NMR (300 MHz, CD3COCD3.S ppm 8.52 (ArH, s, 1H), 8.24 (ArH, d, J = 8.48 Hz, 1H), 7.91-7.80 (ArH, m, 2H), 7.62 (ArH, d, J = 8.47 Hz, 1H), 7.46 (ArH, t, J = 7.94, 7.94 Hz, 1H), 6A8 (NH, br, s, 1H), 4.84-4.70 (CH2, m, 1H), 4.53-4.33 (CH2, m, 3H), 4.09-3.79 (CH2, ma 5H), 3.80-3.56 (CH2, m, 5H), 3.49-3.40 (CH2," m, 1H), 3.23 - 3.28 (CH2, m, 1H), 2.20 (CH2, d, J = 6.66 Hz, 1H), 2,11-1.81 (CH2, m, 4H), 1.39 (CH3, d, J - 6.75 Hz, 3H), 1.26 (CH3, ds J
10 =6.75 Hz, 3H).
13CNMR(75 MHz, CD3COCD3) 5 ppm 171.66,160.37,147.37,145.71, 138.93,138.46, 135.69,128.98,126.26, 126.11,122.52, 121.58,118.91, 118.37, 104.46,77.93,70.31,70.13, 68.81, 66.28, 66.15, 51.77, 46.41, 43.85, 29.98, 25.06,14.39 and 13.92. Tested in Alternative Enzyme Assay: Ex. (15a) 0.0043uM; Ex. (15c) 0.33u.M.
15 Tested in phospho-Ser473 Akt assay: Ex. (15b) 0.5051uM.
Example 16 (Compound 16a)

The aminopyridine substrate was reported in Example 1.
Example lu (1 equiv) was dissolved in pyridine (0.11 M). Acetic anhydride (5.0 equiv) was added and the reaction mixture was heated at 70°C for 6 hours. Upon completion the sample 25 was concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired product.

WO 2008/023161

PCT/GB20H7/003179

Table 16:

NMR data for Example 16a
lHNMR (300MHz,CDCl3) 5 ppm 8.18-8.12 (ArH, m, 2H), 8.05 (ArH, d, J = 8.42 Hz, 1H), 5 7.52-7.40 (ArH, m, 3H), 4.96 (CH, d, br, J = 4.93 Hz, 1H), 4.66 (CH, d, br, / = 12.90 Hz, 1H), 4.40 (d, br, J = 6.71 Hz, 1H), 4.07-3.54 (CH2, m, 11H); 3.47-3.35 (CH, m, 1H), 1.51 (CH3, d, J = 6.79 Hz, 3H), 1.39 (CH3, d, J = 6.82 Hz, 3H)
l3CNMR (75 MHz, CDC13) 5ppm 165.41,162.93, 161.83,160.02, 137.14, 136.13, 134.84, 129.19,128.77,112.99,105.03,71.29,70.91,67.26,66.91,52.85,46.95,44.46,39.34,14.73 10 and 14.37.
Tested in Alternative Enzyme Assay: Ex. (16a) 0.034uM.
Example 17 15 (Compound 17a)



N
O...
R^ /*° O N N N ^'

The chloro-substrate was reported in Example 1.
The appropriate chloro-substrate (1 equiv) was dissolved in toluene (0.07 M). Phenol (1.0 20 equiv), palladium acetate (0.05 equiv), BINAP (0.05 equiv) and tripotassium phosphate (1.0

WO 2008/023161

PCT/GB2007/00317V

equiv) were added. The reaction vessel was sealed and exposed to microwave radiation (140 °C, medium absorption setting) for 10 minutes. Upon completion the samples was concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired product.

Tested in Alternative Enzyme Assay: Ex. (17a) 0.52uM.

Table 17:
To a mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (2.5 equiv), and the appropriate boronic acid (1.1 equiv) in acetonitrile/water (1:1) (0.033 M of chloro-
15 substrate) was added tetrakis(triphenylphosphine) palladium0 (0.05 equiv). The suspension was sonicated while degassed with nitrogen for 5 minutes then heated to 95 °C for 2 hours. Upon completion the reaction mixture was allowed to cool down to room temperature. The reaction mixture was concentrated in vacuo to half original volume. The crude residue was extracted with CH2CI2 and the combined organic phases were washed with brine, dried
20 (MgSO^), filtered and concentrated in vacuo to give a yellow solid. The residue was sonicated in diethyl ether, collected by vacuum filtration to give the desired product as a yellow powder.

WO 2008/02316J PCT/GB2007/003179



{5-[2-CMoro-4^(S)-3-methyl-morpholia-4-yl>pyrido[2,3-d]pyrimidin-7-yl]-2-methoxy-phenyl}-methanol: (78 % yield, 100 % purity) m/z (LC-MS, ESP): 401 [M+H]4 R/T = 3.47 min

{3-[2-Chloro-4-((S)-3-methyl-morpholiri-4-yl)-pyrido[2)3-d]pyrimidiii-7-yl]-phenyl}-methanol: (90 % yield, 90 % purity) m/z (LC-MS, ESP): 371 [M+H]+ R/T = 4.06 min
10 Alternatively, to a stirred mixture of bis(pinacolato)diboron (1.05 equiv) and potassium acetate (3 equiv) in N-methylpyrrolidine (13.5 equiv), purged with nitrogen, was added the corresponding bromobenzylalcohol (1 equiv) followed by PdCl2(dppf) (0.02 equiv). The mixture was then heated to 60 °C and held for 1 Omin, then heated to 70 °C and held for 15min and finally heated to 80 °C and held for Ih. The appropriate chloro-substrate (1 equiv) was
15 then added followed by PdCkCdppf) (0.02 equiv) and N-methylpyrrolidine (4.5 equiv). The temperature was then held at 75 °C, then 4.3M aqueous potassium carbonate (3.5 equiv) was added over 13min, then water (12 equiv) was added and the reaction was stirred at 75 °C for 90min. Water (144 equiv) was then added slowly over 70min with stirring while the temperature was reduced to 66 °C. The temperature of the stirred mixture was then kept at 64
20 °C for 30min, then cooled to 20 °C over 2.5b, and held at 20 °C overnight The resulting slurry was filtered, and the solid washed first with a 3:1 water:N-methylpyrrolidone mixture (18 equiv of water), then washed with water (24 equiv) and then washed with ethyl actetate (4 x 4.4equiv). The solid was then dried in a vacuum oven at 50 °C to leave the title compound

WO 2008/023161

PCT/GB2007/003179

in suitable clean form to be used without any further purification. For example, {5-[2-Chloro-4-((S)-3-methyl-morpholin-4-yI)-pyrido[2,3-d]pyrimidin-7-yl]"2-methoxy-phenyI}-methanol: (73% yield)

Conditions A:
The appropriate chloro-substrate (1 equiv) was dissolved in DMA (0.04 M). Tripotassium
10 phosphate (1.5 equiv) and the appropriate nucleophile (secondary amine) (1.5 equiv) were then added. The reaction vessel was sealed and the mixture exposed to microwave radiation (200 °C, medium absorption setting) for 30 minutes. Upon completion the samples were filtered through a silica cartridge, washed with EtOAc and then concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired products.
15 Conditions B:
The appropriate chloro-substrate (1 equiv) was suspended in a propan-2-ol and aqueous ammonia (1:3) solution (0.02 M). The reaction vessel was sealed and the mixture exposed to microwave radiation (140 °C, medium absorption setting) for 20 minutes. The crude residue was then purified by preparative HPLC to give the desired products.
20 Conditions C:
The appropriate chloro-substrate (1 equiv) was dissolved in dioxane (0.04 M). Diisopropylethylamine (5.0 equiv) and the appropriate nucleophile (secondary amine) (1.5 equiv) were then added. The reaction vessel was sealed and the mixture exposed to microwave radiation (130 °C, medium absorption setting) for 20 minutes. Upon completion
25 the samples were concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired products. Conditions D;
The appropriate chloro-substrate (1 equiv) was dissolved in dioxane (0.04 M). Tripotassium phosphate (3.0 equiv), xantphos (0.05 equiv), palladium acetate (0.05 equiv) and the

WO 20(18/023161

PCT/GB2007/003179

appropriate nucleophile (amine) (1.5 equiv) were then added. The reaction vessel was sealed and the mixture exposed to microwave radiation (150 °C, medium absorption setting) for 20 minutes. Upon completion the samples were filtered through a silica cartridge, washed with EtOAc and then concentrated in vacuo. The crude residue was then purified by preparative 5 HPLC to give the desired products. Conditions E:
The appropriate chloro-substrate (1.0 equiv) was dissolved in dioxane (0.04 M). Diisopropylethylarnine (5.0 equiv) and the appropriate nucleophile (secondary amine, with BOC-protected amino side chain) (1.5 equiv) were then added. The reaction vessel was sealed
10 and the mixture exposed to microwave radiation (130 DC. medium absorption setting) for 20 minutes. Upon completion the samples were concentrated in vacuo. To the crude residue was then added a 4 M solution of HC1 in dioxane (0.15 M). The reaction mixtures were stirred at room temperature for 3 hours. Upon completion the samples were basified with a 2 N sodium hydroxide solution. The crude residue was then purified by preparative HPLC to give the
15 desired products. Conditions F:
The appropriate nucleophile (substituted imidazole) (10.0 equiv) was dissolved in DMF (0. 4 M). Sodium hydride (5.0 equiv) was then added. The reaction mixture was stirred at room temperature for 10 minutes under nitrogen and a solution of the appropriate chloro-substrate
20 (1.0 equiv) in DMF (0.075 M) was added. The reaction vessel was sealed and the mixture exposed to microwave radiation (150 °C, medium absorption setting) for 30 minutes. Upon completion the samples were filtered through a silica cartridge, eluted with CH2CI2 and then concentrated in vacuo. The crude residue were then purified by preparative HPLC to give the desired products.
25 Conditions G;
The appropriate chloro-substrate (1 equiv) was dissolved in dioxane (0.04 M). Diisopropylethylarnine (5.0 equiv) and the appropriate nucleophile (secondary amine) (4.5 equiv) were then added. The reaction vessel was sealed and the mixture exposed to microwave radiation (130 °C, medium absorption setting) for 40 minutes. Upon completion
30 the samples were concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired products. Conditions H:

WO 2008/023161

PCT/GB2007/003179

The appropriate chloro-substrate (1 equiv) was dissolved in dioxane (0.04 M).
Diisopropylethylamine (5.0 equiv) and the appropriate nucleophile (secondary, amine) (10.0
equiv) were then added. The reaction vessel was sealed and the mixture exposed to
microwave radiation (130 °C, medium absorption setting) for 60 minutes. Upon completion 5 the samples were concentrated in vacuo. The crude residue was then purified by preparative
HPLC to give the desired products.
Conditions I:
The appropriate chloro-substrate (1 .equiv) was dissolved in a solution of 1 % DMA in
dioxane (0.04 M). Diisopropylethylamine (5.0 equiv) and the appropriate nucleophile 10 (secondary amine) (10.0 equiv) were then added. The reaction vessel was sealed and the
mixture exposed to microwave radiation (180 °C, medium absorption setting) for 60 minutes.
Upon completion the samples were concentrated in vacuo. The crude residue was then
purified by preparative HPLC to give the desired products.
Conditions J: 15 The appropriate chloro-substrate (1 equiv) was dissolved in a solution of 1 % DMA in
dioxane (0.04 M). Diisopropylethylamine (7.0 equiv) and the appropriate nucleophile
(secondary amine) (3.0 equiv) were then added. The reaction vessel was sealed and the
mixture exposed to microwave radiation (150 °C, medium absorption setting) for 60 minutes.
Upon completion tlie samples were concentrated in vacuo. The crude residue was then 20 purified by preparative HPLC to give the desired products.
Conditions IC:
The appropriate chloro-substrate (1 equiv) was dissolved in DMF (0.075 M). Potassium
carbonate (5.0 equiv) and the appropriate nucleophile (alcohol) (10.0 equiv) were then added.
The reaction vessel was sealed and the mixture exposed to microwave radiation (120 °C, 25 medium absorption setting) for 20 minutes. Upon completion the samples were concentrated
in vacuo. The crude residue was then purified by preparative HPLC to give the desired
products.
Conditions L:
The appropriate chloro-substrate (1 equiv) was dissolved in DMF (0.075 M). Potassium 30 carbonate (5.0 equiv) and the appropriate nucleophile (alcohol) (20.0 equiv) were men added.
The reaction vessel was sealed and the mixture exposed to microwave radiation (150 °C,
medium absorption setting) for 40 minutes. Upon completion the samples were concentrated

WO 2008/023161

PCT/GB2007/003179

in vacuo. The crude residue was then purified by preparative HPLC to give the desired products. Conditions M:
The appropriate chloro-substrate (1 equiv) was dissolved in DMA (0.13 M). 5 Diisopropylethylamrne (2.0 equiv) and the appropriate nucleophile (amine) (2.0 equiv) were then added. The reaction vessel was heated to 100 °C for 3 hours. Upon completion, the reaction mixture was partitioned between dichloromethane and water and the aqueous layer further extracted with dichloromethane. The combined organic phases were dried (MgSO*), filtered and the filtrate was concentrated in vacuo to give a yellow residue which was purified
10 by recrystallisation from diethyl ether. Conditions N:
542-Chloro^-((S)-3-memyl-morpholm^-yl)-pyrido[233"d]pyrirrridin-7-yl]-pyridin-2-ylamine (1 equiv) was dissolved in DMA (0.21 M). Diisopropylethylamrne (1.0 equiv) and the appropriate nucleophile (amine) (1.1 equiv) were then added. The reaction vessel was sealed
15 and the mixture exposed to microwave radiation (130 °C, medium absorption setting) for 10 minutes. Upon completion, the reaction mixture was partitioned between dichloromethane and water and the aqueous layer further extracted with CH2CI2. The combined organic phases were dried (MgS04), filtered and the filtrate was concentrated in vacuo to give a yellow residue which was purified by column chromatography on silica gel eluting with 0 % to 10 %
20 MeOH in CH2CI2 to give the desired product. Conditions O:
The appropriate chloro-substrate (1 equiv) was dissolved in DMA (0.16 M). Diisopropylethylamrne (1.0 equiv) and the appropriate nucleophile (amine) (1.2 equiv) were then added. The reaction vessel was heated to 80 °C for 48 hours. Upon completion, the
25 reaction mixture was partitioned between ethyl acetate and water and the organic layer washed with brine. The combined organic phases were dried (MgSC^), filtered and the filtrate was concentrated in vacuo to give a residue which was purified by preparative HPLC to give the desired product. Conditions P:
30 The appropriate chloro-substrate (1 equiv) was dissolved in anisole (0.25 M) (10 vol). Diisopropylethylamine (1.3 equiv) and the appropriate nucleophile (amine) (1.3 equiv) were then added. The reaction vessel was heated to 125 °C and stirred for 1 lh. Upon completion.

WO 2008/023161

PCT/GB2007/003179

the reaction mixture was allowed to cool to 50 °C, Aqueous 20% citric acid solution (7 vol) was added, stirred for 5min and then allowed to separate partitioned. The aqueous layer was removed and retained. The organic layer was then extracted with a further aliquot of aqueous 20 % citric acid solution (3 vol). The orgainic layer discarded, and the aqueous layers 5 combined. The combined aqueous layers were washed first with anisole (5 vol), then 50% aqueous sodium hydroxide solution (1.23 vol) was added slowly. The resulting aqueous phase was extracted with ethyl acetate (lOvol). The aqueous layer was discarded and the organic layer was washed first with 10% aqueous sodium hydroxide solution (5 vol) and then water (5 vol). The organic layer was then slurried with silicycle Si-thiourea scavenger at 50
10 °C for 2h, then the scavenger was filtered off and washed with ethyl actetate (2 x 1 vol). The organic phase was cooled to 20 °C3 seeded to start crystallization and stirred until a slurry obtained. The slurry was heated to 50 °C under vacuum and ethyl acetate (3 vol) was removed by vacuum distillation. 2-Methylpentane (3.4vol) was added and the mixture heated to 60 °C and then slowly cooled to 20 QC over 2h. The resulting slurry was filtered, and the
15 solid washed with 1:1 ethyl actetate:pentane (2 x 0.5vol). The solid was then dried in a vacuum oven at 50 °C to leave the desired product. For example, compound la was obtained (50.4% yield). The crude product (1 equiv) was dissolved in DMSO (5 vol based on product weight) at 50 °C. Water (2 vol) was added and the mixture stirred at 50 °C until product crystallizes. The slurry was heated to 60 °C and then water (3vol) was added slowly over
20 30min so that the temperature was maintained at 60 °C. The mixture was slowly cooled to 20 °C over 2h, and then held at 20 °C for 30min. The resulting slurry was filtered, and the solid washed with 2:1 watenDMSO (0.5:lvol), and then water (3 x 2vol). The solid was then dried in a vacuum oven at 50 °C to leave the desired product.
25

WO 2008/023161 PCT/GB2007/003179
Table 18:


WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161 PCT/GB2007/003179


WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/G03179





WO 2008/023161

PCT/CB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023361

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB20II7/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT7GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179

NMR data for Example 18b
'HNMR (300 MHz, CDC13) 5 ppm 8.85 (ArH, d, /= 2.46 Hz, IH), 8.64 (ArH, dd,J = 8.75, 2.48 Hz, IH), 8.30 (OH, s, br, IH), 8.04 (ArH, d, J = 8.47 Hz, IH), 7.59 (ArH, d, J= 8.54 Hz, IH), 7.14 (ArH, d, J= 8.83 Hz, IH), 5.03-4.91 (CH2, m, IH), 4.66 (CH2, dd,J = 13.05, 5 0.77 Hz, 1H),4.41 (CH2, d, J = 6.75 Hz, IH), 4.07 (OCH3,s,3H), 4.04-3.98 (CH2, m, IH), 3.97-3.68 (CH2, m, 1 IH), 3.60 (CH2s d, / = 2.75 Hz, IH), 3.41 (CH2, s, IH), 1.50 (CH3, d, J = 6.77 Hz, 3H), 1.39 (CH3, d,J= 6.81 Hz, 3H)
NMR data for Example 18k
10 'HNMR(300MHz, CHCI3) 8ppm 10.59-10.51 (OH, m, IH), 8.18 (ArH, dd, J = 4.42,2.17 Hz, 2H), 7.99 (ArH, d, J = 8.45 Hz, 1H)3 7.44 (ArH, d, J = 8.48 Hz, IH), 7.01 (ArH, d, J = 9.22 Hz, IH), 4.81 (CH2OH, s, 2H), 4.37-4.11 (CH2, m, 3H), 4.09-3.65 (OCH3 + CH2, m, 13H), 2.02-1.94 (CH2, m, IH), 1.73 - 1.38 (CH2, m, IH), 1.50 (CH3) d, J = 6.77 Hz, 3H) 13CNMR(75 MHz, CDC13) 5 ppm 162.05,161.84,161.81, 159.16,150.47,134.52,129.29,
15 128.68,128.43,127.47, 117.04,112.75,110.28,104.93,104.30, 70.96,67.12, 66.95, 66.77, 61.97, 55.57, 52.75, 50.99, 44.48 and 14.72.
NMR data for Example 18v
'HNMR (300 MHz, CDC13) 5ppm 8.07 (ArH, dd, J = 7.09,2.14 Hz, 2H), 7.89 (ArH, d, J =--20 8.47 Hz, 1H)S 7.33 (ArH, d, J = 8.49 Hz, IH), 6.91 (ArH, d,,/ = 9.31 Hz, 1H), 6.88 (NH, S,
Br, IH), 5.34 (NH, s, Br, IH), 4.95 (CH2, dd, J= 12.22, 0.66 Hz, 2H), 4.70 (CH2OH, s, 2H),
4.34-4.20 (CH, m, IH), 3.93-3.53 (OCH3, + CH2, m, 10H),2.91 (CH2,d, J= 12.29 Hz, 2H),
2.38 (CH2, s, 2H), 1.89 (CH2, dd, J = 6.92, 6.38 Hz, 2H), 1.76-1.54 (CH2, m, 3H), 1.38 (CH3,
d,J = 6.76Hz,3H) 25 13CNMR(75MHz,CDCl3)6ppm 176.91,165.39, 162.98, 161.80,160.14, 159.13,134.52,
131.20, 129.28,128.84,128.44,119.99,112.70,110.26,104.34, 70.97, 67.10,66.94, 61.97,
55.57, 52.76,44.52, 43.73, 43.69, 43.16, 26.88, and 14.70.
NMR data for Example 18ab 30 lH NMR(300 MHz, CDCI3) 8ppm 8.19 (ArH, d, J = 7.14 Hz, 2H), 8.00 (ArH, d, / = 8.47 Hz, IH), 7.43 (ArH, d, / = 8.42 Hz, IH), 7.01 (ArH, d, 7= 9.13 Hz, IH), 4.82 (CH2OH, s,

WO 2008/023161

PCT/GB2007/003179

2H), 4.71-4.59 (CH2, m, IH), 4.47-4.35 (CH2, m, IH), 3.97 (OCH3, s, 3H), 3.85 (CH25 ddd, J = 17.63,13.74, 9.24 Hz, 8H), 2.12 (CH2, s, Br, 5H), 1.50 (CH3d,/= 6.75 Hz, 3H). l3CNMR(75MHz,CDCl3)ppm 159.35,159.10,134.61, 131.26, 129-22,128.89,128.54, 112.41,110.21,104.39,71.06, 66.95, 61.99, 55.56, 52.80, 44.51, 27.01 and 14.78. 5
NMR data for Example 18ax
lH NMR (300 MHz, CDC13) 5 ppm 8.27-8.17 (ArH, m, 2H), 8.00 (ArH, d, J = 8.50 Hz, IH), 7.45 (ArH, d, J = 8.51 Hz, IH), 7.01 (ArH, d, J = 8.65 Hz, IH), 5.40 (NH, br, s, IH), 4.81 (CH2OH, s, 2H), 4.49-4.35 (CH2, m, IH), 3.97 (OCH3, s, 3H), 3.93-3.64 (CH2, rn, 6H), 3.58-
10 3.48 (CH2, m, 2H), 3.43 (OCH3, s, 3H), 1.49 (CH3, d, / = 6.71 Hz, 3H), 1.34 (CH3, d, J = 6.68 Hz, 3H)
l3CNMR(75MHz,CDCl3)5ppm 165.67,161.56,160.64, 159.19, 134.53,129.27,128.83, 128.39, 112.53,110.30, 76.23, 70.98, 67.00,62.02, 59.18, 55.57, 52.73,44.31,18.23, 18.20 and 14.85.
15
NMR data for Example 18bn
THNMR (300 MHz, CDC13) 5 ppm 8.23-8.15 (ArH, m, 2H), 7.99 (ArH, d, J - 8.45 Hz, IH), 7.42 (ArH, d, J= 8.46 Hz, IH), 7.00 (ArH, d, J= 8.35 Hz, IH), 4.81 (CH2OH, s, 2H), 4.65 (CH, s, br, IH), 4.05-3.64 (OCH3 + CH2, m, 13H), 3.24 (OH, s, IH), 1.50 (CH3, d, J = 6.73
20 Hz,3H).
l3CNMR(75MHz,CDCl3)Sppm 165.18,162.87, 159.34,159.06, 134.57, 131.25, 129.26, 128.84,128.47, 112.36,110.20,104.35,71.00,70.97, 66.94,61.91, 55.55, 52.82,44.43,27.01 and 14.87.
25 NMR data for Example 18bo
"HNMR(300 MHz, CDC13) 5 ppm 8.28-8.17 (ArH, m, 2H), 8.00 (ArH, d,J = 8.49 Hz, IH), 7.45 (ArH, d,J= 8.50 Hz, IH), 7.02 (ArH, d, J = 8.60 Hz, IH), 5.51-5.34 (CH, m, IH), 4.81 (CH2OH, s, 2H), 4.47-4.34 (CH, m, IH), 4.00 (CH2, d, J = 1.94 Hz, IH), 3.97 (OCH3, s, 3H), 3.93-3.89 (CH2, m, 2H), 3.83-3.63 (CH2, m, 4H), 3.53 (CH2, d,br, J = 4.02 Hz, 2H), 3.43
30 (OCH3; s, 3H), 1.50 (CH3, d, J = 6.73 Hz, 3H), 1.34 (CH3, d, J = 6.69 Hz, 3H)

WO 2008/023161

PCT/GB2007/003179

,3CNMR (75 MHz, CDCb) 5 ppm 165.69,161.55, 160.67,159.19,134.52, 131.15,129.26, 128.84, 128.41, 119.72,112.58,110.30, 70.98, 67.12, 67.00, 62.05, 59.18, 55.58, 52.73, 44.32,18.20 and 14.84.
NMR data for Example 18di
!HNMR (300 MHz, CDC13) 5ppm 8.10-8.03 (ArH, m, 2H), 7.95 (ArH, d, J = 8.41 Hz, IH), 7.42-7.30 (ArH, m, 3H), 5.52-5.27 (NH2, m, br,2H), 4.98 (CH2, dd, /= 12.74, 0.96 Hz, 2H), 4.31-4.29 (CH, m, IH), 3.97-3.55 (CH2, m, SH), 3.07-2.B6(CH2, m, 2H), 2.45-2.35 (CH2, m, 10 IH), 1.99-1.88 (CH2,m,br,2H), 1.70 (CH2,m,2H), 1.41 (CH3, d, J = 6.76 Hz, 3H)
,3CNMR(75MHz,CDCl3)Sppm 176.75, 170.03, 165.35,162.99, 161.12, 160.17, 137.17, 136.10,134.83,129.19,128.76,112.81,104.86,100.00,70.95,67.12, 66.91, 52.83, 44.50, 43.72, 43.68, 43.10, 28.88 and 14.73.
15 NMR data for Example 18dk
1HNMR(300MHz, CDCb 5ppm8.67 (ArH, d, J= 1.86 Hz, IH), 8.38 (ArH, dd, J= 8.76, 2.36 Hz, IH), 8.29 (NH, s, weak signal, IH), 7.91 (ArH, d, J - 8.45 Hz, IH), 7.27 (ArH, d, J = 8.46 Hz, IH), 6.58 (ArH, d, J = 8.75 Hz, IH), 5.54-5.45 (CH2, m, IH), 4.97 (NH2, br, s, 2H), 4.37-4.24 (CH2, m, IH), 3.97-3.54 (CH2, m, 6H), 3.09-2.87 (CH2, m, 2H), 2.77 (NHCH3,
20 d, J = 4.82 Hz, 3H), 2.42-2.24 (CH2,m, IH), 1.87 (CH2,d,t/-0.84Hz, 2H), 1.79-1.59 (CH2, m, 2H), 1.40 (CH3, d, J = 6.76 Hz, 3H).
NMR data for Example 18dl
lHNMR (300 MHz, CDCb 5 ppm 8.68 (ArH, d, J = 1.98 Hz, IH), 8.49 (ArH, dd, J = 8.87, 25 2.32 Hz, IH), 8.38 (NH, s, br,weak signal IH), 7.99 (ArH, d, J = 8.47 Hz, IH), 7.34 (ArH, d, J = 8.49 Hz, IH), 6.67 (ArH, d, / = 8.85 Hz, IH), 4.38 (CH2, d, J = 6.11 Hz, IH), 4.05-3.82 (CH2, m, 7H), 3.81-3.62 (CH2, m, 7H), 1.47 (CH3, d, J =6.11 Hz, 3H). 13C NMR (75 MHz, CDCI3) § ppm 165.16,162.79,160.31, 159.09, 158.45, 143.98, 139.12, 135.00,124.48,111.80,110.03,104.70, 70.92,67.00, 66.90, 52.81,44.57,44.40 and 14.78.

WO 2008/023161

PCT/GB2007/003179

NMR data for Example 18dm
,'HNMR (300 MHz, CDC13.5 ppm 8.67 (ArH, d, J = 2.05 Hz, IH), 8.36 (ArH, dd, J = 8.76, 2.27 Hz, IH), 7.90 (ArH, dd, J = 8.45, 2.12 Hz, IH), 7.26 (ArH, dd, J = 8.47, 0.73 Hz, IH), 6.57 (ArH, d, J= 8.76 Hz, IH), 5.10-4.87 (NH2, m, 2H), 4.37-4.22 (CH2, m, IH), 3.96-3.51 5 (CH2, m, 6H), 3.08 (NCH3 + CH2, s, 4H), 2.95-2.91 (NCH3, s, 3H), 2.80-2.59 (CH2, m, III), 1.76 (CH2, d, J= 2.61 Hz, 3H), 1.64-1.44 (CH, m, IH), 1.38 (CH3, t, ./ = 6.34, 6.34 Hz, 3H). 13CNMR(75MHz,CDCl3)5ppm 173.59, 165.46, 165.31,163.03,160.17, 158.89,145.99, 138.35, 134.76,124.89,111.71,109.15,104.41, 70.96,66.96, 52.81, 46.85,44.38, 39.43, 37.26, 35.56, 28.06, 24.95 and 14.71.
10
NMR data for Example 18dn
*H NMR (300 MHz, CDClj 5 ppm 8.10 (ArH, d, J= 7.89 Hz, 2H), 7.97 (ArH, d, J= 8.49 Hz, IH), 7.42 (ArH, d, J= 8.46 Hz, IH), 6.98 (ArH, d, J= 8.55 Hz, IH), 4.88 (CH2, d, J = 5.25 Hz, IH), 4.77 (CH2OH, s, 2H), 4.56 (CH2, d, J= 13.38 Hz, IH), 4.38 -4.36 (CH2, m, IH),
15 4.02 - 3.51 (OCH3 + CH2, m, 1 IH), 3.43 -3.33 (CH2, m, IH), 1.47 (CH3j d, ./ = 6.77 Hz, 3H), 1.35 (CH3, d, J = 6.78 Hz, 3H).
13CNMR(75 MHz, CD3COCD3)§ ppm 165.11, 162.27,161.87,159.54,159.23, 134.74,
130.76,129.41, 128.86,128.39,113.09, 110.32,104.45, 71.20, 70.95, 67.17, 66.91, 61.80,
55.57, 52.82, 47.05, 44.44, 39.45,14.74 and 14.44. 20
NMR data for Example 18do
'HNMR (300 MHz, CDC13 5ppm 8.10 (ArH, d, J= 8.76 Hz, 2H), 7.98 (ArH, d, J= 8.49 Hz,
IH), 7.42 (ArH, d,J= 8.46 Hz, IB), 6.97 (ArH, d, J= 8.37 Hz, IH), 4.88 (CH2, d, ./= 5.46
Hz, IH), 4.77 (CH2OH, s, 2H), 4.58 - 4.49 (CH2, m, IH), 4.39 - 4.36 (CH2, d J= 7.41 Hz, 25 IH), 4.02 - 3.51 (OCH3 + CH2, m, 11H), 3.43 - 3.33 (CH2, m, IH), 1.48 (CH3, d, y= 6.78
Hz, 3H), 1.35 (CH3, d, J- 6.78 Hz, 3H).
13CNMR(75 MHz, CD3COCD3) 5 ppm 165.05,161.87,159.45, 159.24, 134.78,130.70,
129.44,128.86,128.38, 113.14, 110.33,104.43, 71.19, 70.95, 67.16, 66.90,61.77, 55.57,
52.82,47.08,44.44,39.47,14.76 and 14.44. 30 Tested in Alternative Enzyme Assay: Ex. (18a) 0.03uM; Ex. (18b) Cl^M; Ex. (18c)
0.066jaM; Ex. (18d) 0.15pM; Ex. (18e) 0.039^M; Ex. (18f) 0.038^M; Ex. (18g) 0.031pM;
Ex. (18h) 0.23jaM; Ex. (18i) 0.03pM; Ex. (18j) 0.088uM; Ex. (18k) 0.019pM; Ex. (181)

WO 2008/023161

PCT/GB2007/003179

0.097^iM; Ex. (18m) 0.042JIM; Ex. (18n) 0.3i^iM; Ex. (18o) 0.51|iM; Ex. (18p) 0.25^iM; Ex.
(18q) O.II^LM; EX. (18r) 0AZ\xU; Ex. (18s) 0.037jiM; Ex. (18t) 0.054^M; Ex. (18u)
0.073^iM; Ex. (18v) O.OH^M; Ex. (18w) 025\M; Ex. (18x) O.OH^iM; Ex. (18y) 0.023 ^M;
Ex. (18z) 0.088nM; Ex. (18aa) 0.019^M; Ex. (18ab) 0.012^iM; Ex. (18ac) 0.014|iM; Ex. 5 (18ad) 0.078^iM; Ex. (18ae) 0.034|iM; Ex. (18af) 0.23^M; Ex. (18ag) 0.25nM; Ex. (18ah)
0.03^M; Ex. (18ai) 0.063^M; Ex. (18aj) 0.022|iM; Ex. (18ak) 0A2\iM; Ex. (18al) 0.36^M;
Ex. (18am) 0.077(JM; EX. (18an) 0.14^iM; Ex. (18ao) 0.073nM; Ex. (18ap) 0.013|aM; Ex.
(18aq) 0.l9\xU; Ex. (I8ar) G.079nM; Ex. (18as) 0.08^M; Ex. (18at) 0.78nM; Ex. (18au)
0.1 ^M; Ex. (18av) 0.27|iM; Ex. (18aw) 0.058^M; Ex. (18ax) 0.026^M; Ex. (18ay) 10 0.087^M; Ex. (18az) 0.092^iM; Ex. (18ba) 0.16|aM; Ex. (18bb) 0.65^iM; Ex. (18bc)
0.043^iM; Ex. (18bd) 0.19^iM; Ex. (18be) 0.79|aM; EX. (18bf) 0.077(iM; Ex. (18bg)
0.047nM; Ex. (18bh) 0.04^M; Ex. (18bi) 0.32^M; Ex. (18bj) 0.024^M; Ex. (18bk) 0.022^M;
Ex. (18bl) 0.61^M; Ex. (18bm) 0.025nM; Ex. (lSbn) O.Ol^M; Ex. (18bo) 0.058^iM; Ex.
(18bp) 0.049fiM; Ex. (18bq) 0.072piM; Ex. (18br) 0.03nM; Ex. 08bs) 0.042^M; Ex. (18bt) 15 0.062^M; Ex. (18bu) 0.047^M; Ex. (18bv) 0.11(iM; Ex. (18bw) O.031nM; Ex. (18bx)
0.035jxM; Ex. (18by) 0.039[iM; Ex. (18bz) O.OljiM; Ex. (18ca) 0.0026^M; Ex. (18cb)
0.25nM; Ex. (18cc) O.OIS^M; Ex. (18cd) 0.025^M; Ex. (18ce) 031\xU; Ex. (18cf) 0.013jaM;
Ex. (18cg) 0.067^M; Ex. (18ch) 0.078jaM; Ex. (ISci) 0.068^M; Ex. (18cj) 0.055^M; Ex.
(18ck) 0.0095jiM; Ex. (18cl) 0.023^iM; Ex. (18cm) O.029^iM; Ex. (18cn) 0.013^iM; Ex. 20 (18co) 0.0052jiM; Ex. (IScp) 0.0057^iM; Ex. (18cq) 0.027^M; Ex. (18cr) 0.0063jaM; Ex.
(18cs) 0.0047^M; Ex. (18ct) 0.097(iM; Ex. (18cu) 0.08^M; Ex. (18cv) 0.043nM; Ex. (18cw)
0.034^iM; Ex. (18cx) 0.024^iM; Ex. (18cy) 0.12|iM; Ex. (18cz) 0.079JJM; EX. (18da) 0.71^iM;
Ex. (18db) 0.0031pM; Ex. (18dc) 0.2^M; Ex. (18dd) O.028^M; Ex. (18de) 0.26^M; Ex.
(18df) 0.4^M; Ex. (18dg) 0.3^M; Ex. (18dh) 0.15nM; Ex. (18di) 0.15JIM; Ex. (18dj) 25 0.052JIM; Ex. (18dm) 0.061(aM; Ex. (18dn) 0.0094|iM; Ex. (18do) 0.026 ^iM.Tested in
phospho-Ser473 Akt assay: Ex. (18dk) 0.6821^M; Ex. (18dl) 0.2951pM.

WO 2008/023161

PCT/GB2007/003179


5 (Compounds 19a to 19x)
Conditions A:
To a mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (3.5 equiv), and the appropriate boronic acid (1.0 equiv) in acetonitrile/water (1:1) (0.026 M of chloro-substrate) was added tetrakis(triphenylphosphine) palladium0 (0.05 equiv). The reaction 10 vessel was sealed and heated at 95 °C for 2 hours. Upon completion the samples were filtered through a silica cartridge, washed with CH2CI2 and methanol and then concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired products.
Conditions B:
15 To a mixture of the appropriate chloro-substrate (1 equiv), cesium fluoride (3.5 equiv), and the appropriate boronic acid (1.0 equiv) in acetonitrile (0.026 M of chloro-substrate) was added tetrakis(triphenylphosphine) palladium0 (0.05 equiv). The reaction vessel was sealed and heated at 95 °C for 2 hours. Upon completion the samples were filtered through a silica cartridge, washed with CH2CI2 and methanol and then concentrated in vacuo. The crude
20 residue was then purified by preparative HPLC to give the desired products.
Conditions C:
To a mixture of the appropriate chloro-substrate (1 equiv), potassium carbonate (2.5 equiv), and the appropriate pinacolate boron ester or boronic acid (1.1 equiv) in acetonitrile/water 25 (1:1) (0.041 M of chloro-substrate) was added tetrakis(triphenylphosphine) palladium0 (0.05 equiv). The reaction vessel was sealed and exposed to microwave radiation (150 °C, medium absorption setting) for 30 minutes under nitrogen atmosphere, Upon completion the samples were concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired products.

WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/00317*)





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179





WO 2008/023161 PCT/GB2007/003179

NMR data for Example 19i
lH NMR (300 MHz, DMSO) S ppm 9.63 (ArH, d, J = 1.49 Hz, IH), 8.84-8.69 (ArH, m, 2H), 8.49-8.37 (ArH, m, IH), 8.19 (ArH, dd, J = 8.61, 2.35 Hz, lH), 8.00 (ArH, d, J= 8.76 Hz, 5 IH), 7.57 (ArH, ddd, J = 7.99,4.81, 0.71 Hz, IH), 7.15 (ArH, d, / = 8.71 Hz, 1H), 5.23 (ArH, dd, J = 2.03,1.13 Hz, 1H),5.23 (CH,m, lH),4.78(CH>d, J= 6.83 Hz, 1H),4.61 (CH2OH, s, 2H), 4.22 (CH2, d, J - 13.08 Hz, IH), 4.03-3.92 (CH2, m, IH), 3.98 (OCH3, s, 3H), 3.88-3.61 (CH2, m, 3H), 2.50 1.49 (CH3, d, J= 6.79 Hz, 3H)
l3C NMR (75 MHz, DMSO) 5 ppm 164.91,161.77,161.25, 160.36, 158.71, 151.75, 149.97, 10 136.10,133.86,131.45, 129.97, 127.92,126.79, 124.08, 117.24, 110.92,108.15, 70.77, 66.83, 66.80, 58.39, 56.04, 52.15, 44.39 and 15.24.
NMR data for Example 19x
]H NMR(300 MHz, CDCI3) 5ppm 9,86-9.80 (ArH, m, IH), 9.00-8.91 (ArH, m, IH), 8.77
15 (ArH, dd, J= 4.80,1.71 Hz, 3H), 8.28 (ArH , ddd, J = 9.24, 8.03, 5.57 Hz, IH), 7.83 (ArH, d, J = 8.64 Hz, 2H), 7.60-7.53 (ArH, m, 2H), 7.53-7.43 (CH, m, IH), 4.72 (CH2, d, J = 6.93 Hz, IH), 4.33-4.23 (CH2, m, IH), 4.00-3.80 (CH2, m, 4H), 1.65 (CH3, d, J= 6.81 Hz, 3H) 13CNMR(75MHz,CDCl3)5ppm 164.55, 161.73,161.53,151.52, 150.66,136.99,136.34, 134.91,133.42,129.34, 129.12, 123.19,119.66,117.15,108.64,106.49, 70.98, 67.02, 52.92,
20 44.49 and 15.16.
Tested in Alternative Enzyme Assay: Ex. (19a) 0.048uM; Ex. (19b) 0.018^M; Ex. (19c) 0.052uM; Ex. (19d) 0.25^M; Ex. (I9e) 0.1 luM; Ex. (19f) 0.096uM; Ex. (19g) 0.0087jiM; Ex. (19h) 0.77uM; Ex. (19i) 0.28uM; Ex. (19j) 0.057uM; Ex. (19k) 0.077^M; Ex. (191)

WO 2008/023161

PCT/GB2007/003179

0.12uM; Ex. (19m) 0.41uM; Ex. (19n) 0.22uM; Ex. (19o) 0.19jiM; Ex. (19p) 0.24uM; Ex. (19q) 0.14uM; Ex. (19r) 0.012uM; Ex. (19s) 2uM; Ex. (19t) 0.097uM; Ex. (19u) 0.055uM; Ex. (19V) 0.07uM; £x. (19w) 0.086uM; Ex. (19x) 0.81uM.
5 Example 20

The amino substrate was reported in Example 18.
(Compounds 20a to 20c)
10 Conditions A:
The appropriate amino-substrate (1 equiv) was suspended in THF (0.04 M). The appropriate sulfonyl chloride (2-0 equiv) was added. The reaction vessel was sealed and exposed to microwave radiation (140 °C, medium absorption setting) for 10 minutes. Upon completion the samples were concentrated in vacuo. The crude residue was then purified by preparative
15 HPLC to give the desired products. Conditions B:
The appropriate amino-substrate (1 equiv) was suspended in DMF (0.04 M). The appropriate acyl chloride (1.2 ecjuiv) and potassium carbonate (2.4 equiv) were added. The reaction vessel was sealed and exposed to microwave radiation (140 ^C, medium absorption setting) for 10
20 minutes. Upon completion the samples were concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired products. Conditions C:
The appropriate amino-substrate (1 equiv) was suspended in DMF (0.09 M). The appropriate acyl chloride (3.0 equiv) was added. The reaction vessel was sealed and exposed to
25 microwave radiation (130 °C medium absorption setting) for 15 minutes. Upon completion the samples were concentrated in vacuo. The crude residue was then purified by preparative HPLC to give the desired products.
]

WO 2008/023161

PCT/GB2007/003179


Table 20:
Tested in Alternative Enzyme Assay: Ex. (20a) 1.4|xM; Ex. (20b) 0.67jxM; Ex. (20c) 5 0.024^M.

The chloro-substrate was reported in Example 18.
Example 21


WO 2008/023161

PCT/GB2007/003179

(Compound 21a)
The appropriate chloro-substrate (1 equiv) was dissolved in ethanol (0.025 M). Sodium formate (11.0 equiv) and palladium on carbon (0.5 equiv) were added. The reaction vessel was sealed and heated at 100 °C for 12 hours. Upon completion the sample was filtered 5 tlirough Celite™, and the filtrate was concentrated in vacuo. The crude residue was then purified by reverse phase chromatography eluting with a gradient of 5 to 95 % acetonitrile in 0.1 % formic acid/water, to give the desired product.
Table 21:

10
NMR data for Example 21a
lH NMR (300 MHz, DMSO) 5 ppm 8.70 (ArH, s, 1H), 8.42 - 8.37 (ArH, m, 2H), 8.16 (ArH, dd,J = 8.59, 2.34 Hz, 1H), 8.01 (ArH, d, J= 8.79 Hz, 1H), 7.14 (ArH, d, J =8.69 Hz, 1H), 5.20 (CH, l,J= 5.67, 5.67 Hz, 1H), 4.59 (CH2, d,J = 5.61 Hz, 2H), 4.05 - 3.93 (CH2, m, 2H),
15 3.89 (OCH3, s, 3H), 3.80-3.59 (CH2, m, 4H), 3.57 (s, 1H), 3.31 (s, 1H), 2.50 (td, J = 3.64, 1.80, 1.80 Hz, 1H), 1.42 (CH3, d, J= 6.79 Hz, 3H)
I3CNMR(75 MHz, DMSO) 5ppm 164.04,161.48,160.52,158.69, 157.38, 136.11,131.43, 129.96, 127.87, 126.77, 117.33, 110.93,109-11, 70.71, 66.83, 58.37, 56.03, 52.14,44.28 and 15.17.
20
Tested in Alternative Enzyme Assay: Ex. (21a) 0.2uM.

WO 2008/023161

PCT/GB2007/003179


Using the method of Example 1, to a cooled (0-5 °C) stirred solution (0.1 M) of the 5 appropriate trichloro substrate (1 equiv) in CH2CI2 was added diisopropylethylamine (1 equiv) in a drop wise fashion. The appropriate amine (1 equiv) was then added to the reaction mixture portionwise over the period of 1 hour. The solution was maintained at room temperature with stirring for a further 1 hour before the mixture was washed with water (2x1 reaction volume). The aqueous extracts were combined and extracted with CH2CI2 (2x1 reaction volume). The 10 organic extracts were then combined, dried (sodium sulphate), filtered and concentrated in vacuo to give an oily residue which solidified upon prolonged drying. The solid was triturated with diethylether and then filtered and the cake washed with cold diethyl ether to leave the title compound in a suitably clean form to be used without any further purification.

15 2J7-Dichioro-4-morpholin-4-yl-pyrido[2,3-d]pyrimidine - Rl= morpholine: (92% yield, 90% purity) m/z (LC-MS, ESP): 285 [M+H]+ R/T = 3.90 min

To a solution (0.2 M) of the appropriate dichloro-substrate (1 equiv) in anhydrous dimethyl acetamide under an inert atmosphere was added diisopropylethylamine (1 equiv) followed by 20 the appropriate amine (1 equiv). The resulting mixture was heated for 48 hours at 70 °C before being cooled to ambient temperature. The reaction was diluted with CH2CI2 (1 reaction volume) and then washed with water (3x1 reaction volumes). The organic extract

WO 2008/023161

PCT/GB2007/003179

was concentrated in vacuo to give a syrup which was dissolved in EtOAC (1 reaction volume) and washed with saturated brine solution before being dried, filtered (sodium sulphate) and concentrated in vacuo to give an oil. The crude residue was purified by flash chromatography (SiC>2, eluted with EtOAc:Hex (7:3) going to (1:1)) to give the title compound as a yellow 5 solid that was suitably clean to be used without any further purification.



7-Chloro-2-((2S,6R)-2,6-dimethyI-morpholin-4-yi)-4-morpholin-4-yI-pyrido[2,3-djpyrimidine - Rl= morpholine, R2=cis-dimethylmorpholine: (42% yield, 100% purity) tn/z
n r.A/TQ PSPV -X6A [\A+1f\+ TJ/T = 0 Qfi m,"n



7-Crdoro-2-((S)^-methyl-morpholin-4-yI)-4-morpholin-4-yl-pyrido[2,3-d]pyrimidine Rl = morpholine, R2= (S)-3-Methyl-morpholine: (70 % yield, 97 % purity) m/z (LC-MS, ESP): 350 [M+H]+R/T = 3.44 rain


CI
15

7-Chloro-2-(2-ethy]-piperidin-l-yl)-4-morpholin-4-yl-pyrido[2,3-d]pyrimidineRl = morpholine, R2= 2-Ethyl-piperidine: (56 % yield, 95 % purity) m/z (LC-MS, ESP): 362 [M+H]+R/T = 3.78 min

WO 2008/023161

PCT/GB2007/003179

Comparative Examples la, lb, lc, lj, and Ik
R4 = morpholine
R2 - (S)-3-methyl-morpholine or cis-dimethylmorpholine or 2-EthyI-piperidine
R7 = aryl or heteros

5
The appropriate chloro-substrate (1 equiv) was dissolved in a toluene/ethanol (1:1) solution
(0.02 M). Sodium carbonate (2 equiv) and the appropriate pinacolate boron ester or boronic
acid (1 equiv) were then added followed by tetrakis(uiphenylphosphine) palladium0 (0.1
equiv). The reaction vessel was sealed and the mixture exposed to microwave radiation (140
10 °C, medium absorption setting) for 30 minutes. Upon completion the samples were filtered
through a silica cartridge, washed with EtOAc and then concentrated in vacuo. The crude
residue was then purified by preparative HPLC to give the desired comparative examples.
The following Comparative Examples were prepared
15


WO 2008/023161

PCT/GB2007/003179


Example 22 Biological Assay
For mTOR enzyme activity assays, mTOR protein was isolated from HeLa cell 5 cytoplasmic extract by immunoprecipitation, and activity determined essentially as described previously using recombinant PHAS-1 as a substrate (ref. 21).
Examples la - 11, lak, lal, lap, lat, laz, 31,4a, 4c, 4d, 4f, 4i, 4w, 4x, 5q were tested
and exhibited IC50 values against mTOR of less than 200 nM. For example 5q was measured
to have an IC50 of 46nm.
10 The comparative Examples were also tested and when compared to the corresponding
Examples, the exhibited IC50 values for the Comparative Examples were higher than those of the corresponding Examples (ie IC50 Comparative Example 1 a > IC50 Example 1 a). For example Example Ik was measured to have an IC50 of 5nm whereas Comparative Example

WO 2008/023161

PCT/GB2007/003179

Ik was measured to have an IC50 of 33nm. Therefore, compounds of the present invention are more active in the mTOR assay.
Example 23 5 Alternative Enzyme Assay
The assay used AlphaScreen technology (Gray et ai, Analytical Biochemistry, 2003, 313: 234-245) to determine the ability of test compounds to inhibit phosphorylation by recombinant mTOR.
A C-terminal truncation of mTOR encompassing amino acid residues 1362 to 2549 of
10 mTOR (EMBL Accession No. L34075) was stably expressed as a FLAG-tagged fusion in HEK293 cells as described by Vilella-Bach et al„ Journal of Biochemistry, 1999, 274, 4266-4272. The HEK293 FLAG-tagged mTOR (1362-2549) stable cell line was routinely maintained at 37°C with 5 % CO2 up to a confluency of 70-90 % in Dulbecco's modified Eagle's growth medium (DMEM; Invitrogen Limited, Paisley, UK Catalogue No. 41966-029)
15 containing 10 % heat-inactivated foetal calf serum (FCS; Sigma, Poole, Dorset, UK, Catalogue No. F0392), 1 % L-glutamine (Gibco, Catalogue No. 25030-024) and 2 mg/ml Geneticin (G418 sulphate; Invitrogen Limited, UK Catalogue No. 10131-027). Following expression in the mammalian HEK293 cell line, expressed protein was purified using the FLAG epitope tag using standard purification techniques.
20 Test compounds-were prepared as 10 mM stock solutions in DMSO and diluted into
water as required to give a range of final assay concentrations. Aliquots (2 |xl) of each compound dilution were placed into a well of a Greiner 384-well low volume (LV) white polystyrene plate (Greiner Bio-one). A 10 pi mixture of recombinant purified mTOR enzyme, 1 uJvl biotinylated peptide substrate (Biotin-Ahx-Lys-Lys-Ala-Asn-Gln-Val-Phe-
25 Leu-Gly-Phe-Thr-Tyr-Val-Ala-Pro-Ser-Val-Leu-Glu-Ser-Val-Lys-Glu-NH2; Bachem UK Ltd), ATP (20 pM) in a buffer solution [comprising Tris-HCl pH7.4 buffer (50 mM), EGTA (0.1 mM), bovine serum albumin (0.5 mg/ml), DTT (1.25 mM) and manganese chloride (10 mM)] were added to the assay plates and incubated with compound for 2 hours at room temperature.
30 Each reaction was stopped by the addition of 5 pi of a mixture of EDTA (50 mM),
bovine serum albumin (BSA; 0.5 mg/ml) and Tris-HCl pH7.4 buffer (50 mM) containing p70 S6 Kinase (T389) 1A5 Monoclonal Antibody (Cell Signalling Technology, Catalogue No.

WO 2008/023161

PCT/GB2007/003179

9206B) and AlphaScreen Streptavidin donor and Protein A acceptor beads (200ng/well Perkin Elmer, Catalogue No. 6760002B and 6760137R respectively). Assay plates were left for approx 16 hours at room temperature before measurement. The resultant signals arising from laser light excitation at 680 nm were measured using a Packard Envision instrument. 5 Phosphorylated biotinylated peptide is formed in situ as a result of mTOR mediated phosphorylation. The phosphorylated biotinylated peptide that is associated with AlphaScreen Streptavidin donor beads forms a complex with the p70 S6 Kinase (T389) 1A5 Monoclonal Antibody that is associated with Alphascreen Protein A acceptor beads. Upon laser light excitation at 680 nm, the donor bead : acceptor bead complex produces a signal that
10 can be measured. Accordingly, the presence of mTOR kinase activity results in an assay signal. In the presence of an mTOR kinase inhibitor, signal strength is reduced.
Control wells that produced a maximum signal corresponding to maximum enzyme activity were created by using 5 % DMSO instead of test compound. Control wells that produced a minimum signal corresponding to fully inhibited enzyme were created "by adding
15 EDTA (83 mM) instead of test compound.
mTOR enzyme inhibition for a given test compound was expressed as an IC50 value. The compounds tested in this assay exhibited IC50 values against mTOR of less than 40pjn.
20 The following compounds exhibited IC50 values against mTOR of less than l^m: lbp, lea, leb, led, 12e, 18df, lm, Iq, Ir, 17,19h, 19m, 18n, 18o, 18ak, 18al, 18at, It, 18bb, 18be, 18bL 18bl, lx, ly, lba, lz, 20b, lae, 7a, 7h, 18ce, 5f, 4af, 4ag, 4aj, 5y, 3b, 5j, 5k, 5p, 3w, 3y, 3z, 1 la, 18da, 3m, 3o, 3p, 3r, 3s, laj, 5r, 5s, len, 2a, 2b, Icq, lcr, 2d, 3ad, 2h, lew and ldd, with the following compounds exhibiting IC50 values against mTOR of less than 300niM: 1c, lbq,
25 lbt, lch, lei, 4ap, 4at, 4aw, 4ax, 4ay, 4bd, 12b, 18de, 18dh, l8di, 18dg, 21a, lo, 18b, I8d, 18h, 19d, 19e, 19i, 191,19n, 19o, 18p, 18q, 18r, 18w, 18af, I8ag, 18an, 18aq, 18au, 18av, lv, 18ay, 18ba, 18bd, lbg, lw, lac, 4p, 9a, lbb, lav, 7b, 7e, 7f, 7g, 7k, 7j= 5c, 5d, 5e, 5g, 4v, 4x, 4y, 4z, 4aa, 4ae, 4ah, 4ai, 5u, 5v, 5w, 5x, 3d, 3f, 18bv, 18cb, 3h, 5h, 5i, 51, 5o, 3i, 3j, 3v, 3x, 3u, 3ab, lal, lam, Ian, lbe, 18cy, 18dc, 13a, 19p, 19q, 3k, 3n, 3q, 13f, 13b, 4g, lau, 5q, lay,
30 18dj, 13c, 13e, 10a, lcl, 2c, 2e, les, 2i, 8d, 13g and leu, with the following compounds
exhibiting IC50 values against mTOR of less than lOOnM: lb, la, Id, lbl, lbm, lbn, If. lbo, li, lg, lh, Ibr, lbs, lbu, lbv, le, lj, Ibw, lbx, lby, lbz, lec, Ice, Ik, lef, leg, 11, lcj, 4al,

WO 2008/023161

PCT/GB2007/003179

4am, 4an, 4ao, 4aq, 4ar, 4as, 4au, 4av, 4az, 4ba, 4bb, 4bc, 4be, 4bf, 12c, 12d, 12a,18a, 6a, las, lax, In, lp, Is, Ick, 18c, 18e, 18f, 18g, 18i, 18j, 18k, lar, 19a, 19b, 19c, 19f, 19g, 19j, 19k, 181,18m, lbd, laq, 18s, 18t, 18u, 18v, 18x, 18y, 18z, 18aa, 18ab, 18ac, 18ad, 18ae, 18ah, I8ai, 18aj, 18am, 18ao, 18ap, 18ar, 18as, 18aw, 18ax, 18az, 18bc, 18bf, 18bg, 18bk, 5 18bh, 18bj, 15a, 18bm, 8b, 4h, 14a, 8a, laa, lab, lad, laf, lag, 14b, lbc, 4i, lah, 4j, 41,4m, 4n, 4o, 18bn, 18bo, 4u, lbh, 16a, lat, 7c, 7d, 7i, 3a, 3c, 5a, 5b, 4w, 4ac, 4ad, 5t, 3e, 3gs 18bp, 18bq, 18br, 18bs, 18W, 18bu, 18bw, 18by, 18bz, 18ca, 18cc, 18cd, 18cf, 18cg, 18ch, 18ci, 18cj, 18ck, 18cl,4ak, 18bx, 18cm, 18cv, lbi, lbj, 4a, law, 3t, 3aa, lap, Ibf, 18cn, 18co, 18cp, 18cs, 18ct, 18cu, 18cw, 18cx, 18cz, 18cq, 19r, 19t, 31, 19u, 19v, 19w, 20c, lu, 4b, 4q, 4t, 4c,.
10 4e, 4f, 18dd, 4d, laz, 4r, 4s, 2f, 2g, 2j and lev. For example, Compound 4aa has an IC50 of 151nM.
The Comparative Examples were also tested and when compared to the corresponding Examples, the exhibited IC30 values for the Comparative Examples were higher than those of the corresponding Examples. For example Example Ik was measured to have an IC50 of
15 15nm whereas Comparative Example Ik was measured to have an IC50 of 225nm, Therefore, compounds of the present invention are more active in reducing cell growth.
Example 24
Cell proliferation assay (GISo)
20 Cell growth was assessed using the sulforhodamine B (SRB) assay (A). T47D
(ECACC, 85102201) cells were routinely passaged in RPMI (Invitrogen, 42401018) plus 10 % foetal calf serum (FCS), 1 % L-glutamine (Gibco BRL, 25030) to a confluence not greater than 80 %. To undertake the assay, T47D cells were seeded at 2.5xl03 cells/ well in 90ul RPMT plus 10 % foetal calf serum, 1 % L-glutamine in 96 well plates (Costar, 3904) and
25 incubated at 37°C (+5 % CO2) in a humidified incubator. Once the cells had fully adhered (typically following 4-5 hours incubation) the plate was removed from the incubator and lOuL of the diluent added to the control wells (Al-12 and Bl-12). Compound was prepared in a six point semi-log dilution at lOx the final concentration required e.g. for a 6 point range of 30jiM to lOOnM in semi-log steps dilution started at 300uM in stock plate. Dosing was
30 completed by addition of lOuL of compound at highest concentration to Cl-12 through to the lowest concentration in Hl-12. The plates were then incubated for 120 hours prior to SRB analysis.

WO 2008/023161

PCT/GB2007/003179

Upon completion of incubation, media was removed and the cells fixed with 100u.l of ice cold 10 % (w/v) trichloroacetic acid. The plates were incubated at 4QC for 20 minutes and then washed four times with water. Each well of cells was then stained with lOOul of 0.4 % (w/v) SRB (Sulforhodamine B, Sigma, Poole, Dorset, UK, Catalogue number S-9012) in 1 5 % acetic acid for 20 minutes before washing four times with 1 % acetic acid. Plates were then dried for 2 hours at room temperature. The dye from the stained cells was solubilized by the addition of lOOui of lOmM Tris Base into each well. Plates were gently shaken and left at room temperature for 30 minutes before measuring the optical density at 564nM on a Microquant microtiter plate reader. The concentration of inhibitor eliciting a 50 % reduction 10 in growth (GI50) was determined by analysis of staining intensity of the treated cells as a percentage of the vehicle control wells using Excelfit software.
(A) Skehan, P., Stoning, R., Scudiero, R, Monks, A., McMahon, J., Vistica, D., Warren, J.
T., Bokesch, H., Kenny, S. and Boyd, M. R. (1990) New colorimetric cytotoxicity assay for 15 anticancer-drug screening. J. Natl. Cancer Inst. 82, 1107-1112.
Examples la-11 were tested and exhibited GI50 values of less than 300 nM.
The Comparative Examples were also tested and when compared to the corresponding
Examples, the exhibited GI50 values for the Comparative Examples were higher than those of
the corresponding Examples (ie GI50 Comparative Example la > GI50 Example la). For 20 example Example Ik was measured to have an GI50 of 32nm whereas Comparative Example
1 k was measured to have an GI50 of 268nm. Therefore, compounds of the present invention
are more active in reducing cell growth.
Example 25 25 In Vitro phospho-Ser473 Akt assay
This assay determines the ability of test compounds to inhibit phosphorylation of
Serine 473 in Akt as assessed using Acumen Explorer technology (Acumen Bioscience
Limited), a plate reader that can be used to rapidly quantitate features of images generated by
laser-scanning.
30 A MDA-MB-468 human breast adenocarcinoma cell line (LGC Promochem,
Teddington, Middlesex, UK, Catalogue No. HTB-132) was routinely maintained at 37°C with 5 % C02 up to a confluency of 70-90 % in DMEM containing 10 % heat-inactivated PCS and 1 % L-glutamine.

WO 2008/023161

PCT/GB200 7/003179

For the assay, the cells were detached from the culture flask using 'Accutase' (Innovative Cell Technologies Inc., San Diego, CA, USA; Catalogue No. AT104) using standard tissue culture methods and resuspended in media to give 1.7x10s cells per ml. Aliquots (90 (il) were seeded into each of the inner 60 wells of a black Packard 96 well plate 5 (PerkinElmer, Boston, MA, USA; Catalogue No. 6005182) to give a density of -15000 cells per well. Aliquots (90 |il) of culture media were placed in the outer wells to prevent edge effects. The cells were incubated overnight at 37°C with 5 % CO2 to allow them to adhere.
On day 2, the cells were treated with test compounds and incubated for 2 hours at 37°C with 5 % CO2. Test compounds were prepared as 10 mM stock solutions in DMSO and
10 serially diluted as required with growth media to give a range of concentrations that were 10-fold the required final test concentrations. Aliquots (10 \i\) of each compound dilution were placed in a well (in triplicate) to give the final required concentrations. As a minimum reponse control, each plate contained wells having a final concentration of 100 uM LY294002 (Calbiochem, Beeston, UK, Catalogue No. 440202). As a maximum response control, wells
15 contained 1 % DMSO instead of test compound. Following incubation, the contents of the plates were fixed by treatment with a 1.6 % aqueous formaldehyde solution (Sigma, Poole, Dorset, UK, Catalogue No. F1635) at room temperature for 1 hour.
All subsequent aspiration and wash steps were carried out using a Tecan 96 well plate washer (aspiration speed 10 mm/sec). The fixing solution was removed and the contents
20 of the plates were washed with phosphate-buffered saline (PBS; 50 [il; Gibco, Catalogue No. 10010015). The contents of the plates were treated for 10 minutes at room temperature with an aliquot (50 jal) of a cell permeabilisation buffer consisting of a mixture of PBS and 0.5 % Tween-20. The 'permeabilisation' buffer was removed and non-specific binding sites were blocked by treatment for 1 hour at room temperature of an aliquot (50 ul) of a blocking
25 buffer consisting of 5 % dried skimmed milk ['Marvel' (registered trade mark); Premier Beverages, Stafford, GB] in a mixture of PBS and 0.05 % Tween-20. The 'blocking5 buffer was removed and the cells were incubated for 1 hour at room temperature with rabbit anti phospho-Akt (Ser473) antibody solution (50 ul per well; Cell Signalling, Hitchin, Herts, U.K., Catalogue No 9277) that had been diluted 1:500 in 'blocking' buffer. Cells were washed
30 three times in a mixture of PBS and 0.05 % Tween-20. Subsequently, cells were incubated for 1 hour at room temperature with Alexafluor488 labelled goat anti-rabbit IgG (50 p.1 per well; Molecular Probes, Invitrogen Limited, Paisley, UK, Catalogue No. All008) that had

WO 2008/023161

PCT/GB2007/003179

been diluted 1:500 in 'blocking' buffer. Cells were washed 3 times with a mixture of PBS and 0.05 % Tween-20. An aliquot of PBS (50 pA) was added to each well and the plates were sealed with black plate sealers and the fluorescence signal was detected and analysed.
Fluorescence dose response data obtained with each compound were analysed and the 5 degree of inhibition of Serine 473 in Akt was expressed as an IC50 value.
The compounds tested in this assay exhibited IC50 values against mTOR of less than 10u.m.
The following compounds exhibited IC50 values against mTOR of less than lp.m: lbu, Ice, 12b, 18de, 18dg, 18j, lar, 19e, 19h, 19i, 191, 19m, 19n, 19o, 18n, 18o, 18z, 18aa, 18ag, 18ai,
10 18al, lv, 18az, lah, 7e, 7i, 7j, 5d, 5f, 4v, 4ab, 4aj, 5t, 5u, 5w, 5x, 5y, 5z, 3f, 3g, 18bp, 18bs, 18bv, 18by, 18cb, 18cv, law, 3u, lbf, ISct, 19q, 19s, 19u, 19v, 19w, lau, 5r, 4t, 18dj3 lei, 2d: 2e, lcs, 2h, 2j and lew, with the following compounds exhibiting IC50 values against mTOR of less than300nM: Ibo, lbp, Ij, lbx, lby, lef, lei, lcj, 4an, 4ap, 4av, 12d, 18dh, 18di, 6a, In, lp, lq, 18e, 18h, 19b, 19c, 19f, 19k, 18p, lbd, 18w, 18ab, 18af, I8aj, 18aq, 18as, 18av,
15 18ay, 18bb, 18bc, 18bf, 18bl, lab, 4p, 9a, lav, 3a, 5b, 5c, 5e, 5g, 4aa, 4ad, 4ah, 5v, 3e, 18bq, 18bt, I8bz, 18ca, I8cd, 18cg, I8ci, 18bx, 5n, lam, lao, 18cn, 18cx, lbk, 13b, 4g, 5s, 4q, 18dd, lep, lcq, 2f, 2g, 13g, lev and let, with the following compounds exhibiting IC50 values against mTOR of less than lOOnM: lb, la, lc, Id, Ibl, lbm, If, li, lg, lh, lbr, lbs, Ibv, le, lbz, lec, Ik, leg, 11, 4al, 4am, 4ao, 4aq, 4as, 4at, 4au, 4aw, 4ax, 4ay, 4az, 4ba, 4bb, 4bc, 4bd,
20 4be,4bf, 12c, 12a, 18a, las, Is, 18c, 18d, 18f, 18g, 18i, 18k, 19j, 18m, 18q, 18r, 18s, 18t, 18u, 18v, 18x, 18y, 18ac, 18ad, 18ae, 18ah, 18ak, 18am, 18an, 18ap, 18ar, 18au, 18aw, 18ax, 18ba, I8bd, 18be, 18bg, 18bi, 18bk, 18bh, 18bj, 18bm, lbg, 8b3 4h, lba, 8a, laa, lac, lae, laf, lag, 14b, lbc, 4i, 4j, 4k, 41,4m, 4n, 4o, 18bn, 18bo, 4u, lbb, lat, 7b, 7c, 7d, 7f, 7g, 7k, 5a, 4w, 4x, 4y, 4z, 4ac, 4af, 4ai, 18br, 18bw, 18cc, 18cf, 18ch, 18cj, 18ck, 18cl, 4ak, 18cm, 4a, 3i, 3y,
25 lak, lal, lap, lbe, 18co, 18cr, 18cs, 18db, 19p, 31, lu, 4b, 5q, 4c, 4e, 4f, 4d, laz, 4r, 4s, len, 1 co and 3ad. For example, Compound 18di has an IC50 of 15inM
The Comparative Examples were also tested and when compared to the corresponding Examples, the exhibited IC50 values for the Comparative Examples were higher than those of the corresponding Examples. For example Example Ik was measured to have an IC50 of
30 83nm whereas Comparative Example Ik was measured to haveanIC50of 4l2nm. Therefore, compounds of the present invention are more active in reducing cell growth.

WO 2008/023161

PCT/GB2007/003179

Reference List
The following documents are all herein incorporated by reference.
1) Brown, et al, Nature, 369,756-758 (1994)
2) Chiu, et al, Proc NatlAcadSci, 91,12574-12578 (1994) 5 3) Sabatini, et al., Cell, 78,35-43, (1994)

4) Sabers, et al., J Biol Chem, 270, 825-822 (1995)
5) Abraham, Curr Opin Immunol, 8,412-418 (1996)
6) Schmelze and Hall, Cell, 103,253-262 (2000)
7) Burnett, et al, Proc Natl AcadSci, 95,1432-1437(1998) 10 8) Terada,e/ al, Proc NatlAcadSci, 91,11477-11481 (1994)
9) Jeffries, et al, EMBOJ, 16 ,3693-3704 (1997)
10) Bjornsti and Houghton, Nat Rev Cancer, 4, 335-348 (2004)
11) Gingras, e/o/., Genes Dev, 13, 1422-1437(1999)
12) Gingras, et al., Genes Dev, IS, 807-826 (2001)
15 13) Neuhaus, et al, Liver Transplantation, 1,473-484 (2001)
14) Woods and Marks, Ann Rev Med, 55, 169-178 (2004)
15) Dahia, Endocrine-Related Cancer, 7,115-129 (2000)
16) Cristofano and Pandolfi, Cell, 100, 387-390 (2000)
17) Samuels, et al, Science, 304, 554 (2004)
20 18) Huang and Houghton, Curr Opin Pharmacol, 3,371-377 (2003)
19) Sawyers, Cancer Cell, 4, 343-348 (2003)
20) Huang and Houghton, Curr Opin in Invest Drugs, 3,295-304 (2002)
21) Brunn, et al,EMBO J, 15, 5256-5267 (1996)
22) Edinger, et al, Cancer Res, 63, 8451-8460, (2003)
25 23) Lawrence, etal, Curr Top Microbiol Immunol, 279,199-213 (2004)
24) Eshleman, etal, Cancer Res, 62, 7291-7297 (2002)
25) Berge, et al, J. Pharm, Sci., 66,1-19 (1977).
26) Green, T. and Wuts, P., "Protective Groups in Organic Synthesis", 3rd Edition, John Wiley and Sons (1999).
30 27) "Handbook of Pharmaceutical Additives", 2nd Edition (eds. M. Ash and I. Ash), 2001 (Synapse information Resources, Inc., Endicott, New York, USA).

WO 2008/023161

PCT/GB2007/003179

28) "Remington's Pharmaceutical Sciences", 20th edition, pub. Lippincott, Williams & Wilkins, 2000.
29) "Handbook of Pharmaceutical Excipients", 2nd edition, 1994.

WO 2008/023161

PCT/CB2007/0D3179





1. A compound of formula I:

5 , or a pharmaceutically acceptable salt thereof, wherein: one or two of X5, X6 and X8 is N. and the others are CH;
R7 is selected from halo, OR01, SRS1, NRN!RN2, NRN7aC(0}RCI, NRN7bS02RS2a, an optionally substituted 5- to 20-memberedheteroaryl group, or an optionally substituted C5-20 aryl group, where R01 and Rsl are selected from H, an optionally substituted C5.20 aryl group, an 10 optionally substituted 5- to 20-memberedheteroaryl group, or an optionally substituted C1.7
.Nl __J DN2
alkyl group; R and R are independently selected from H, an optionally substituted C|_7 alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, an optionally substituted C5.20 aryl group or RN1 and RN2 together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms;
15 RC1 is selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5-to 20-membered heteroaryl group, an optionally substituted C1.7 alkyl group or NRN8RN9, where RN8 and RN9 are independently selected from H, an optionally substituted C1.7 alkyl group, an optionally substituted 5- to 20-memberedheteroaryl group, an optionally substituted C5-20 aryl group or RN8 and RN9 together with the nitrogen to which they are bound form an
20 optionally substituted heterocyclic ring containing between 3 and 8 ring atoms;
RS2a is selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5-
to 20-memberedheteroaryl group, or an optionally substituted Ci_7 alkyl group;
RN7a and RN7b are selected from H and a CM alkyl group;
R2 is selected from H, halo, OR02, SRs2b, NRN5RN6, an optionally substituted 5- to 20-
25 membered heteroaryl group, and an optionally substituted C5.20 aryl group,
wherein R02 and RS2b are selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted C1.7 alkyl group; RN5 and RN6 are independently selected from H, an optionally substituted C1.7 alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, and an optionally

WO 2008/023161

PCT/GB2007/003179

substituted 05:20 aryl group, or RN5 and RN(5 together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms.
2. A comnound of formula la or lb:

5
, or a pharmaceutically acceptable salt thereof, wherein:
one or two of X , X and X is N, and the others are CH;
R7 is selected from halo, OR01, SRSI, NRN1RN2, NRN7aC(0)RC!, NRN7bS02RS2a, an optionally
substituted 5- to 20-memberedheteroaryI group, or an optionally substituted C5.20 aryl group,
10 where R01 and RS! are selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted Ci_7 alkyl group; RN1 and RN2 are independently selected from H, an optionally substituted C1.7 alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, an optionally substituted C5.20 aryl group or RNi and RN2 together with the nitrogen to which they are bound
15 form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms;
RCI is selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5-to 20-membered heteroaryl group, an optionally substituted C1.7 alkyl group or NRNBRN9, where RN8 and RN9 are independently selected from H, an optionally substituted C1.7 alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, an optionally substituted
20 C5-20 aryl group or RN8 and RN5 together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 3 and S ring atoms; R a is selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5-to 20-membered heteroaryl group, or an optionally substituted C1-7 alkyl group; RN7a and RN7b are selected from H and a CM alkyl group;
25 R2 is selected from H, halo, OR02, SRS2\ NRN5RN6, an optionally substituted 5- to 20-membered heteroaryl group, and an optionally substituted C5-20 aryl group, wherein R02 and RS2b are selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted C1.7 alkyl group; RN5 and RN6 are independently selected from H, an optionally substituted C1.7

WO 2008/023161

PCT/CB2007/003179

alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, and an optionally substituted C5-20 aryl group, or RN5 and RN6 together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms.
5 3. A compound of formula la:

, or a pharmaceutically acceptable salt thereof, wherein:
one or two of X5, X6 and X8 is N, and the others are CH;
R7 is selected from halo, OR01, SRS1, NRN1RN2, NRN7aC(0)RC1, NRN7bS02RS2a, an optionally
10 substituted S- to 20-memberedheteroaryl group, or an optionally substituted C5-20 aryl g,roup, where R01 and RS1 are selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted C1_7 alkyl group; RN1 and RN2 are independently selected from H, an optionally substituted C1.7 alkyl group, an optionally substituted 5- to 20-membered heteroaryl group, an optionally
15 substituted C5.20 aryl group or RN1 and RN2 together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 3 and S ring atoms; R ' is selected from H, an optionally substituted C5.20 aryl group, an optionally substituted 5-to 20-membered heteroaryl group, an optionally substituted Ci.7 alkyl group or NRN8RN9, where Rm and RN9 are independently selected from H, an optionally substituted C1.7 alkyl
20 group, an optionally substituted 5- to 20-membered heteroaryl group, an optionally substituted Cs-20 aryl group or RNS and RN9 together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms; RS2a is selected from H, an optionally substituted C5-20 aryl group, an optionally substituted 5-to 20-membered heteroaryl group, or an optionally substituted Q.7 alkyl group;
25 RN7a and RN7b are selected from H and a CM alkyl group;
R2 is selected from H, halo, OR02, SRS2b, NRN5RN6, an optionally substituted 5- to 20-membered heteroaryl group, and an optionally substituted C5.20 aryl group, wherein R02 and RS2b are selected from H, an optionally substituted C5-20 aryl group, an optionally substituted 5- to 20-membered heteroaryl group, or an optionally substituted C1.7

WO 2008/023161

PCT/GB2007/003179

alkyl group; RN5 and RN6 are independently selected from H, an optionally substituted C1-7 alkyl group, an optionaUy substituted 5- to 20-membered heteroaryl group, and an optionally substituted C5-20 aryl group, or RN5 and RN6 together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 3 and 8 ring atoms. 5
4. A compound according to any one of claims 1 to 3, wherein only one of X5, X6 and X8
isN.
5. A compound according to any one of claims 1 to 3, wherein X6 is CH and X5 and X8 10 areN.
6. A compound according to any one of claims 1 to 4, wherein X8 is N.
7. A compound according to any one of the preceding claims, wherein R7 is selected 15 from an optionally substituted C5-20 aryl group, OR01, NRN1RN2, NRN7aC(0)Rcl and
NRN7bS02Rs2a
8. A compound according to any one of claims 1 to 6, wherein R7 is an optionally
substituted phenyl or pyridyl group.
20
9. A compound according to any one of claims 1 to 6, wherein R7 is OR01, and R01 is a
Cj.7 alkyl group, which may be substituted.
10. A compound according to any one of claims 1 to 6, wherein R7 is NRN1RN2, and RN2 is 25 H.
11. A compound according to claim 10, wherein RN] is C3.7 cycloalkyl.
12. A compound according to claim 10, wherein RN1 is C5-6 aryl. 30
13. A compound according to any one of claims 1 to 6, wherein R7 is NRN7aC(0)RC], and
RN7a is preferably H.

WO 2008/023161

PCT/GB2007/003179

14. A compound according to claim 13, wherein R is selected from an optionally
substituted C5-20 aryl group, C3-20 heterocyclyl, C1-7 alkyl and NRNSRN9, where RNS is
hydrogen, and RN9 is C 1.7 alkyl.
5
15. A compound according to any one of claims 1 to 6, wherein R7 is NRN7bS02RS2a, and
RN7bisH.
16. A compound according to claim 15, wherein RS2a is C1-7 alkyl.
10
17. A compound according to Claim 8 wherein R7 is an optionally substituted phenyl
group, wherein the optional substituents are preferably selected from halo, hydroxyl, cyano,
C1.7 alkyl, Cj-7alkoxy, sulfonamino (for example -NHS(=0)2C].7alkyl) amino (for example
-NH2, C5^arylamino, Ci-7alkyIamino, and di-(Ci-7alkyl)amino), and amido (for example
15 -CONH2j -CONHCi.7alkyl, -CON(C,.7alkyl)2 and -CONHheterocycyl) and wherein the susbtitutent alkyl, alkoxy, or aryl groups may be further optionally substituted by one or more groups selected from halo, hydroxyl, C1-7 alkyl, C1.7 alkoxy, C5-6aryl, -NHS(=0)2C].7alkyl, Cs-earylamino, di-(C1-7alkyl)amino and Ci^alkylarnino.
20 18. A compound according to claim 8 wherein R7 is an optionally substituted phenyl group selected from


WO 2008/023161

PCT/GB2007/003179

Z is H, For OR03;
R03 is selected from hydrogen or an optionally substituted C].6 alkyl group; RNi0 is selected from hydrogen, C(0)RC2, C(S)RC33 S02Rs\ an optionally substituted C5.20 heterocyclyl group, an optionally substituted C5_2o aryl group, or an optionally 5 substituted CMO alkyl group where Rc2 and RC3 are selected from H, an optionally substituted C5-20 aryl group, an optionally substituted C5.20 heterocyclyl group, an optionally substituted C1.7 alkyl group or NRNnRN12, where RN11 and RN12 are independently selected from H, an optionally substituted C1.7 alkyl group, an optionally substituted C5.2o heterocyclyl group, an optionally substituted C5.20 aryl group or RNn and RN12 together with the nitrogen to which
10 they are bound form a heterocyclic ring containing between 3 and 8 ring atoms; and R is selected from H, an optionally substituted C5-20 aryl group, an optionally substituted C5-20 heteroaryl group, or an optionally substituted C1-7 alkyl group; RN,0a is selected from hydrogen or an optionally substituted C1-10 alkyl group; or Rwa and RWQ together with the nitrogen to which they are bound form an optionally
15 substituted heterocyclic ring containing between 3 and 8 ring atoms.
19. A compound according to claim 8, wherein R7 is an optionally substituted phenyl
1_-i_J C



20 wherein
R°3 is selected from hydrogen or an optionally substituted C1-6alkyl group.
20. A compound according to claim 8, wherein R7 is

25 wherein

WO 2008/023161

PCT/GB2007/Q03179

ZisH,ForORUj;
RN1° is selected from hydrogen, -C(0)CH3, -C(0)CH2OH, -CH3) -CH2CH3) -CH3CH2OH, -CH(CH3)2) -CH2CH2OMe, -CH2C(CH3)2, -CH2CH2C(CH3)2, -CH(CH3)CH2C(CH3)2, -CH2CH2CH2N(CH3)2, cycloproyl, cyclopentyl, cyclohexyl, cycloheptyl, -CH2cyclopropyl, 5 methylcyclohexyl, cyanocyclohexyl, pyrazolyl, hydroxypyrrolidinyl, -CH2imidazole; RN10a is hydrogen; or
RNi0 and RN10a together with the nitrogen to which they are bound form an optionally substituted heterocyclic ring containing between 5 or 6 ring atoms; and where the optional subtsituents are selected from halo, hydroxyl, Ci^alkyloxy. 10
7 .
21. A compound according to claim 8 wherein R is an optionally substituted phenyl
group, wherein the optional substituents are selected from fluoro, hydroxyl, cyano, nitro,
methyl, niethoxy, -OCH2CH3, -NH2, -NHS02CH3j -CH2NHSO2CH3, -OCHF2, -CH2OH,
-C02H, -CONH2, -CONHMe, -CONHEt, -CONHCH(CH3)2, -CONHCH2CH2F,
15 -CONHCH2CHF2, -CONHCH2CH2OH, -CONMeEt, -CONMe2,
N-methylpiperazinylcarbonyl and 4-hydroxypiperidinylcarbonyl
22. A compound according to claim 7 wherein R7 is selected from


WO 2008/023161

PCT/GB2007/003179


5 23. A compound according to any one of the preceding claims, wherein R2 is NRN5RN6, where RN5 and RN6 together with the nitrogen to which they are bound form a heterocyclic ring containing between 3 and 8 ring atoms, which may optionally be substituted.
24. A compound according to claim 23, wherem R2 is selected from morpholino, 10 thiomorpholino, piperadinyl, piperazinyl, homopiperazinyl and pyrrolidinyl.
25. A compound according to Claim 23 wherein R2 wherein R2 is


WO 2008/023161

PCT/GB2007/003179






26.

A compound according to claim 23, wherein R2 is

WO 2008/023161

PCT/GB2007/003179





WO 2008/023161

PCT/GB2007/003179



28. A compound according to Claim 1 selected from any one of the Examples.
29. A compound according to Claim 1 selected from any one of Examples lbu, Ice, 12b, 18de, 18dg, 18j, lar, 19e, 19h, 19i, 191, 19m, 19n, 19o, 18n, 18o, 18z, 18aa, 18ag, 18ai, 18al,
10 lv, 18az, lah, 7e, 7i, 7j, 5d, 5f, 4v, 4ab, 4aj, 5t, 5u, 5w, 5x, 5y, 5z, 3f, 3g, 18bp, 18bs, lSbv, 18by, lScb, 18cv, law, 3u, Ibf, 18ct, 19q, 19s, 19u, 19v, 19w, lau, 5r, 4t, 18dj, lei, 2d, 2e, lcs, 2h, 2j, lew, lbo, lbp, lj, Ibx, lby, lef, lei, lcj, 4an, 4ap, 4av, 12d, 18dh, I8di, 6a, In, lp, lq, 18e, 18h, 19b, 19c, 19f, 19k, 18p, lbd, 18w, 18ab, 18af, 18aj, 18aq, 18as, 18av, 18ay, 18bb, I8bc, 18bf, I8bl, lab, 4p, 9a, lav, 3a, 5b, 5c, 5e, 5g, 4aa, 4ad, 4ah, 5v, 3e, 18bq, 18bt,
15 18bz, 18ca, 18cd, 18cg, 18ci, 18bx, 5n, lam, lao, 18cn, 18cx, lbk, 13b, 4g, 5s, 4q, 18dd, lep, Icq, 2f, 2g, I3g, lev, let, lb, la, tc, Id, lbl, lbm, If, li, lg, lh, lbr, lbs, lbv, le, lbz, lec, Ik. leg, 11, 4al, 4am, 4ao, 4aq, 4as, 4at, 4au, 4aw, 4ax, 4ay, 4az, 4ba, 4bb, 4bc, 4bd, 4be, 4bf, 12c, 12a, 18a, las, Is, 18c, 18d, 18f, 18g, 18i, 18k, 19j, 18m, 18q, 18r, 18s, 18t, 18u, 18v,

WO 2008/023161

PCT/GB2007/003179

18x, 18y, 18ac, 18ad, 18ae, 18ah, 18ak, 18am, 18an, 18ap, 18ar, 18au, 18aw, 18ax, 18ba, 18bd, 18be, 18bg, 18bi, 18bk, 18bh, 18bj, 18bm, lbg, 8b, 4h, lba, 8a, laa, lac, lae, laf, lag, 14b, Ibc, 4i, 4j, 4k, 41, 4m, 4n, 4o, 18bn, ISbo, 4u, lbb, 3at, 7b, 7c, 7d, 7f, 7g, 7k, 5a, 4w, 4x, 4y, 4z, 4ac, 4af, 4ai, 18br, 18bw, 18cc, 18cf, 18ch, 18cj, 18ck, 18cl, 4ak, 18cm, 4a, 3i, 3y, 5 lak, lal, lap, Ibe, 18co, 18cr, 18cs, 18db, 19p, 31, lu, 4b, 5q, 4c, 4e, 4f, 4d, laz, 4r, 4s, len, lco, 3ad, lcr, lew, ley, Idv, 15c, lei, 1cm, lcn, lcq, lev, lex, ldi, Idj, leb, lcj, lck, let, leu, lcz, ldb, ldc, ldd, lde, Idg, ldh, ldk, ldl, 1dm, ldn, Ido, ldp, ldq, ldt, ldu, ldw, idy, ldz, lea, lee, led, lee, 18dm, lSdnand 18do.
10 30. A compound according to Claim 1 selected from any one of Examples lbo, lbp, lj, lbx, lby, let lei, lcj, 4an, 4ap, 4av, 12& 18dh, 18di, 6a, In, lp, lq, 18e, 18h, 19b, 19c, 19f, 19k, 18p, lbd, 18w, 18ab, 18af, 18aj, 18aq, 18as, 18av, 18ay, 18bb, 18bc, 18bf, 18bl, lab, 4p, 9a, lav, 3a, 5b, 5c, 5e, 5g, 4aa, 4ad, 4ah, 5v, 3e, 18bq, 18bt, 18bz, 18ca, 18cd, 18cg, I8ci, ISbx, 5n, lam, lao, 18cn, 18cx, lbk, 13b, 4g, 5s, 4q, 18dd, lep, lcq, 2f, 2g, 13g, lev, let, lb,
15 la, lc, Id, lbl, lbm, If, li, ig, lh, lbr, lbs, lbv, le, Ibz, Ice, Ik, leg, 11, 4al, 4am, 4ao, 4aq, 4as, 4at, 4au, 4aw, 4ax, 4ay, 4az, 4ba, 4bb, 4bc, 4bd, 4be, 4bf, 12c, 12a, 18a, las, Is, 18c, 18d, 18f, 18g, 18i, 18k, 19j, 18m, 18q, 18r, 18s, 18t, 18u, 18v, 18x, 18y, 18ac, 18ad, 18ae, 18ah, 18ak, 18am, 18an, 18ap, 18ar, 18au, 18aw, 18ax, 18ba, 18bd, 18be, 18bg, 18bi, 18bk, 18bh, 18bj, 18bm, lbg, 8b, 4h, lba, 8a, laa, lac, lae, laf, lag, 14b, lbc, 4i, 4j, 4k, 41, 4m, 4n,
20 4o, 18bn, 18bo, 4u, lbb, lat, 7b, 7c, 7d, 7f, 7g, 7k, 5a, 4w, 4x, 4y, 4z, 4ac, 4af, 4ai, 18br, 18bw, 18cc, 18cf, 18ch, 18cj, ISck, 18cl, 4ak, 18cm, 4a, 3i, 3y, lak, lal, lap, lbe, 18co, 18cr, 18cs, 18db, 19p, 31, lu, 4b, 5q, 4c, 4e, 4f, 4d, laz, 4r, 4s, lcn, lco, 3ad, lcl, 1cm, lcn, lcq, lev, lex, ldi, Idj, leb, lcj, lck, let, leu, lcz, ldb, ldc, ldd, lde, Idg, ldh, ldk, ldl, 1dm, ldn, ldo, ldp, ldq, ldt, ldu, ldw, ldy, ldz, lea, lee, led, lee, 18dm, 18dn and 18do.
25
31. A compound according to Claim 1 selected from any one of Examples lb, la, lc, Id, lbl, lbm, If, H, lg, lb, lbr, lbs, lbv, le, Ibz, lee, Ik, leg, 11, 4al, 4am, 4ao, 4aq, 4as, 4at, 4au, 4aw, 4ax, 4ay, 4az, 4ba, 4bb, 4bc, 4bd, 4be, 4bf, 12c, 12a, 18a, las, Is, 18c, 18d, ISf, 18g, 18i, 18k, 19j, 18m, 18q, 18r, 18s, 18t, 18u, 18v, 18x, 18y, 18ac, 18ad, 18ae, 18ah, 18ak,
30 18am, 18an, 18ap, 18ar, 18au, 18aw, 18ax, 18ba, 18bd, 18be, 18bg, 18bi, 18bk, 18bh, 18bj, 18bm, lbg, 8b, Ah, lba, 8a, laa, lac, Ue, laf, lag, 14b, lbc, 4i, 4j, 4k, 41, 4m, 4n, 4o, 18bn, 18bo, 4u, lbb, lat, 7b, 7c, 7d, 7f, 7g, 7k, 5a, 4w, 4x, 4y, 4z, 4ac, 4af, 4ai, 18br, 18bw, IScc,

WO 2008/023161

PCT/GB2007/003J79

18cf, 18ch, 18cj, 18ck, 18cl, 4ak, 18cm, 4a, 3i, 3y, lak, lal, lap, Ibe, 18co, 18cr, 18cs, 18db, 19p, 31, lu, 4b, 5q, 4c, 4e, 4f, 4d, laz, 4r, 4s, len, lco, 3ad, lcj, Ick, let, leu, lcz, ldb, Idc, ldd, lde, ldg, ldh, ldk, ldl, 1dm, ldn, ldo, ldp, ldq, ldt, ldu, ldw, Idy, Idz, lea, lee, led, lee, 18dm, lSdnand 18do. 5
32. A compound according to Claim 1 selected from any one of Examples la, lu, lal, lap, lat, laz, lco, lde, ldg, ldh, ldk, ldl, ldp, ldq, ldr, Ids, ldt, ldu, ldy, lee, lee, 12d, 14b, 18dnandl8do.
10 33. A pharmaceutical composition comprising a compound according to any one of claims 1 to 32, and a pharmaceutically acceptable carrier or diluent,
34. A compound according to any one of claims 1 to 32 for use in a method of treatment
of the human or animal body.
15
35. The use of a compound according to any one of claims 1 to 32 in the preparation of a
medicament for treating a disease ameliorated by the inhibition of mTOR.
36. A compound according to any one of claims 1 to 32, or a pharmaceutically acceptable
20 salt thereof, for use as a medicament.
37. The use of a compound according to any one of claims 1 to 32, or a pharmaceutically
acceptable salt thereof, in the manufacture of a medicament for use in the production of a
mTOR inhibitory effect in a warm-blooded animal such as man.
25
38. The use of a compound according to any one of claims 1 to 32, or a pharmaceutically
acceptable salt thereof, in the manufacture of a medicament for use in the production of an
anti-cancer effect in a warm-blooded animal such as man.
30 39. The use of a compound according to any one of claims 1 to 32, or a pharmaceutically acceptable .salt thereof, in the manufacture of a medicament for use in the treatment of melanoma, glioma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian

WO 2008/023161

PCT/GB2007/003179

cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, endometrium, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries.
5 40. The use of a compound according to any one of claims 1 to 32, or a pharmaceutically acceptable salt thereof, in the production of a mTOR inhibitory effect in a warm-blooded animal such as man.
41. The use of a compound according to any one of claims 1 to 32, or a pharmaceutically
10 acceptable salt thereof, in the production of an anti-cancer effect in a warm-blooded animal
such as man.
42. The use of a compound according to any one of claims 1 to 32, or a pharmaceutically
acceptable salt thereof, in the treatment of melanoma, glioma, papillary thyroid tumours,
15 cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, endometrium, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries.
20 43. A method for producing a mTOR inhibitory effect in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound according to any one of claims 1 to 32, or a pharmaceutically acceptable salt thereof.
25 44. A method for producing an anti-cancer effect in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound according to any one of claims 1 to 32, or a pharmaceutically acceptable salt thereof.
30 45. A method of treating melanoma, glioma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, endometrium,

WO 2008/023161

PCT/GB2007/003179



10

breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries, in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound according to any one of claims 1 to 32, or a pharmaceutically acceptable salt thereof.
46. A pharmaceutical composition which comprises a compound according to any one of claims 1 to 32, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically-acceptable diluent or carrier for use in the production of a mTOR inhibitory effect in a warm-blooded animal such as man.
47. A pharmaceutical composition which comprises a compound according to any one of claims 1 to 32, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically-acceptable diluent or carrier for use in the production of an anti-cancer effect in a warm-blooded animal such as man.

15
48. A phannaceutical composition which comprises a compound according to any one of claims 1 to 32, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically-acceptable diluent or carrier for use in the treatment of melanoma, glioma, papillary thyroid tumours, cholangiocarcinomas, colon.cancer, ovarian cancer, lung cancer, 20 leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, endometrium, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries in a warm-blooded animal such as man.
Dated this 19lh day of February, 2009.


Documents:

398-MUMNP-2009-ABSTRACT(29-4-2013).pdf

398-mumnp-2009-abstract.doc

398-mumnp-2009-abstract.pdf

398-MUMNP-2009-CANCELLED PAGES(29-4-2013).pdf

398-MUMNP-2009-CLAIMS(20-2-2009).pdf

398-MUMNP-2009-CLAIMS(AMENDED)-(29-4-2013).pdf

398-mumnp-2009-claims.doc

398-mumnp-2009-claims.pdf

398-MUMNP-2009-CORRESPONDENCE(1-8-2012).pdf

398-MUMNP-2009-CORRESPONDENCE(12-3-2013).pdf

398-MUMNP-2009-CORRESPONDENCE(21-8-2009).pdf

398-MUMNP-2009-CORRESPONDENCE(6-8-2010).pdf

398-mumnp-2009-correspondence.pdf

398-MUMNP-2009-DESCRIPTION(COMPLETE)-(20-2-2009).pdf

398-mumnp-2009-description(complete).doc

398-mumnp-2009-description(complete).pdf

398-MUMNP-2009-FORM 1(1-8-2012).pdf

398-MUMNP-2009-FORM 1(20-2-2009).pdf

398-MUMNP-2009-FORM 1(29-4-2013).pdf

398-mumnp-2009-form 1.pdf

398-MUMNP-2009-FORM 13(1-8-2012).pdf

398-MUMNP-2009-FORM 18(6-8-2010).pdf

398-MUMNP-2009-FORM 2(COMPLETE)-(20-2-2009).pdf

398-MUMNP-2009-FORM 2(TITLE PAGE)-(20-2-2009).pdf

398-MUMNP-2009-FORM 2(TITLE PAGE)-(29-4-2013).pdf

398-mumnp-2009-form 2(title page).pdf

398-mumnp-2009-form 2.doc

398-mumnp-2009-form 2.pdf

398-MUMNP-2009-FORM 26(21-8-2009).pdf

398-MUMNP-2009-FORM 3(12-3-2013).pdf

398-MUMNP-2009-FORM 3(21-8-2009).pdf

398-MUMNP-2009-FORM 3(29-4-2013).pdf

398-mumnp-2009-form 3.pdf

398-mumnp-2009-form 5.pdf

398-MUMNP-2009-OTHER DOCUMENT(12-3-2013).pdf

398-MUMNP-2009-OTHER DOCUMENT(29-4-2013).pdf

398-mumnp-2009-pct-ib-304.pdf

398-mumnp-2009-pct-isa-210.pdf

398-MUMNP-2009-REPLY TO EXAMINATION REPORT(29-4-2013).pdf

398-mumnp-2009-wo international publication report a1.pdf


Patent Number 256458
Indian Patent Application Number 398/MUMNP/2009
PG Journal Number 25/2013
Publication Date 21-Jun-2013
Grant Date 19-Jun-2013
Date of Filing 20-Feb-2009
Name of Patentee KUDOS PHARMACEUTICALS LIMITED
Applicant Address 15 STANHOPE GATE, LONDON, W1K 1LN,
Inventors:
# Inventor's Name Inventor's Address
1 MARTIN, NIALL, MORRISSON, BARR KUDOS PHARMACEUTICALS LIMITED, 410 CAMBRIDGE SCIENCE PARK, MILTON ROAD, CAMBRIDGE CB4 0PE.
2 LEROUX, FREDERIC, GEORGES, MARIE KUDOS PHARMACEUTICALS LIMITED, 26 FOUNDRY LANE, HORSHAM, SUSSED RH13 5PX,
3 MALAGU, KARINE KUDOS PHARMACEUTICALS LIMITED, 26 FOUNDARY LANE, HORSHAM, SUSSEX RH13 5PX,
4 MENEAR, KEITH, ALLAN KUDOS PHARMACEUTICALS LIMITED, 26 FOUNDARY LANE, HORSHAM, SUSSEX RH13 5PX,
5 DUGGAN, HEATHER, MARY ELLEN KUDOS PHARAMACEUTICALS LIMITED, 26 FOUNDRY LANE, HORSHAM, SUSSEX RH13 5PX,
6 SMITH, GRAEME, CAMERON, MURRAY KUDOS PHARMACEUTICALS LIMITED, 410 CAMBRIDGE SCIENCE PARK, MILTON ROAD, CAMBRIDGE, CB4 0PE,
PCT International Classification Number C07D 471/04
PCT International Application Number PCT/GB2007/003179
PCT International Filing date 2007-08-21
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/938,776 2007-05-18 U.S.A.
2 60/823,311 2006-08-23 U.S.A.