Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PYRROLO[2,3-D]PYRIMIDINE DERIVATIVES AND THEIR USE IN THE TREATMENT OF CANCER
Document Type and Number:
WIPO Patent Application WO/2021/074251
Kind Code:
A1
Abstract:
The invention provides compounds of the formula (1): or a salt or tautomer thereof wherein A, R1, R2, R3, R4, R5 and R6 are as defined herein. The compounds are inhibitors of Wee1 and/or PLK1 kinase and are envisaged to be useful in the treatment of cancers.

Inventors:
BOYLE ROBERT GEORGE (GB)
WALKER DAVID WINTER (GB)
MAJOR MERIEL RUTH (GB)
Application Number:
PCT/EP2020/078960
Publication Date:
April 22, 2021
Filing Date:
October 14, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
SENTINEL ONCOLOGY LTD (GB)
International Classes:
C07D487/04; A61K31/519; A61P35/00; C07D519/00
Domestic Patent References:
WO2018098561A12018-06-07
WO2015038417A12015-03-19
WO2009098236A12009-08-13
Foreign References:
CN110467615A2019-11-19
CN111718348A2020-09-29
Other References:
MORIARTY K J ET AL: "The synthesis and SAR of 2-amino-pyrrolo[2,3-d]pyrimidines: A new class of Aurora-A kinase inhibitors", BIORGANIC & MEDICINAL CHEMISTRY LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 16, no. 22, 15 November 2006 (2006-11-15), pages 5778 - 5783, XP027966083, ISSN: 0960-894X, [retrieved on 20061115]
Attorney, Agent or Firm:
HUTCHINS, Michael Richard (GB)
Download PDF:
Claims:
CLAIMS

1. A compound of the formula (1 ): or a salt or tautomer thereof; wherein:

A is CH or N;

R1 is selected from -C(OH)(Alk1)(Alk2), -N=S(0)(Alk3)(Alk4) and a group:

Aik1 and Aik2 are the same or different and each is a C1-3 saturated hydrocarbyl group; or Aik1 and Aik2 together with the carbon atom to which they are attached form a C3-4 cycloalkyl ring;

Aik3 and Aik4 are the same or different and each is a C1-3 hydrocarbyl group; or Aik3 and Aik4 together with the sulphur atom to which they are attached form a 4-6 membered thiacycloalkyl ring; p is 1 or 2, and q is 1 or 2, provided that the sum of p + q is either 2 or 3;

R8 is hydrogen, fluorine or methyl;

R2 is selected from hydrogen, halogen, C1-3 alkyl and C1-3 alkoxy and C1-3 fluoroalkyl;

R3 is selected from hydrogen, fluorine and methyl;

R4 is selected from hydrogen, fluorine, methyl and cyano; R5 is L-Cyc1 ;

L is -(CH2)m-B-(CH2)n- m and n are independently selected from 0 and 1 ;

B is absent or is selected from -C(0)N(Rc)-, -N(Rc)C(0)-, -N(RC)-, -0-, -N(Rc)CH2C(0)-, S, SCO) and -S(0)2-;

Rc is hydrogen or a C1-4 hydrocarbyl group;

Cyc1 is selected from:

• a C3-6 cycloalkyl group optionally substituted with one or more substituents selected from R7;

• a 4- to 7-membered monocyclic, heterocyclic group containing 1 or 2 heteroatoms selected from N and O and being optionally substituted with one or more substituents selected from R7;

• a 7- to 11 - membered bicyclic heterocyclic group containing 1 or 2 heteroatoms selected from N and O and being optionally substituted with one or more substituents selected from R7; and

• phenyl optionally substituted with one or more substituents R7;

R6 is selected from hydrogen, halogen, C1-4 alkyl, C1-4 alkoxy and C1-4 fluoroalkyl;

R7 is selected from C1-4 hydrocarbyl, halogen, hydroxy, oxo, C(0)Ra, C(0)0Ra, C(0)N(Ra)(Rb), N(Rb)C(0)Ra, N(Rb)C(0)N(Ra)(Rb), and a C2.5alkane-diyl group, wherein the C2.5alkane-diyl group together with an atom or atoms of Cyc1 to which it is attached forms a cyclic group;

Ra is selected from hydrogen and a C1-3 hydrocarbyl group; and

Rb is selected from hydrogen and a C1-3 hydrocarbyl group; and wherein, in each substituent consisting of or containing a hydrocarbyl group, the hydrocarbyl group is selected from alkyl, alkenyl, alkynyl and cycloalkyl groups and combinations thereof.

2. A compound according to claim 1 wherein A is N.

3. A compound according to claim 1 or claim 2 wherein R1 is selected from - C(CH3)2OH, -N=S(0)(CH3)2 and: 4. A compound according to any one of claims 1 to 3 wherein R2 is hydrogen.

5. A compound according to any one of claims 1 to 4 wherein R3 is hydrogen.

6. A compound according to any one of claims 1 to 5 wherein R4 is hydrogen or fluorine.

7. A compound according to any one of claims 1 to 6 wherein B is absent or is selected from -C(0)N(Rc)-, -N(Rc)C(0)-, -N(RC)-, and -0-.

8. A compound according to any one of claims 1 to 7 wherein Cyc1 is selected from:

• cyclopropyl;

• a 5- to 7-membered monocyclic, non-aromatic heterocyclic group containing 1 or 2 heteroatom ring members selected from N and O and being optionally substituted with one or more substituents selected from R7;

• a 7- to 11 - membered bicyclic non-aromatic group heterocyclic group containing 2 nitrogen heteroatom ring members and being optionally substituted with one or more substituents selected from R7; and

• phenyl optionally substituted with one or more substituents R7.

9. A compound according to claim 8 wherein Cyc1 is selected from:

• cyclopropyl;

• a tetrahydrofuran, tetrahydropyran, pyrrolidine, piperidine, piperazine, homopiperazine, homopiperidine or morpholine group, each optionally substituted with one or more substituents selected from R7;

• a 2,5-diazabicyclo[2.2.1 ]heptane, 4,7-diazaspiro[2.5]octane or 3,9- diazaspiro[5.5]undecane, each optionally substituted with one or more substituents selected from R7; and

• phenyl optionally substituted with one or more substituents R7. 10. A compound according to any one of claims 1 to 9 wherein R7 is selected from Ci- 3 alkyl, C2-3 alkanoyl, cyclopropylcarbonyl, oxo, and C1-4 alkoxycarbonyl.

11. A compound according to any one of claims 1 to 10 wherein R6 is selected from hydrogen and chlorine. 12. A pharmaceutical composition comprising a compound as defined in any one of claims 1 to 11 and a pharmaceutically acceptable excipient.

13. A compound as defined in any one of claims 1 to 11 for use in medicine, for example for use as a Wee1 kinase inhibitor in the treatment of cancer.

14. A compound as defined in any one of claims 1 to 11 for use in medicine, for example for use as a PLK1 kinase inhibitor in the treatment of cancer.

15. An invention as defined in any one of Embodiments 1 .1 to 1.77, 2.1 to 2.72, 3.1 and 4.1 to 4.12.

Description:
PYRROLO[2,3-D]PYRIMIDINE DERIVATIVES AND THEIR USE IN THE TREATMENT OF CANCER

This invention relates to compounds that inhibit or modulate the activity of Wee1 and/or PLK1 kinases, pharmaceutical compositions containing the compounds and the therapeutic uses of the compounds.

Background of the Invention

The cell cycle is a highly controlled process and there are numerous cell-cycle checkpoints that prevent or delay mitosis in the event of detected DNA damage. This provide the cell with time to repair the damaged DNA before proliferating via mitosis.

The G2-M DNA damage checkpoint ensures that cells do not initiate mitosis until damaged or incompletely repaired DNA is sufficiently repaired following replication. Entry into mitosis from the G2-M checkpoint is controlled by phosphorylation of Cdk1 and its association with cyclin B.

Cyclic-dependent kinases (Cdk) are a family of serine/threonine protein kinases that are involved in the GM-2 checkpoint, which controls the entry of the cell into mitosis.

In normal cells, Cdk1 complexes with Cyclin B into order to induce mitosis. The Cdk1 /cyclin B complex is activated by dephosphorylation of its Tyr15 residue by phosphatase, Cdc25c and the resulting activated complex causes the cell to enter mitosis.

The Wee1 enzyme is a nuclear kinase belonging to the Tyrosine family of protein kinases, which is capable of deactivating the Cdk1 /Cyclin B complex by phosphorylation.

DNA damage is detected by the ataxia-telangiectasia-related (ATR) protein kinase pathway and results in phosphorylation and activation of Chk1 kinase. The Chk1 kinase subsequently phosphorylates Wee1 .

At high levels of phosphorylation, Wee1 phosphorylates Cdk1 at its Tyr15 residue to maintain it in its inactive state in association with cyclin B and prevent the damaged cell from entering mitosis. Wee1 is therefore a key negative regulator of cell cycle progression.

Once the DNA damage has been repaired, Wee1 activity decreases and therefore inhibitory phosphorylation of Cdk1 is lost and the cell enters mitosis. Wee1 is highly expressed in numerous types of cancers that rely on the G2-M checkpoint for DNA repair, particularly those that have a deficient G1 checkpoint and therefore rely on the G2-M checkpoint for survival. These cancers rely on the G2-M checkpoint in order to prevent excessive DNA damage and to prevent apoptosis resulting from mitotic catastrophe. Examples of such cancers include p53 mutant cancers.

Inhibition of Wee1 is therefore seen as an attractive target for these cancers in order to abrogate the G2-M checkpoint leading to unscheduled mitosis and cell death by mitotic catastrophe.

A wide range of cancers have been reported to be sensitive to Wee1 inhibitors, both as a monotherapy and in combination with DNA damaging agents (such as Gemcitabine or ionising radiation).

One type of cancer reported to be susceptible to treatment with Wee1 inhibitors is medulloblastoma. Medulloblastoma is a common primary brain cancer in children. The cancer typically originates towards the back and the bottom of the brain, on the floor of the skull and/or in the cerebellum or posterior fossa. Medulloblastomas are invasive and rapidly growing tumours. Unlike many brain tumours, Medulloblastomas can spread through the cerebrospinal fluid and therefore commonly metastasise to other surfaces of the brain and to the spinal cord. It has been found that small molecule inhibition of Wee1 suppresses medulloblastoma growth (Harris etal, Mol Cancer. 2014; 13: 72).

A number of other types of cancer have been reported to be sensitive to Wee1 inhibition, including:

• Diffuse Intrinsic Pontine Glioma (DIPG) (see Caretti etal., “WEE1 kinase inhibition enhances the radiation response of diffuse intrinsic pontine gliomas”, Mol Cancer Ther., 2013, 12(2), 141-50);

• Glioblastoma (see Mir etal. “In silico analysis of kinase expression identifies WEE1 as a gatekeeper against mitotic catastrophe in glioblastoma”, Cancer Cell. 2010,18(3), 244-57 and Sarcar etal., “Targeting Radiation-Induced G2 Checkpoint Activation with the Wee-1 Inhibitor MK-1775 in Glioblastoma Cell Lines”, Mol Cancer Ther., 2011, 10(12), 2405-14);

• Ependymoma (see Kool et al, “Therapeutic Targeting of Ependymoma as Informed by Oncogenic Enhancer Profiling”, Nature. 2018 January 04; 553(7686):

101-105. doi:10.1038/nature25169. • Acute myeloid leukemia (see Porter etal., “Integrated genomic analyses identify WEE1 as a critical mediator of cell fate and a novel therapeutic target in acute myeloid leukemia”, Leukemia, 2012, 26, 1266-1276);

• Cervical cancer (see Hirai etal., “Small-molecule inhibition of Wee1 kinase by MK-1775 selectively sensitizes p53-deficient tumour cells to DNA-damaging agents”, Mol Cancer Ther., 2009, 8(11), 2992-3000);

• Colorectal cancer (see Hirai etal., “Small-molecule inhibition of Wee1 kinase by MK-1775 selectively sensitizes p53-deficient tumour cells to DNA-damaging agents”, Mol Cancer Ther., 2009, 8(11), 2992-3000);

• Head and neck squamous cell carcinoma (see Diab etal., “Multiple Defects Sensitize p53-Deficient Head and Neck Cancer Cells to the WEE1 Kinase Inhibition”, Mol Cancer Res, 2019, 17(5), 1115-1128);

• Malignant melanoma (see Magnuseen etal., “High expression of Wee1 is associated with poor disease-free survival in malignant melanoma: potential for targeted therapy”, PLoS One., 2012;7(6):e38254);

• Ovarian cancer (see Zhang etal., “WEE1 inhibition by MK1775 as a single-agent therapy inhibits ovarian cancer viability”, Oncol Lett., 2017, 14(3), 3580-3586);

• Pancreatic cancer (see Cuneo etal., “Dose Escalation Trial of the Wee1 Inhibitor Adavosertib (AZD1775) in Combination With Gemcitabine and Radiation for Patients With Locally Advanced Pancreatic Cancer”, J Clin Oncol. 2019 Aug 9:JC01900730 and Rajeshkumar etal., “MK-1775, a potent Wee1 inhibitor, synergizes with gemcitabine to achieve tumour regressions, selectively in p53- deficient pancreatic cancer xenografts”, Clin Cancer Res., 2011, 17(9), 2799-806);

• Sarcoma (see Kreahling etal., “MK1775, a selective Wee1 inhibitor, shows single agent antitumour activity against sarcoma cells”, Mol Cancer Ther., 2012, 11(1), 174-82);

• Small cell lung cancer (see Richer etal., “WEE1 Kinase Inhibitor AZD1775 Has Preclinical Efficacy in LKB1 -Deficient Non-Small Cell Lung Cancer”, Cancer Res., 2017, 77(17), 4663-72 and Lallo etal., “The Combination of the PARP Inhibitor Olaparib and the WEE1 Inhibitor AZD1775 as a New Therapeutic Option for Small Cell Lung Cancer”, Clin Cancer Res, 2018, 24(20), 5153-5164); and • Triple-negative breast cancer (see Chen etal., “Cyclin E Overexpression Sensitizes Triple-Negative Breast Cancer to Wee1 Kinase Inhibition”, Clin Cancer Res., 2018, 24(24), 6594-6610).

• Uterine Serous Carcinoma (USC) or Uterine Carcinosarcoma (see Liu et at., ASCO 2020 virtual meeting abstracts and phase II clinical trial (NCT03668340) for Wee1 inhibitor AZD1775 in women with recurrent or persistent uterine serous carcinoma or uterine carcinosarcoma

A small-molecule inhibitor of Wee1 , AZD1775 (also referred to as Adavosertib or MK1775):

(AZD1775) has been developed and shows good inhibitory activity against Wee1 in cell-based assays (~80nM) and also in vivo when used as a sole therapeutic agent or in combination with a further chemotherapeutic agent (e.g. gemcitabine or carboplatin). However, data have been published suggesting that AZD1775 has sub-optimal ADME (Absorption, Distribution, Metabolism, Excretion) properties. It is highly effluxed from the brain when dosed orally (see Tellingen et al, 2018, Investigational New Drugs, ATP-binding cassette transporters limit the brain penetration of Wee1 inhibitors). It is anticipated that this would limit clinical efficacy of the compound against brain malignancies. Furthermore, data from US patent application US 2019/0084985 show that, following a 20 mg/kg single oral dose of AZD1775 to mice bearing dorsal flank tumour xenografts, only trace amounts of AZD1775 penetrated the brain. In addition, it was reported that when AZD1775 was dosed in a Phase I trial it was found to have an apparent oral clearance of 2.67 L/hour which is high considering that it exceeds human liver blood flow (see Do et al, 2015, Phase I Study of Single-Agent AZD1775 (MK-1775), a Wee1 Kinase Inhibitor, in Patients With Refractory Solid Tumors). It would therefore be beneficial to develop further compounds that have the ability to inhibit Wee1 kinase.

Cancer cells can acquire resistance to chemotherapeutic drugs as a result of the target protein developing mutations or having altered expression levels meaning that the target protein can no longer be inhibited by the drug or inhibition no longer results in therapeutic efficacy. Lewis eta!., “Upregulation of Myt1 Promotes Acquired Resistance of Cancer Cells to Wee1 Inhibition”, Cancer Res., 2019, 79(23), pp.5971 -5985 describes primary resistance to Wee1 inhibition in cancer cells following exposure to AZD1775.

Chemotherapeutic drugs that act on more than one target and/or more than one pathway are less prone to resistance as, in the event that resistance is developed at one pathway, the drug is still able to exert its effect via another alternative pathway.

PLK1 is a serine/threonine kinase consisting of 603 amino acids and having a molecular weight of 66 kDa and is an important regulator of the cell cycle. In particular, PLK1 is important to mitosis and is involved in the formation of and the changes in the mitotic spindle and in the activation of CDK/cyclin complexes during the M-phase of the cell cycle.

Tumour protein p53 functions as a tumour suppressor and plays a role in apoptosis, genomic stability and inhibition of angiogenesis. It is known that tumours with both p53- deficiency and high PLK1 expression may be particularly sensitive to PLK1 inhibitors (Yim et al., Mutat Res Rev Mutat Res, (2014). 761 , 31 -39).

The protein expressed by the normal KRAS gene performs an essential function in normal tissue signalling. The mutation of a KRAS gene by a single amino acid substitution, and in particular a single nucleotide substitution, is responsible for an activating mutation which is an essential step in the development of many cancers. The mutated protein that results is implicated in various malignancies, including lung adenocarcinoma, mucinous adenoma, ductal carcinoma of the pancreas and colorectal carcinoma. Like other members of the Ras family, the KRAS protein is a GTPase and is involved in many signal transduction pathways.

KRAS acts as a molecular on/off switch. Once it is turned on, it recruits and activates proteins necessary for the propagation of growth factor and other receptors' signal such as c Raf and PI-3 Kinase. Normal KRAS binds to GTP in the active state and possesses an intrinsic enzymatic activity which cleaves the terminal phosphate of the nucleotide converting it to GDP. Upon conversion of GTP to GDP, KRAS is turned off. The rate of conversion is usually slow but can be sped up dramatically by an accessory protein of the GTPase-activating protein (GAP) class, for example RasGAP. In turn, KRAS can bind to proteins of the Guanine Nucleotide Exchange Factor (GEF) class, for example SOS1 , which forces the release of bound nucleotide. Subsequently, KRAS binds GTP present in the cytosol and the GEF is released from ras-GTP. In mutant KRAS, its GTPase activity is directly removed, rendering KRAS constitutively in the active state. Mutant KRAS is often characterised by mutations in codons 12, 13, 61 or mixtures thereof. The viability of cancer cells carrying a mutant KRAS is known to be dependent on Polo- Like Kinase 1 (PLK1) and it has been shown that silencing PLK1 leads to the death of cells containing mutant KRAS (see Luo et al., Cell. 2009 May 29; 137(5): 835-848). Compounds that inhibit PLK1 should therefore be useful in treating cancers that arise from KRAS mutations. Therefore, it would also be beneficial to develop compounds that have the ability to inhibit Wee1 kinase and an additional kinase, such as PLK1.

The Invention

The present invention provides a class of novel pyrrolopyrimidine compounds as inhibitors of Wee1 and/or PLK1 kinases. or a salt or tautomer thereof; wherein:

A is CH or N;

R 1 is selected from -C(OH)(Alk 1 )(Alk 2 ), -N=S(0)(Alk 3 )(Alk 4 ) and a group:

Aik 1 and Aik 2 are the same or different and each is a C1-3 saturated hydrocarbyl group; or Aik 1 and Aik 2 together with the carbon atom to which they are attached form a C3-4 cycloalkyl ring;

Aik 3 and Aik 4 are the same or different and each is a C1-3 hydrocarbyl group; or Aik 3 and Aik 4 together with the sulphur atom to which they are attached form a 4-6 membered thiacycloalkyl ring; p is 1 or 2, and q is 1 or 2, provided that the sum of p + q is either 2 or 3;

R 8 is hydrogen, fluorine, methyl, hydroxy and methoxy;

R 2 is selected from hydrogen, halogen, C1-3 alkyl and C1-3 alkoxy and C1-3 fluoroalkyl;

R 3 is selected from hydrogen, fluorine and methyl;

R 4 is selected from hydrogen, fluorine, methyl and cyano;

R 5 is L-Cyc 1 ;

L is -(CH 2 )m-B-(CH 2 )n-; m and n are independently selected from 0 and 1 ;

B is absent or is selected from -C(0)N(R c )-, -N(R c )C(0)-, -N(R C )-, -0-, -N(R C )CH 2 C(0)-, S, SCO) and -S(0) 2 -;

R c is hydrogen or a C 1-4 hydrocarbyl group;

Cyc 1 is selected from:

• a C3-6 cycloalkyl group optionally substituted with one or more substituents selected from R 7 ; • a 4- to 7-membered monocyclic, heterocyclic group containing 1 or 2 heteroatoms selected from N and O and being optionally substituted with one or more substituents selected from R 7 ;

• a 7- to 11 - membered bicyclic heterocyclic group containing 1 or 2 heteroatoms selected from N and O and being optionally substituted with one or more substituents selected from R 7 ; and

• phenyl optionally substituted with one or more substituents R 7 ;

R 6 is selected from hydrogen, halogen, C 1-4 alkyl, C 1-4 alkoxy and C 1-4 fluoroalkyl;

R 7 is selected from C1-4 hydrocarbyl, halogen, hydroxy, oxo, C(0)R a , C(0)0R a , C(0)N(R a )(R b ), N(R b )C(0)R a , N(R b )C(0)N(R a )(R b ), and a C -5 alkane-diyl group, wherein the C2- 5 alkane-diyl group together with an atom or atoms of Cyc 1 to which it is attached forms a cyclic group;

R a is selected from hydrogen and a C 1-3 hydrocarbyl group; and

R b is selected from hydrogen and a C 1-3 hydrocarbyl group; and wherein, in each substituent consisting of or containing a hydrocarbyl group, the hydrocarbyl group is selected from alkyl, alkenyl, alkynyl and cycloalkyl groups and combinations thereof.

Particular and preferred compounds of the formula (1 ) are as defined in the Embodiments

1 .2 to 1 .77 below.

1 .2 A compound according to Embodiment 1 .1 wherein A is CH.

1 .3 A compound according to Embodiment 1.1 wherein A is N.

1 .4 A compound according to any one of Embodiments 1 .1 to 1 .3 wherein R 1 is -C(OH)(Alk 1 )(Alk 2 )

1 .5 A compound according to Embodiment 1 .4 wherein Aik 1 and Aik 2 are the same or different and each is a C1-2 alkyl group; or Aik 1 and Aik 2 together with the carbon atom to which they are attached form a cyclopropyl ring.

1 .6 A compound according to Embodiment 1 .5 wherein Aik 1 and Aik 2 are both methyl groups; or Aik 1 and Aik 2 together with the carbon atom to which they are attached form a cyclopropyl ring. 1 .7 A compound according to Embodiment 1 .6 wherein Aik 1 and Aik 2 are both methyl groups.

1 .8 A compound according to any one of Embodiments 1 .1 to 1 .3 wherein R 1 is -N=S(0)(Alk 3 )(Alk 4 ). 1 .8A A compound according to Embodiment 1 .8 wherein Aik 3 and Aik 4 are the same or different and each is a Ci-3hydrocarbyl group.

1 .9 A compound according to Embodiment 1 .8 or Embodiment 1 .8A wherein Aik 3 and Aik 4 are the same or different and each is a C1-3 alkyl group.

1.10 A compound according to Embodiment 1 .9 wherein Aik 3 and Aik 4 are the same or different and each is a C1-2 alkyl group.

1.11 A compound according to Embodiment 1 .9 wherein Aik 3 and Aik 4 are both methyl groups.

1.12 A compound according to any one of Embodiments 1 .1 to 1 .3 wherein R 1 is a group: 1.13 A compound according to Embodiment 1 .12 wherein p and q are both 1 .

1 .14 A compound according to Embodiment 1 .12 or Embodiment 1 .13 wherein R 8 is fluorine.

1.15 A compound according to any one of Embodiments 1.1 to 1 .3 wherein R 1 is selected from -C(CH 3 ) 2 OH, -N=S(0)(CH 3 ) 2 and:

1.16 A compound according to any one of Embodiments 1 .1 to 1 .3 wherein R 1 is -C(CH 3 ) 2 OH. 1.17 A compound according to any one of Embodiments 1 .1 to 1 .3 wherein R 1 is -N=S(0)(CH 3 ) 2 .

1.18 A compound according to any one of Embodiments 1 .1 to 1 .3 wherein R 1 is

1.19 A compound according to any one of Embodiments 1 .1 to 1 .18 wherein R 2 is selected from hydrogen, fluorine, chlorine, C1-3 alkyl, C1-3 alkoxy, difluoromethyl and trifluoromethyl.

1 .20 A compound according to Embodiment 1 .19 wherein R 2 is selected from hydrogen, fluorine, chlorine, methyl, ethyl, methoxy, trifluoromethyl and difluoromethyl.

1 .21 A compound according to Embodiment 1 .20 wherein R 2 is selected from hydrogen, fluorine, methyl and methoxy.

1 .22 A compound according to Embodiment 1 .21 wherein R 2 is hydrogen or fluorine.

1 .23 A compound according to Embodiment 1 .22 wherein R 2 is hydrogen.

1 .24 A compound according to any one of Embodiments 1 .1 to 1 .23 wherein R 3 is hydrogen.

1 .25 A compound according to any one of Embodiments 1 .1 to 1 .24 wherein R 4 is selected from hydrogen, fluorine and cyano.

1 .26 A compound according to Embodiment 1 .25 wherein R 4 is hydrogen or fluorine.

1 .27 A compound according to Embodiment 1 .25 wherein R 4 is fluorine.

1 .28 A compound according to Embodiment 1 .25 wherein R 4 is hydrogen.

1 .29 A compound according to Embodiment 1 .25 wherein R 4 is cyano.

1 .30 A compound according to any one of Embodiments 1 .1 to 1 .29 wherein m is 0.

1.31 A compound according to any one of Embodiments 1 .1 to 1 .29 wherein m is 1 . 1 .32 A compound according to any one of Embodiments 1 .1 to 1 .31 wherein n is 0.

1 .33 A compound according to any one of Embodiments 1 .1 to 1 .31 wherein n is 1 .

1 .34 A compound according to any one of Embodiments 1 .1 to 1 .33 wherein B is absent or is selected from -C(0)N(R c )-, -N(R c )C(0)-, -N(R C )-, and -0-.

1 .35 A compound according to Embodiment 1 .34 wherein B is -C(0)N(R c )-.

1 .36 A compound according to Embodiment 1 .34 wherein B is -N(R c )C(0)-.

1 .37 A compound according to Embodiment 1 .34 wherein B is -N(R C )-.

1 .38 A compound according to any one of Embodiments 1 .1 to 1 .37 wherein R c is selected from hydrogen, C1-3 alkyl, C3-4 cycloalkyl and cyclopropylmethyl.

1 .39 A compound according to Embodiment 1 .38 wherein R c is hydrogen, methyl or ethyl.

1 .40 A compound according to Embodiment 1 .38 wherein R c is hydrogen or methyl.

1 .41 A compound according to Embodiment 1 .38 wherein R c is hydrogen.

1 .42 A compound according to Embodiment 1 .38 wherein R c is methyl.

1 .43 A compound according to Embodiment 1 .34 wherein B is absent.

1 .44 A compound according to Embodiment 1 .34 wherein B is -0-.

1 .45 A compound according to any one of Embodiments 1 .1 to 1 .33 wherein L is absent or is selected from -0-, -CONH-, -NH-, -NHCO- and -IMHCH2-.

1 .46 A compound according to any one of Embodiments 1 .1 to 1 .45 wherein Cyc 1 is selected from:

• a C3-6 cycloalkyl group optionally substituted with one or more substituents selected from R 7 ;

• a 5- to 7-membered monocyclic, heterocyclic group (e.g. a non-aromatic group) containing 1 or 2 heteroatom ring members selected from N and O and being optionally substituted with one or more substituents selected from R 7 ; • a 7- to 11 - membered bicyclic heterocyclic group (e.g. a non-aromatic group) containing 1 or 2 heteroatom ring members (for example 2 nitrogen heteroatom ring members) selected from N and O and being optionally substituted with one or more substituents selected from R 7 ; and

• phenyl optionally substituted with one or more substituents R 7 . A compound according to Embodiment 1 .46 wherein Cyc 1 is selected from:

• a C cycloalkyl group optionally substituted with one or more substituents selected from R 7 ;

• a 5- to 7-membered monocyclic, heterocyclic group (e.g. a non-aromatic group) containing 1 or 2 heteroatom ring members selected from N and O and being optionally substituted with one or more substituents selected from R 7 ;

• a 7- to 11 - membered bicyclic heterocyclic group (e.g. a non-aromatic group such as a non-aromatic bridged bicyclic or spiro-bicyclic group) containing 1 or 2 nitrogen heteroatom ring members (for example 2 nitrogen heteroatom ring members) and being optionally substituted with one or more substituents selected from R 7 ; and

• phenyl optionally substituted with one or more substituents R 7 . A compound according to Embodiment 1 .47 wherein Cyc 1 is selected from:

• a cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl group optionally substituted with one or more substituents selected from R 7 ;

• a 5- to 7-membered monocyclic, non-aromatic heterocyclic group containing 1 or 2 heteroatom ring members selected from N and O and being optionally substituted with one or more substituents selected from R 7 ;

• a 7- to 11 - membered bicyclic non-aromatic (e.g. a bridged bicyclic or spiro- bicyclic group) heterocyclic group containing 1 or 2 nitrogen heteroatom ring members (for example 2 nitrogen ring members) and being optionally substituted with one or more substituents selected from R 7 ; and

• phenyl optionally substituted with one or more substituents R 7 . A compound according to Embodiment 1 .48 wherein Cyc 1 is selected from: cyclopropyl; • a 5- to 7-membered monocyclic, non-aromatic heterocyclic group containing 1 or 2 heteroatom ring members selected from N and O and being optionally substituted with one or more substituents selected from R 7 ;

• a 7- to 11 - membered bicyclic non-aromatic group heterocyclic group (e.g. a bridged bicyclic or spiro-bicyclic group) containing 2 nitrogen heteroatom ring members and being optionally substituted with one or more substituents selected from R 7 ; and

• phenyl optionally substituted with one or more substituents R 7 .

1 .50 A compound according to Embodiment 1 .49 wherein Cyc 1 is selected from:

• cyclopropyl;

• a 5- or 6-membered monocyclic, non-aromatic group heterocyclic group containing 1 or 2 heteroatoms selected from N and O and being optionally substituted with one or more substituents selected from R 7 ;

• a 7- to 11 - membered bicyclic non-aromatic group heterocyclic group (e.g. a bridged bicyclic or spiro-bicyclic group) containing 2 nitrogen heteroatoms and being optionally substituted with one or more substituents selected from R 7 ; and

• phenyl optionally substituted with one or more substituents R 7 .

1 .51 A compound according to Embodiment 1 .46 wherein Cyc 1 is selected from:

• cyclopropyl;

• a tetrahydrofuran, tetrahydropyran, pyrrolidine, piperidine, piperazine, homopiperazine, homopiperidine or morpholine group, each optionally substituted with one or more substituents selected from R 7 ;

• a 2,5-diazabicyclo[2.2.1 ]heptane, 4,7-diazaspiro[2.5]octane or 3,9- diazaspiro[5.5]undecane, each optionally substituted with one or more substituents selected from R 7 ; and

• phenyl optionally substituted with one or more substituents R 7 .

1 .52 A compound according to any one of Embodiments 1 .1 to 1.51 wherein R 7 is selected from C1-4 hydrocarbyl, halogen, hydroxy, oxo, C(0)R a , C(0)N(R a )(R b ), N(R b )C(0)R a , N(R b )C(0)N(R a )(R b ) and a C^alkane-diyl group, wherein the C^alkane- diyl group together with an atom or atoms of Cyc 1 to which it is attached forms a 3-6 membered cyclic group.

1 .53 A compound according to Embodiment 1 .52 wherein R 7 is selected from C1-4 hydrocarbyl, halogen, hydroxy, oxo, C(0)R a , C(0)N(R a )(R b ), N(R b )C(0)R a , and a C 2-5 alkane-diyl group, wherein the C 2-5 alkane-diyl group together with a carbon atom or atoms of Cyc 1 to which it is attached forms a C 3-6 cyclic group.

1 .54 A compound according to Embodiment 1 .53 wherein R 7 is selected from C1-4 hydrocarbyl, halogen, hydroxy, oxo, C(0)R a , C(0)N(R a )(R b ), N(R b )C(0)R a , and a C2-3 alkane-diyl group, wherein the C2-3 alkane-diyl group together with a carbon atom or atoms of Cyc 1 to which it is attached forms a C3-4 carbocyclic group.

1 .55 A compound according to Embodiment 1 .54 wherein R 7 is selected from C 1-4 hydrocarbyl, halogen, hydroxy, oxo, C(0)R a , and a C2 alkane-diyl group, wherein the C2 alkane-diyl group together with a carbon atom or atoms of Cyc 1 to which it is attached forms a cyclopropyl ring.

1 .56 A compound according to any one of Embodiments 1 .1 to 1.55 wherein R a is selected from hydrogen, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, cyclopropylmethyl and f-butyl.

1 .57 A compound according to Embodiment 1 .56 wherein R a is selected from hydrogen, methyl, ethyl and cyclopropyl.

1 .58 A compound according to any one of Embodiments 1 .1 to 1.57 wherein R b is selected from hydrogen, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, cyclopropylmethyl, methylcyclopropyl and f-butyl.

1 .59 A compound according to Embodiment 1 .58 wherein R b is selected from hydrogen, methyl, ethyl and cyclopropyl.

1 .60 A compound according to Embodiment 1 .59 wherein R b is hydrogen.

1 .61 A compound according to any one of Embodiments 1 .1 to 1.52 wherein R 7 is selected from C 1-3 alkyl, C 2-3 alkanoyl, cyclopropylcarbonyl, oxo, and C 1-4 alkoxycarbonyl.

1 .62 A compound according to Embodiment 1.61 wherein R 7 is selected from methyl, ethyl, acetyl, oxo, cyclopropylcarbonyl, propionyl and ferf-butoxycarbonyl. 1 .63 A compound according to any one of Embodiments 1 .1 to 1.45 wherein Cyc 1 is selected from groups AA to BK in Table 1 below, wherein an asterisk ( * ) denotes the point of attachment to group L:

Table 1

1 .64 A compound according to any one of Embodiments 1 .1 to 1.63 wherein R 6 is selected from hydrogen, halogen, C1-4 alkyl, C1-2 alkoxy and C1-2 haloalkyl.

1 .65 A compound according to Embodiment 1 .64 wherein R 6 is selected from hydrogen, fluorine, chlorine, methyl, ethyl, methoxy, trifluoromethyl and difluoromethyl.

1 .66 A compound according to Embodiment 1 .65 wherein R 6 is selected from hydrogen, fluorine, chlorine and methyl. 1 .66A A compound according to Embodiment 1 .65 wherein R 6 is selected from hydrogen, fluorine and chlorine.

1 .67 A compound according to Embodiment 1 .66 wherein R 6 is selected from hydrogen and chlorine.

1 .68 A compound according to Embodiment 1 .67 wherein R 6 is hydrogen.

1 .69 A compound according to Embodiment 1 .66 wherein R 6 is chlorine.

1 .70 A compound selected from the title compounds of Examples 1 to 51 herein.

1 .71 A compound selected from the title compounds of Examples 1 to 57 herein.

1 .72 A compound according to any one of Embodiments 1.1 to 1.71 which is in the form of a salt.

1 .73 A compound according to Embodiment 1 .72 wherein the salt is an acid addition salt.

1 .74 A compound according to Embodiment 1.72 or Embodiment 1 .73 wherein the salt is a pharmaceutically acceptable salt.

1 .75 A compound according to any one of Embodiments 1.1 to 1.71 which is in the form of a non-salt (e.g. free base).

1 .76 A compound according to any one of Embodiments 1 .1 to 1.75 which is in the form of a solvate.

1 .77 A compound according to Embodiment 1 .76 wherein the solvate is a hydrate. Definitions

References to “carbocyclic” and “heterocyclic” groups as used herein shall, unless the context indicates otherwise, include both aromatic and non -aromatic ring systems. Thus, for example, the term “carbocyclic and heterocyclic groups” includes within its scope aromatic, non-aromatic, unsaturated, partially saturated and fully saturated carbocyclic and heterocyclic ring systems.

The carbocyclic or heterocyclic groups can be aryl or heteroaryl groups. The aryl or heteroaryl groups can be monocyclic or bicyclic groups, as defined herein. The term “aryl” as used herein refers to a carbocyclic group having aromatic character and the term “heteroaryl” is used herein to denote a heterocyclic group having aromatic character. Where the context permits, the terms “aryl” and “heteroaryl” may embrace bicyclic ring systems wherein both rings are aromatic or one ring is non-aromatic and the other is aromatic. In such bicyclic systems containing one aromatic and one non -aromatic group, the group may be attached by the aromatic ring, or by the non-aromatic ring.

The term “non-aromatic group” refers to unsaturated ring systems without aromatic character, partially saturated and fully saturated carbocyclic and heterocyclic ring systems. The terms “unsaturated” and “partially saturated” refer to rings wherein the ring structure(s) contains atoms sharing more than one valence bond e.g. the ring contains at least one multiple bond e.g. a C=C N=C bond. The term “saturated” refers to rings where there are no multiple bonds between ring atoms. Saturated carbocyclic groups include the cycloalkyl groups cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. Partially saturated carbocyclic groups include the cycloalkenyl groups cyclopentenyl, cyclohexenyl, cycloheptenyl and cyclooctenyl. Non-aromatic heterocyclic groups include azetidine, pyrrolidine, piperidine, azepane, piperazine, morpholine, thiomorpholine, thiomorpholine S-oxide and S,S-dioxide, pyran (2H-pyran or 4H-pyran), dihydrothiophene, dihydropyran, dihydrofuran, dihydrothiazole, tetrahydrofuran, tetrahydrothiophene, dioxane, tetrahydropyran, imidazoline, imidazolidinone, oxazoline, thiazoline, pyrazoline and pyrazolidine.

The term “hydrocarbyl” as used herein refers to aliphatic, alicyclic and aromatic groups having an all-carbon backbone and consisting of carbon and hydrogen atoms, except where otherwise stated. Examples of hydrocarbyl groups include alkyl, cycloalkyl, cycloalkenyl, carbocyclic aryl, alkenyl, alkynyl, cycloalkylalkyl, cycloalkenylalkyl, and carbocyclic aralkyl, aralkenyl and aralkynyl groups. Such groups can be unsubstituted or, where stated, substituted by one or more substituents as defined herein. In certain cases, as defined herein, one or more, but not all, of the carbon atoms of the hydrocarbyl group may be replaced by another atom or group of atoms.

The term “alkylene” (e.g. as in C1-4 straight chain or branched chain alkylene) as used herein refers to an alkanediyl group, i.e. a divalent saturated acyclic straight chain or branched chain hydrocarbon group. Examples of straight chain alkanediyl groups include methylene (CH 2 ), ethylene (CH 2 CH 2 ) and propylene ((CH 2 CH 2 CH 2 ). Examples of branched chain alkanediyl groups include CH(CH 3 ), CH 2 CH(CH 3 )CH2 and CH 2 (CH 3 )CH 2 CH2.

The term “haloalkyl” as used herein refers to alkyl group substituted with one or more halogen atom substituents, and in particular fluorine substituents. Particular examples of haloalkyl groups are trifluoromethyl and difluoromethyl.

Salts

The compounds of the invention as defined in Embodiments 1.1 to 1 .71 may be presented in the form of salts.

The salts referred to above (and also defined in embodiments 1 .72, 1 .73 and 1.74) are typically acid addition salts.

The salts can be synthesized from the parent compound by conventional chemical methods such as methods described in Pharmaceutical Salts: Properties, Selection, and Use, P. Heinrich Stahl (Editor), Camille G. Wermuth (Editor), ISBN: 3-90639-026-8, Hardcover, 388 pages, August 2002. Generally, such salts can be prepared by reacting the free base form of the compound with the acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media such as ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are used.

Acid addition salts (as defined in Embodiment 1.73) may be formed with a wide variety of acids, both inorganic and organic. Examples of acid addition salts include salts formed with an acid selected from the group consisting of acetic, 2,2-dichloroacetic, adipic, alginic, ascorbic (e.g. L-ascorbic), L-aspartic, benzenesulphonic, benzoic, 4- acetamidobenzoic, butanoic, (+) camphoric, camphor-sulphonic, (+)-(1 S)-camphor-10- sulphonic, capric, caproic, caprylic, cinnamic, citric, cyclamic, dodecylsulphuric, ethane- 1 ,2-disulphonic, ethanesulphonic, 2-hydroxyethanesulphonic, formic, fumaric, galactaric, gentisic, glucoheptonic, D-gluconic, glucuronic (e.g. D-glucuronic), glutamic (e.g. L- glutamic), a-oxoglutaric, glycolic, hippuric, hydrobromic, hydrochloric, hydriodic, isethionic, (+)-L-lactic, (±)-DL-lactic, lactobionic, maleic, malic, (-)-L-malic, malonic, (±)- DL-mandelic, methanesulphonic, naphthalene-2-sulphonic, naphthalene-1 ,5-disulphonic, 1-hydroxy-2-naphthoic, nicotinic, nitric, oleic, orotic, oxalic, palmitic, pamoic, phosphoric, propionic, L-pyroglutamic, salicylic, 4-amino-salicylic, sebacic, stearic, succinic, sulphuric, tannic, (+)-L-tartaric, thiocyanic, p-toluenesulphonic, undecylenic and valeric acids, as well as acylated amino acids and cation exchange resins.

The salt forms of the compounds of the invention are typically pharmaceutically acceptable salts, and examples of pharmaceutically acceptable salts are discussed in Berge etal., 1977, "Pharmaceutically Acceptable Salts," J. Pharm. Sci., Vol. 66, pp. 1 -19. However, salts that are not pharmaceutically acceptable may also be prepared as intermediate forms which may then be converted into pharmaceutically acceptable salts. Such non-pharmaceutically acceptable salts forms, which may be useful, for example, in the purification or separation of the compounds of the invention, also form part of the invention.

N-Oxides

Many compounds of the Embodiments 1.1 to 1.77 may form N-oxides. Where a compound contains several amine functions, one or more than one nitrogen atom may be oxidised to form an N-oxide. Particular examples of N-oxides are the N-oxides of a tertiary amine or a nitrogen atom of a nitrogen-containing heterocycle.

N-Oxides can be formed by treatment of the corresponding amine with an oxidizing agent such as hydrogen peroxide or a per-acid (e.g. a peroxycarboxylic acid), see for example Advanced Organic Chemistry, by Jerry March, 4 th Edition, Wiley Interscience, pages. More particularly, N-oxides can be made by the procedure of L. W. Deady ( Syn . Comm. 1977, 7, 509-514) in which the amine compound is reacted with m-chloroperoxybenzoic acid (MCPBA), for example, in an inert solvent such as dichloromethane.

Further examples of conditions for forming N-oxides are disclosed in our earlier application WO2008/139152.

Geometric isomers and tautomers

The compounds of the invention may exist in a number of different geometric isomeric, and tautomeric forms and references to the compounds of formula (1 ) as defined in Embodiments 1 .1 to 1 .77 include all such forms. For the avoidance of doubt, where a compound can exist in one of several geometric isomeric or tautomeric forms and only one is specifically described or shown, all others are nevertheless embraced by formula (1) or subgroups, subsets, preferences and examples thereof. Optical Isomers

Where compounds of the formula contain one or more chiral centres, and can exist in the form of two or more optical isomers, references to the compounds include all optical isomeric forms thereof (e.g. enantiomers, epimers and diastereoisomers), either as individual optical isomers, or mixtures (e.g. racemic mixtures) or two or more optical isomers, unless the context requires otherwise.

The optical isomers may be characterised and identified by their optical activity (i.e. as + and - isomers, or c/and / isomers) or they may be characterised in terms of their absolute stereochemistry using the “R and S” nomenclature developed by Cahn, Ingold and Prelog, see Advanced Organic Chemistry by Jerry March, 4 th Edition, John Wiley & Sons, New York, 1992, pages 109-114, and see also Cahn, Ingold & Prelog, Angew. Chem. Int. Ed. Engl., 1966, 5, 385-415.

Optical isomers can be separated by a number of techniques including chiral chromatography (chromatography on a chiral support) and such techniques are well known to the person skilled in the art.

As an alternative to chiral chromatography, optical isomers can be separated by forming diastereoisomeric salts with chiral acids such as (+)-tartaric acid, (-)-pyroglutamic acid, (-)-di-toluoyl-L-tartaric acid, (+)-mandelic acid, (-)-malic acid, and (-)-camphorsulphonic, separating the diastereoisomers by preferential crystallisation, and then dissociating the salts to give the individual enantiomer of the free base.

Where compounds of the invention exist as two or more optical isomeric forms, one enantiomer in a pair of enantiomers may exhibit advantages over the other enantiomer, for example, in terms of biological activity. Thus, in certain circumstances, it may be desirable to use as a therapeutic agent only one of a pair of enantiomers, or only one of a plurality of diastereoisomers. Accordingly, the invention provides compositions containing a compound having one or more chiral centres, wherein at least 55% (e.g. at least 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%) of the compound of the formula (1) is present as a single optical isomer (e.g. enantiomer or diastereoisomer). In one general embodiment, 99% or more (e.g. substantially all) of the total amount of the compound of the formula (1 ) may be present as a single optical isomer (e.g. enantiomer or diastereoisomer). Isotopes

The compounds of the invention as defined in any one of Embodiments 1.1 to 1 .77 may contain one or more isotopic substitutions, and a reference to a particular element includes within its scope all isotopes of the element. For example, a reference to hydrogen includes within its scope 1 H, 2 H (D), and 3 H (T). Similarly, references to carbon, nitrogen, oxygen and fluorine include within their scope respectively 11 C, 12 C, 13 C and 14 C; 13 N and 14 N; 15 0, 16 0 and 18 0; and 18 F and 19 F.

The isotopes may be radioactive or non -radioactive. In one embodiment of the invention, the compounds contain no radioactive isotopes. Such compounds are preferred for therapeutic use. In another embodiment, however, the compound may contain one or more radioisotopes. Compounds containing such radioisotopes may be useful in a diagnostic context.

Solvates

Compounds of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 may form solvates.

Preferred solvates are solvates formed by the incorporation into the solid-state structure (e.g. crystal structure) of the compounds of the invention of molecules of a non-toxic pharmaceutically acceptable solvent (referred to below as the solvating solvent). Examples of such solvents include water, alcohols (such as ethanol, isopropanol and butanol) and dimethylsulphoxide. Solvates can be prepared by recrystallising the compounds of the invention with a solvent or mixture of solvents containing the solvating solvent. Whether or not a solvate has been formed in any given instance can be determined by subjecting crystals of the compound to analysis using well known and standard techniques such as thermogravimetric analysis (TGE), differential scanning calorimetry (DSC) and X-ray crystallography.

The solvates can be stoichiometric or non-stoichiometric solvates.

Particularly preferred solvates are hydrates, and examples of hydrates include hemihydrates, monohydrates and dihydrates. For a more detailed discussion of solvates and the methods used to make and characterise them, see Bryn et al., Solid-State Chemistry of Drugs, Second Edition, published by SSCI, Inc of West Lafayette, IN, USA, 1999, ISBN 0-967-06710-3.

Prodrugs

The compounds of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 may be presented in the form of a pro-drug.

By “prodrugs” is meant for example any compound that is converted in vivo into a biologically active compound of the formula (1), as defined in any one of Embodiments 1.1 to 1.77.

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 cleaved to yield the active drug. Such esters may be formed by esterification, for example, of any hydroxyl groups present in the parent compound with, where appropriate, prior protection of any other reactive groups present in the parent compound, followed by deprotection if required.

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, as in ADEPT, GDEPT, LIDEPT, etc.). For example, the prodrug may be a sugar derivative or other glycoside conjugate, or may be an amino acid ester derivative.

Also encompassed by formula (1) or subgroups, subsets, preferences and examples thereof are complexes (e.g. inclusion complexes or clathrates with compounds such as cyclodextrins, or complexes with metals) of the compounds.

Bioloaical Activity

Compounds of the formula (1 ) as defined in any one of Embodiments 1.1 to 1.77 have activity as inhibitors of Wee1 kinase and/or PLK1 . As such, they may be useful in preventing or treating disease states and conditions in which Wee1 kinase or mutant forms thereof or PLK1 or mutant forms thereof play an active part. As shown in Biological Examples a) and b) below, the compounds of the invention bind with Wee1 and PLK1 kinases in competition with ATP. Accordingly, the compounds of the invention are considered to inhibit Wee1/PLK1 by binding to the active site of the enzymes (rather than being allosteric inhibitors).

Therefore, it is envisaged that the compounds of Embodiments 1.1 to 1 .77 will be useful in treating a range of diseases or conditions, for example proliferative disorders such as cancers.

Accordingly, in further embodiments (Embodiments 2.1 to 2.9), the invention provides:

2.1 A compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 for use in medicine or therapy.

2.2 A compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 for use in preventing or treating disease states and conditions mediated by Wee1 kinase or mutant forms thereof.

2.3 A compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 for use in preventing or treating disease states and conditions characterised by abnormal expression of Wee1 kinase (e.g. over-expression or expression of a mutant form of Wee1 kinase).

2.4 A compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 for use in preventing or treating disease states and conditions mediated by PLK1 kinase or mutant forms thereof.

2.5 A compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 for use in preventing or treating disease states and conditions characterised by abnormal expression of PLK1 kinase (e.g. over-expression or expression of a mutant form of PLK1 kinase).

2.6 A compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 for use as an anti-cancer agent.

2.7 The use of a compound of the formula (1 ) as defined in any one of Embodiments 1 .1 to 1 .77 for the manufacture of a medicament for the treatment of cancer.

2.8 A method of treating a cancer, which method comprises administering to a subject in need thereof a therapeutically effective amount of a compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1.77, optionally together with another anti cancer agent or radiation therapy.

2.9 A compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 for use in enhancing a therapeutic effect of radiation therapy or chemotherapy in the treatment of a proliferative disease such as cancer.

2.10 The use of a compound of the formula (1) as defined in any one of Embodiments 1 .1 to 1 .77 for the manufacture of a medicament for enhancing a therapeutic effect of radiation therapy or chemotherapy in the treatment of a proliferative disease such as cancer.

2.11 A method for the prophylaxis or treatment of a proliferative disease such as cancer, which method comprises administering to a patient in combination with radiotherapy or chemotherapy a compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1.77.

Examples of proliferative disorders (e.g. cancers) as defined in Embodiments 2.6 to 2.11 include, but are not limited to carcinomas, for example carcinomas of the bladder, breast, colon, kidney, epidermis, liver, lung, oesophagus, gall bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, gastrointestinal system, or skin, hematopoieitic tumours such as leukaemia, B-cell lymphoma, T-cell lymphoma, Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, hairy cell lymphoma, or Burkett's lymphoma; hematopoieitic tumours of myeloid lineage, for example acute and chronic myelogenous leukaemias, myelodysplastic syndrome, or promyelocytic leukaemia; thyroid follicular cancer; tumours of mesenchymal origin, for example fibrosarcoma or rhabdomyosarcoma ; tumours of the central or peripheral nervous system, for example astrocytoma, neuroblastoma, glioma or schwannoma; melanoma; seminoma; teratocarcinoma; osteosarcoma; xeroderma pigmentosum; keratoctanthoma; thyroid follicular cancer; or Kaposi's sarcoma.

Further examples of proliferative disorders (e.g. cancers) as defined in Embodiments 2.6 to 2.11 are carcinoma of the uterus, glioblastoma (GBM); medulloblastoma and ependymoma.

One particular subset of cancers against which the compounds of Embodiments 1 .1 to 1 .77 should prove particularly active are cancers which are characterised by Wee1 overexpression or elevated expression of Wee1 or elevated activation (phosphorylation). The ability of the compounds of the invention to inhibit Wee1 kinase can be determined by means of the protocols set out in the Examples section below.

As described above, several types of cancer have been reported to be sensitive to Wee1 inhibition. Accordingly, further particular examples of cancers against which the compounds of Embodiments 1.1 to 1 .77 should prove particularly active are:

• Medulloblastoma;

• Diffuse Intrinsic Pontine Glioma (DIPG);

• Glioblastoma;

• Acute myeloid leukemia;

• Cervical cancer;

• Colorectal cancer;

• Head and neck squamous cell carcinoma;

• Malignant melanoma;

• Ovarian cancer;

• Pancreatic cancer;

• Sarcoma;

• Small cell lung cancer; and

• Triple-negative breast cancer; as well as

• Uterine Serous Carcinoma (USC) or Uterine Carcinosarcoma; and

• Ependymoma

A further sub-set of cancers against which the compounds of Embodiments 1 .1 to 1 .77 should prove particularly active includes cancers of the brain such as:

• Medulloblastoma

• Diffuse Intrinsic Pontine Glioma (DIPG); and

• Glioblastoma; and optionally

• Ependymoma. It has also been shown that Wee1 inhibitors can sensitize p53 deficient cancer cells to other DNA-damaging chemotherapeutic agents (see Hirai etai, ibid). Accordingly, it is envisaged that the compounds of the present invention can be used in the treatment of p53 deficient or mutated cancers.

Furthermore, it has been determined that tumours with BRCA1 mutations are especially sensitive to Wee1 inhibition due to the inability of the cancer to repair double-strand DNA breaks (see Do et al, 2015, Phase I Study of Single-Agent AZD1775 (MK-1775), a Wee1 Kinase Inhibitor, in Patients With Refractory Solid Tumors).

Other markers of sensitivity to Wee1 inhibition include, but are not limited to: a mutation in KRAS (an oncogene), a mutation in LKB1 (a tumour suppressor) or co-mutation (See Whitsett etal, 2017, WEE1 Kinase Inhibitor AZD1775 Has Preclinical Efficacy in LKB1 - Deficient Non-Small Cell Lung Cancer).

In addition, tumours with high levels of replication stress (RS) caused by inactivation of a tumour suppressor e.g. RB1 or those harbouring a DNA Damage Response (DDR) defect can be sensitive (see review by Fu etal, 2018, Strategic development of AZD1775, a Wee1 kinase inhibitor, for cancer therapy). Furthermore, RS in certain tumours such as medulloblastoma can be driven by the transcription factor n-Myc and such tumours are sensitive to small molecule Wee1 inhibition (see Moreira etal, 2020, Targeting MYC-driven replication stress in medulloblastoma with AZD1775 and gemcitabine).

It is also known that Wee1 inhibition sensitizes cancer cells to immunotherapy such as a PD-1 monoclonal antibody (mAb) checkpoint inhibitor (Allen etal, 2018, WEE1 kinase inhibition reverses G2/M cell cycle checkpoint activation to sensitize cancer cells to immunotherapy).

Accordingly, in further Embodiments 2.12 to 2.32, the invention also provides

2.12 A compound according to any one of Embodiments 1 .1 to 1.77, optionally in combination with a further therapeutic agent (e.g. a further therapeutic agent described herein), for use in the treatment of a cancer which is characterised by mutant or deficient tumour protein p53.

2.13 A method for the treatment of a subject (e.g. a human patient) suffering from a cancer characterised by mutant or deficient tumour protein p53, which method comprises administering to the subject and effective therapeutic amount of a compound of any one of Embodiments 1.1 to 1.77, optionally in combination with a further therapeutic agent (e.g. a further therapeutic agent described herein).

2.14 The use of a compound of any one of Embodiments 1 .1 to 1 .77, optionally in combination with a further therapeutic agent (e.g. a further therapeutic agent described herein), for the manufacture of a medicament for the treatment of a subject (e.g. a human patient) suffering from a cancer characterised by mutant or deficient tumour protein p53.

2.15 A compound as defined in any one of Embodiments 1 .1 to 1 .77 for use in the treatment of a cancer in a patient who has been screened and has been determined as suffering from a cancer which is characterised by mutant or deficient tumour protein p53.

2.16 The use of a compound as defined in any one of Embodiments 1 .1 to 1 .77 for the manufacture of a medicament for the treatment of a cancer in a patient who has been screened and has been determined as suffering from a cancer which is characterised by mutant or deficient tumour protein p53.

2.17 A method for the diagnosis and treatment of a cancer which is characterised by mutant or deficient tumour protein p53, which method comprises (i) screening a patient to determine whether a cancer from which the patient is suffering is one which is characterised by mutant or deficient tumour protein p53; and (ii) where it is indicated that the cancer is one which is characterised by mutant or deficient tumour protein p53, thereafter administering to the patient a therapeutically effective amount of a compound as defined in any one of Embodiments 1.1 to 1.77.

2.18 A compound of the formula (1) as defined in any one of Embodiments 1.1 to 1 .77 for use in preventing or treating cancers which are sensitive to Wee1 kinase inhibition.

2.19 A compound for use according to Embodiment 2.18 wherein the cancer which is sensitive to Wee1 kinase inhibition is a cancer which contains one or more mutations selected from mutations of BRCA1 , KRAS and LKB1 .

2.20 A compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 for use in treating a cancer in a patient who has been screened and has been determined as suffering from a cancer which is sensitive to Wee1 kinase inhibition.

2.21 A compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 for use in treating a cancer in a patient who has been screened and has been determined as suffering from a cancer which is sensitive to Wee1 kinase inhibition and contains one or more mutations selected from mutations of BRCA1 , KRAS and LKB1 .

2.22 The use of a compound of the formula (1 ) as defined in any one of Embodiments 1 .1 to 1 .77 for the manufacture of a medicament for a use as defined in any one of Embodiments 2.9 to 2.21 .

2.23 A method of treating a subject suffering from a cancer which is sensitive to Wee1 inhibition, which method comprises administering to the subject an effective amount of a compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77.

2.24 A method of treating a subject who has been diagnosed and has been found to be suffering from a cancer which is sensitive to Wee1 kinase inhibition, which method comprises administering to the subject an effective amount of a compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1.77.

2.25 A method of treating a subject who has been diagnosed and has been found to be suffering from a cancer which is sensitive to Wee1 kinase inhibition and contains one or more mutations selected from mutations of BRCA1 , KRAS and LKB1 , which method comprises administering to the subject an effective amount of a compound of the formula (1 ) as defined in any one of Embodiments 1 .1 to 1.77.

2.26 A method for the diagnosis and treatment of a disease state or condition mediated by Wee1 kinase which method comprises (i) screening a patient to determine whether a disease or condition from which the patient is or may be suffering is one which would be susceptible to treatment with a compound having activity against Wee1 kinase; and (ii) where it is indicated that the disease or condition from which the patient is thus susceptible, thereafter administering to the patient a compound as defined in any one of Embodiments 1.1 to 1.77.

2.27 The use of a compound as defined in any one of Embodiments 1 .1 to 1 .77 for the manufacture of a medicament for the treatment or prophylaxis of a disease state or condition in a patient who has been screened and has been determined as suffering from, or being at risk of suffering from, a disease or condition which would be susceptible to treatment with a compound having activity against Wee1 kinase.

2.28 A compound as defined in any one of Embodiments 1 .1 to 1 .77 for use in the treatment or prophylaxis of a disease state or condition in a patient who has been screened and has been determined as suffering from, or being at risk of suffering from, a disease or condition which would be susceptible to treatment with a compound having activity against Wee1 kinase.

2.29 A method for the diagnosis and treatment of a disease state or condition characterised by up-regulation of Wee1 kinase or the presence of a mutated form of Wee1 kinase, which method comprises (i) screening a patient to determine whether a disease or condition from which the patient is or may be suffering is one which would be susceptible to treatment with a compound having activity against Wee1 kinase; and (ii) where it is indicated that the disease or condition from which the patient is thus susceptible, thereafter administering to the patient a compound as defined in any one of Embodiments 1.1 to 1.77.

2.30 A method for the treatment of a disease state or condition characterised by up- regulation of Wee1 kinase or the presence of a mutated form of Wee1 , which method comprises administering a therapeutically effective amount of a compound as defined in any one of Embodiments 1 .1 to 1.77 to a patient who has been screened and has been determined as suffering from, or being at risk of suffering from, a disease or condition which would be susceptible to treatment with a compound having activity against Wee1 kinase.

2.31 The use of a compound as defined in any one of Embodiments 1 .1 to 1 .77 for the manufacture of a medicament for the treatment or prophylaxis of a disease, condition or disorder as defined in any one of Embodiments 2.1 to 2.28 in a patient who has been screened and has been determined as suffering from, or being at risk of suffering, from a said disease, condition or disorder which would be susceptible to treatment with a compound having activity against Wee1 kinase.

2.32 A compound as defined in any one of Embodiments 1 .1 to 1 .77 for use in the treatment or prophylaxis of a disease or condition or disorder in a patient who has been screened and has been determined as suffering from, or being at risk of suffering from, a disease, condition or disorder as defined in any one of Embodiments 2.1 to 2.31 which would be susceptible to treatment with a compound having activity against Wee1 kinase.

In addition, preferred compounds of the invention possess good PLK1 inhibitory activity. PLK1 is believed to inhibit p53 in cancer cells. Therefore, upon treatment with PLK1 inhibitors, p53 in tumour cells should be activated and hence should induce apoptosis. Accordingly, it is believed that compounds of the invention may be useful in treating cancers characterised by p53 deficiency or mutation in the TP53 gene.

Compounds that inhibit multiple kinase targets may be advantageous in the treatment of certain diseases, such as cancer, as in the event that resistance is developed a pathway in which one kinase is involved, the drug is still able to exert its effect via another alternative pathway in which the second kinase is involved.

Accordingly, in further Embodiments 2.33 to 2.30, the invention also provides:

2.33 A compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 for use in preventing or treating cancers which are sensitive to PLK1 kinase inhibition.

2.34 A compound for use according to Embodiment 2.33 wherein the cancer which is sensitive to PLK1 kinase inhibition is a cancer which contains a KRAS mutation.

2.35 A compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 for use in treating a cancer in a patient who has been screened and has been determined as suffering from a cancer which is sensitive to PLK1 kinase inhibition.

2.36 A compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 for use in treating a cancer in a patient who has been screened and has been determined as suffering from a cancer which is sensitive to PLK1 kinase inhibition and has a KRAS mutation.

2.37 A method of treating a subject suffering from a cancer which is sensitive to PLK1 inhibition, which method comprises administering to the subject an effective amount of a compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77.

2.38 A method of treating a subject who has been diagnosed and has been found to be suffering from a cancer which is sensitive to PLK1 kinase inhibition, which method comprises administering to the subject an effective amount of a compound of the formula (1 ) as defined in any one of Embodiments 1 .1 to 1.77.

2.39 A method of treating a subject who has been diagnosed and has been found to be suffering from a cancer which is sensitive to PLK1 kinase inhibition and contains a KRAS mutation, which method comprises administering to the subject an effective amount of a compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77. 2.40 A method for the diagnosis and treatment of a disease state or condition mediated by PLK1 kinase which method comprises (i) screening a patient to determine whether a disease or condition from which the patient is or may be suffering is one which would be susceptible to treatment with a compound having activity against PLK1 kinase; and (ii) where it is indicated that the disease or condition from which the patient is thus susceptible, thereafter administering to the patient a compound as defined in any one of Embodiments 1.1 to 1.77.

2.41 The use of a compound as defined in any one of Embodiments 1 .1 to 1 .77 for the manufacture of a medicament for the treatment or prophylaxis of a disease state or condition in a patient who has been screened and has been determined as suffering from, or being at risk of suffering from, a disease or condition which would be susceptible to treatment with a compound having activity against PLK1 kinase.

2.42 A compound as defined in any one of Embodiments 1 .1 to 1 .77 for use in the treatment or prophylaxis of a disease state or condition in a patient who has been screened and has been determined as suffering from, or being at risk of suffering from, a disease or condition which would be susceptible to treatment with a compound having activity against PLK1 kinase.

2.43 A method for the diagnosis and treatment of a disease state or condition characterised by up-regulation of PLK1 kinase or the presence of a mutated form of PLK1 kinase, which method comprises (i) screening a patient to determine whether a disease or condition from which the patient is or may be suffering is one which would be susceptible to treatment with a compound having activity against PLK1 kinase; and (ii) where it is indicated that the disease or condition from which the patient is thus susceptible, thereafter administering to the patient a compound as defined in any one of Embodiments 1.1 to 1.77.

2.44 A method for the treatment of a disease state or condition characterised by up- regulation of PLK1 kinase or the presence of a mutated form of PLK1 , which method comprises administering a therapeutically effective amount of a compound as defined in any one of Embodiments 1 .1 to 1.77 to a patient who has been screened and has been determined as suffering from, or being at risk of suffering from, a disease or condition which would be susceptible to treatment with a compound having activity against PLK1 kinase. 2.45 The use of a compound as defined in any one of Embodiments 1 .1 to 1 .77 for the manufacture of a medicament for the treatment or prophylaxis of a disease, condition or disorder as defined in any one of Embodiments 2.1 to 2.37 in a patient who has been screened and has been determined as suffering from, or being at risk of suffering, from a said disease, condition or disorder which would be susceptible to treatment with a compound having activity against PLK1 kinase.

2.46 A compound as defined in any one of Embodiments 1 .1 to 1 .77 for use in the treatment or prophylaxis of a disease or condition or disorder in a patient who has been screened and has been determined as suffering from, or being at risk of suffering from, a disease, condition or disorder as defined in any one of Embodiments 2.1 to 2.45 which would be susceptible to treatment with a compound having activity against PLK1 kinase.

2.47 A compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 for use in preventing or treating cancers which are sensitive to both Wee1 and PLK1 kinase inhibition.

2.48 A compound for use according to Embodiment 2.33 wherein the cancer which is sensitive to Wee1 and PLK1 kinase inhibition is a cancer which contains a KRAS mutation.

2.49 A compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 for use in treating a cancer in a patient who has been screened and has been determined as suffering from a cancer which is sensitive to Wee1 and PLK1 kinase inhibition.

2.50 A compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .77 for use in treating a cancer in a patient who has been screened and has been determined as suffering from a cancer which is sensitive to Wee1 and PLK1 kinase inhibition and has a KRAS mutation.

2.51 A method of treating a subject suffering from a cancer which is sensitive to Wee1 and PLK1 inhibition, which method comprises administering to the subject an effective amount of a compound of the formula (1 ) as defined in any one of Embodiments 1 .1 to 1.77.

2.52 A method of treating a subject who has been diagnosed and has been found to be suffering from a cancer which is sensitive to both Wee1 and PLK1 kinase inhibition, which method comprises administering to the subject an effective amount of a compound of the formula (1 ) as defined in any one of Embodiments 1 .1 to 1.77.

2.53 A method of treating a subject who has been diagnosed and has been found to be suffering from a cancer which is sensitive to both Wee1 and PLK1 kinase inhibition and contains a KRAS mutation, which method comprises administering to the subject an effective amount of a compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1.77.

2.54 A method for the diagnosis and treatment of a disease state or condition mediated by Wee1 and PLK1 kinase which method comprises (i) screening a patient to determine whether a disease or condition from which the patient is or may be suffering is one which would be susceptible to treatment with a compound having activity against both Wee1 and PLK1 kinase; and (ii) where it is indicated that the disease or condition from which the patient is thus susceptible, thereafter administering to the patient a compound as defined in any one of Embodiments 1.1 to 1.77.

2.55 The use of a compound as defined in any one of Embodiments 1 .1 to 1 .77 for the manufacture of a medicament for the treatment or prophylaxis of a disease state or condition in a patient who has been screened and has been determined as suffering from, or being at risk of suffering from, a disease or condition which would be susceptible to treatment with a compound having activity against Wee1 and PLK1 kinase.

2.56 A compound as defined in any one of Embodiments 1 .1 to 1 .77 for use in the treatment or prophylaxis of a disease state or condition in a patient who has been screened and has been determined as suffering from, or being at risk of suffering from, a disease or condition which would be susceptible to treatment with a compound having activity against Wee1 and PLK1 kinase.

2.57 A method for the diagnosis and treatment of a disease state or condition characterised by up-regulation of Wee1 and/or PLK1 kinase and/or the presence of a mutated form of Wee1 and/or PLK1 kinase, which method comprises (i) screening a patient to determine whether a disease or condition from which the patient is or may be suffering is one which would be susceptible to treatment with a compound having activity against Wee1 and PLK1 kinase; and (ii) where it is indicated that the disease or condition from which the patient is thus susceptible, thereafter administering to the patient a compound as defined in any one of Embodiments 1 .1 to 1 .77. 2.58 A method for the treatment of a disease state or condition characterised by up- regulation of Wee1 and/or PLK1 kinase and/or the presence of a mutated form of Wee1 and/or PLK1 , which method comprises administering a therapeutically effective amount of a compound as defined in any one of Embodiments 1 .1 to 1 .77 to a patient who has been screened and has been determined as suffering from, or being at risk of suffering from, a disease or condition which would be susceptible to treatment with a compound having activity against Wee1 and PLK1 kinase.

2.59 The use of a compound as defined in any one of Embodiments 1 .1 to 1 .77 for the manufacture of a medicament for the treatment or prophylaxis of a disease, condition or disorder as defined in any one of Embodiments 2.1 to 2.58 in a patient who has been screened and has been determined as suffering from, or being at risk of suffering, from a said disease, condition or disorder which would be susceptible to treatment with a compound having activity against Wee1 and PLK1 kinase.

2.60 A compound as defined in any one of Embodiments 1 .1 to 1 .77 for use in the treatment or prophylaxis of a disease or condition or disorder in a patient who has been screened and has been determined as suffering from, or being at risk of suffering from, a disease, condition or disorder as defined in any one of Embodiments 2.1 to 2.59 which would be susceptible to treatment with a compound having activity against Wee1 and PLK1 kinase.

The diagnostic methods used to determine whether a particular cancer is susceptible to treatment with the compounds of the invention can be as described below in the section headed “Methods of Diagnosis”.

Determination of biological properties

The ability of the compounds of Embodiments 1 .1 to 1 .77 to inhibit cell proliferation can also be determined using the protocols set out in the Examples section below.

Preferred compounds of Embodiments 1.1 to 1 .77 are those having an IC 5 o against Wee1 kinase of less than 5 mM, or less than 1 mM and preferably less than 0.1 mM.

Accordingly, in further embodiments (Embodiments 2.61 to 2.64), the invention provides:

2.61 A compound according to any one of Embodiments 1 .1 to 1 .77 having an IC 5 o against Wee1 kinase of less than 5 mM. 2.62 A compound according to any one of Embodiments 1 .1 to 1 .77 having an IC 5 o against Wee1 kinase of or less than 1 mM.

2.63 A compound according to any one of Embodiments 1 .1 to 1 .77 having an IC 5 o against Wee1 kinase of less than 0.1 mM.

2.64 A compound according to any one of Embodiments 2.61 to 2.63 for use in a therapy, treatment, method or use according to any one of Embodiments 2.1 to 2.60.

Other preferred compounds are those having an IC 5 o against PLK1 of less than 5 mM, or less than 1 mM and preferably less than 0.1 mM.

Accordingly, in further embodiments (Embodiments 2.65 to 2.72), the invention provides:

2.65 A compound according to any one of Embodiments 1 .1 to 1 .77 having an IC 5 o against PLK1 kinase of less than 5 mM.

2.66 A compound according to any one of Embodiments 1 .1 to 1 .77 having an IC 5 o against PLK1 kinase of or less than 1 mM.

2.67 A compound according to any one of Embodiments 1 .1 to 1 .77 having an IC 5 o against PLK1 kinase of less than 0.1 mM.

2.68 A compound according to any one of Embodiments 2.65 to 2.67 for use in a therapy, treatment, method or use according to any one of Embodiments 2.1 to 2.60.

2.69 A compound according to any one of Embodiments 2.65 to 2.67 having an IC 5 o against Wee1 kinase of less than 5 mM.

2.70 A compound according to any one of Embodiments 2.65 to 2.67 having an IC 5 o against Wee1 kinase of or less than 1 mM.

2.71 A compound according to any one of Embodiments 2.65 to 2.67 having an IC 5 o against Wee1 kinase of less than 0.1 mM.

2.72 A compound according to any one of Embodiments 2.69 to 2.71 for use in a therapy, treatment, method or use according to any one of Embodiments 2.1 to 2.60.

Methods for the Preparation of Compounds of the Invention The invention also provides methods for the preparation of a compound of the formula (1 )·

Accordingly, in another embodiment (Embodiment 3.1 ), the invention provides a method for preparing a compound as defined in any one of Embodiments 1.1 to 1 .77 which method comprises the reaction of a compound of the formula (10): or a protected form thereof, wherein Hal is a halogen such as bromine or chlorine, with a compound of the formula (11 ): or a protected form thereof; and thereafter optionally: a) removing any protecting group present; and/or b) interconverting a compound of formula (1 ) or a protected derivative thereof to a further compound of formula (1 ) or a protected derivative thereof and optionally removing any protecting group present. The reaction between the compounds of formulae (10) and (11 ) may be carried out in the presence of a strong but non-nucleophilic base. Examples of suitable bases are lithium amide bases, such as lithium bis(trimethylsilyl)amide (Li-HDMS) or lithium diisopropylamide (LDA) The reaction is typically carried out in a polar, aprotic solvent (such as tetrahydrofuran). After initial mixing at a relatively low temperature (typically a temperature of 10°C or less, preferably 5°C or less, for example at approximately 0°C), the reaction mixture may then be heated to a higher temperature of 80°C or more, preferably at least 90°C, for example at approximately 100°C.

Alternatively, the reaction between the compounds of formulae (10) and (11) may be carried out under Buchwald-Hartwig amination conditions in the presence of a palladium catalyst, a ligand and a base. The palladium catalyst may be Pd2(dba)3 and the ligand may be 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene. Typically, the base is a non- nucleophilic base (such as an alkoxide base, e.g. sodium tert-butoxide). The reaction may be carried out in a polar solvent such as dimethyl formamide (DMF) or dioxane, and the reaction mixture is typically subjected to heating, for example to a temperature in excess of 90 e C and for a time period of 12 hours or more.

Compounds of formula (10) can be prepared by the reaction of a compound of formula (12): wherein Hal is a halogen (e.g. chlorine) with a compound of formula (13): wherein X is selected from halogen, a boronic acid/ester group (e.g. B(OH) 2 ) or a trialkyl tin group (e.g. Sn(CH 3 ) 3 ).

The reaction between the compounds of formulae (12) and (13) typically takes place in the presence of a coupling catalyst. When X is a boronic acid/ester group, the catalyst may be a copper catalyst (e.g. copper (II) acetate) or a palladium (0) catalyst. The reaction is typically carried out in the presence of a non-nucleophilic base (such as pyridine) and is conducted at room temperature. When X is halogen (e.g. bromine), the catalyst may be a copper (I) catalyst (e.g. copper iodide). A diamine ligand, such as trans-N,N'-dimethylcyclohexane-1 ,2-diamine is typically also present. The reaction may be conducted at a temperature above 90°C and for a time period of over 1 hour.

For compounds of the formula (1 ) where R 4 is cyano, an analogue of the compound of formula (12), but wherein R 4 is bromine, may be used as a starting material. The bromo- analogue is reacted with the compound of formula (13) under the general conditions described above to give the corresponding bromo-analogue (R 4 is Br) of the compound of formula (10). Subsequent reaction with the compound of formula (11) gives an analogue of the compound of formula (1) wherein R 4 is bromine.

The bromo-analogue of the compound of formula (1) can then be converted to a compound of the formula (1 ) where R 4 is cyano by reaction with a cyanide salt (e.g. Zn(CN) 2 ) in the presence of a palladium catalyst (such as Pd 2 (dba)3) and a suitable ligand (such as 1 ,1'-bis(diphenylphosphino)ferrocene). The reaction is typically carried out in a polar, aprotic solvent (such as tetrahydrofuran) and may be subject to heating at temperatures above 150°C for a period of 20 minutes or greater.

Once formed, one compound of the formula (1 ), or a protected derivative thereof, can be converted into another compound of the formula (1) by methods well known to the skilled person. Examples of synthetic procedures for converting one functional group into another functional group are set out in standard texts such as Advanced Organic Chemistry, by Jerry March, 4 th edition, 119, Wiley Interscience, New York; Fiesers' Reagents for Organic Synthesis, Volumes 1 -17, John Wiley, edited by Mary Fieser (ISBN: 0-471 -58283-2); and Organic Syntheses, Volumes 1 -8, John Wiley, edited by Jeremiah P. Freeman (ISBN: 0-471 -31192-8)).

Examples of a number of interconversions of compounds of formula (1) are described below.

A compound of formula (1) wherein R 6 is hydrogen can be converted to a compound of formula (1) wherein R 6 is chlorine by reaction with an electrophilic source of chlorine (such as N-chlorosuccinimide) in a polar, chlorinated solvent (as such dichloromethane). The reaction is typically conducted at a temperature of 25°C or less and for a period of 9 hours. A compound of formula (1 ) wherein Cyc 1 is a piperazine or piperidine group be converted to the corresponding compound wherein Cyc 1 is an acylated piperazine or piperidine by reaction with an acylating agent (e.g. an anhydride of a carboxylic acid or an acyl chloride) in the presence of a weakly basic solvent (e.g. pyridine).

In many of the reactions described above, it may be necessary to protect one or more groups to prevent reaction from taking place at an undesirable location on the molecule. Examples of protecting groups, and methods of protecting and deprotecting functional groups, can be found in Protective Groups in Organic Synthesis (T. Green and P. Wuts; 3rd Edition; John Wiley and Sons, 1999).

Compounds made by the foregoing methods may be isolated and purified by any of a variety of methods well known to those skilled in the art and examples of such methods include recrystallisation and chromatographic techniques such as column chromatography (e.g. flash chromatography) and HPLC.

Pharmaceutical Formulations

The compounds of the invention are typically administered to patients in the form of a pharmaceutical composition. Accordingly, in another Embodiment of the invention (Embodiment 4.1 ), the invention provides a pharmaceutical composition comprising a compound according to any one of Embodiments 1.1 to 1 .77 and a pharmaceutically acceptable excipient.

In further embodiments, there are provided:

4.2 A pharmaceutical composition according to Embodiment 4.1 which comprises from approximately 1% (w/w) to approximately 95% (w/w) of a compound of any one of Embodiments 1 .1 to 1 .77 and from 99% (w/w) to 5% (w/w) of a pharmaceutically acceptable excipient or combination of excipients and optionally one or more further therapeutically active ingredients.

4.3 A pharmaceutical composition according to Embodiment 4.2 which comprises from approximately 5% (w/w) to approximately 90% (w/w) of a compound of any one of Embodiments 1 .1 to 1 .77 and from 95% (w/w) to 10% of a pharmaceutically excipient or combination of excipients and optionally one or more further therapeutically active ingredients. 4.4 A pharmaceutical composition according to Embodiment 4.3 which comprises from approximately 10% (w/w) to approximately 90% (w/w) of a compound of any one of Embodiments 1 .1 to 1 .77 and from 90% (w/w) to 10% of a pharmaceutically excipient or combination of excipients.

4.5 A pharmaceutical composition according to Embodiment 4.4 which comprises from approximately 20% (w/w) to approximately 90% (w/w) of a compound of any one of Embodiments 1 .1 to 1 .77 and from 80% (w/w) to 10% of a pharmaceutically excipient or combination of excipients.

4.6 A pharmaceutical composition according to Embodiment 4.5 which comprises from approximately 25% (w/w) to approximately 80% (w/w) of a compound of any one of Embodiments 1 .1 to 1 .77 and from 75% (w/w) to 20% of a pharmaceutically excipient or combination of excipients.

The pharmaceutical compositions of the invention can be in any form suitable for oral, parenteral, topical, intranasal, intrabronchial, ophthalmic, otic, rectal, intra-vaginal, or transdermal administration. Where the compositions are intended for parenteral administration, they can be formulated for intravenous, intramuscular, intraperitoneal, subcutaneous administration or for direct delivery into a target organ or tissue by injection, infusion or other means of delivery.

Pharmaceutical dosage forms suitable for oral administration include tablets, capsules, caplets, pills, lozenges, syrups, solutions, sprays, powders, granules, elixirs and suspensions, sublingual tablets, sprays, wafers or patches and buccal patches.

Accordingly, in further embodiments, the invention provides:

4.7 A pharmaceutical composition according to any one of Embodiments 4.1 to 4.6 which is suitable for oral administration.

4.8 A pharmaceutical composition according to Embodiment 4.7 which is selected from tablets, capsules, caplets, pills, lozenges, syrups, solutions, sprays, powders, granules, elixirs and suspensions, sublingual tablets, sprays, wafers or patches and buccal patches.

4.9 A pharmaceutical composition according to Embodiment 4.8 which is selected from tablets and capsules. 4.10 A pharmaceutical composition according to any one of Embodiments 4.1 to 4.6 which is suitable for parenteral administration.

4.11 A pharmaceutical composition according to Embodiment 4.10 which is formulated for intravenous, intramuscular, intraperitoneal, subcutaneous administration or for direct delivery into a target organ or tissue by injection, infusion or other means of delivery.

4.12 A pharmaceutical composition according to Embodiment 4.11 which is a solution or suspension for injection or infusion.

Pharmaceutical compositions (e.g. as defined in any one of Embodiments 4.1 to 4.12) containing a compound according to Embodiments 1 .1 to 1.77 of the invention can be formulated in accordance with known techniques, see for example, Remington’s Pharmaceutical Sciences, Mack Publishing Company, Easton, PA, USA.

Thus, tablet compositions (as in Embodiment 4.9) can contain a unit dosage of active compound together with an inert diluent or carrier such as a sugar or sugar alcohol, e.g.; lactose, sucrose, sorbitol or mannitol; and/or a non-sugar derived diluent such as sodium carbonate, calcium phosphate, talc, calcium carbonate, or a cellulose or derivative thereof such as methyl cellulose, ethyl cellulose, hydroxypropyl methyl cellulose, and starches such as corn starch. Tablets may also contain such standard ingredients as binding and granulating agents such as polyvinylpyrrolidone, disintegrants (e.g. swellable crosslinked polymers such as crosslinked carboxymethylcellulose), lubricating agents (e.g. stearates), preservatives (e.g. parabens), antioxidants (e.g. BHT), buffering agents (for example phosphate or citrate buffers), and effervescent agents such as citrate/bicarbonate mixtures. Such excipients are well known and do not need to be discussed in detail here.

Capsule formulations (as in Embodiment 4.9) may be of the hard gelatin or soft gelatin variety and can contain the active component in solid, semi-solid, or liquid form. Gelatin capsules can be formed from animal gelatin or synthetic or plant-derived equivalents thereof.

The solid dosage forms (e.g.; tablets, capsules etc.) can be coated or un-coated, but typically have a coating, for example a protective film coating (e.g. a wax or varnish) or a release controlling coating. The coating (e.g. a Eudragit ™ type polymer) can be designed to release the active component at a desired location within the gastro -intestinal tract. Thus, the coating can be selected so as to degrade under certain pH conditions within the gastrointestinal tract, thereby selectively release the compound in the stomach or in the ileum or duodenum.

Instead of, or in addition to, a coating, the drug can be presented in a solid matrix comprising a release controlling agent, for example a release delaying agent which may be adapted to selectively release the compound under conditions of varying acidity or alkalinity in the gastrointestinal tract. Alternatively, the matrix material or release retarding coating can take the form of an erodible polymer (e.g. a maleic anhydride polymer) which is substantially continuously eroded as the dosage form passes through the gastrointestinal tract.

Compositions for topical use include ointments, creams, sprays, patches, gels, liquid drops and inserts (for example intraocular inserts). Such compositions can be formulated in accordance with known methods.

Compositions for parenteral administration (as in Embodiments 4.10 to 4.12) are typically presented as sterile aqueous or oily solutions or fine suspensions, or may be provided in finely divided sterile powder form for making up extemporaneously with sterile water for injection.

Examples of formulations for rectal or intra-vaginal administration include pessaries and suppositories which may be, for example, formed from a shaped mouldable or waxy material containing the active compound.

Compositions for administration by inhalation may take the form of inhalable powder compositions or liquid or powder sprays, and can be administrated in standard form using powder inhaler devices or aerosol dispensing devices. Such devices are well known. For administration by inhalation, the powdered formulations typically comprise the active compound together with an inert solid powdered diluent such as lactose.

The compounds of the inventions will generally be presented in unit dosage form and, as such, will typically contain sufficient compound to provide a desired level of biological activity. For example, according to any one of Embodiments 4.1 to 4.9), a composition intended for oral administration may contain from 2 milligrams to 200 milligrams of active ingredient, more usually from 10 milligrams to 100 milligrams, for example, 12.5 milligrams, 25 milligrams and 50 milligrams.

Combination Therapy It is envisaged that the compounds of Embodiments 1.1 to 1 .77 will be useful either as sole chemotherapeutic agents or, more usually, in combination therapy with chemotherapeutic agents or radiation therapy in the prophylaxis or treatment of a range of proliferative disease states or conditions. Examples of such disease states and conditions are set out above.

Particular examples of chemotherapeutic agents that may be co-administered with the compounds of Embodiments 1.1 to 1 .77 include:

• Topoisomerase I inhibitors e.g. Topotecan, Irinotecan

• Antimetabolites e.g. 5-fluoro uracil (5-FU)

• Tubulin targeting agents

• Topoisomerase II inhibitors e.g. etoposide

• EGFR inhibitors (e.g. Gefitinib - see Biochemical Pharmacology 782009 460- 468)

• mTOFt inhibitors (e.g. Everolimus)

• PI3K pathway inhibitors (e.g. PI3K, PDK1)

• Akt inhibitors

• Alkylating Agents (e.g. temozolomide, cyclophosphamide)

• Monoclonal Antibodies (e.g. antibodies targeting CTLA-4, PD-1 , PD-L1 , 0X40, CD52 or CD20). Examples include nivolumab, pembrolizumab, avelumab, durvalumab, atezolizumab)

• Anti-Hormones

• Signal Transduction Inhibitors

• Proteasome Inhibitors

• DNA methyl transferases

• Cytokines and retinoids

• Hypoxia triggered DNA damaging agents (e.g. Tirapazamine)

• Aromatase inhibitors

• Anti Her2 antibodies (for example, those described in WO 2007/056118) • Inhibitors of angiogenesis

• HDAC inhibitors

• MEK inhibitors

• B-Raf inhibitors · ERK inhibitors

• HER2 small molecule inhibitors (e.g. lapatinib)

• Bcr-Abl tyrosine-kinase inhibitors (e.g. imatinib)

• CDK4/6 inhibitors e.g. Ibrance

• VEGFR inhibitors · IGFR-1 inhibitors

• Inhibitors of the Hedgehog signalling pathway

• PARP inhibitors e.g. Olaparib

• Immune Checkpoint inhibitors

• Gemcitabine

Further examples of chemotherapeutic agents that may be co-administered with a compound as defined in any one Embodiments 1.1 to 1.77 include:

• Tore 1 inhibitors

• Taxanes (e.g. paclitaxel, docetaxel, cabazitaxel)

• Platinum agents (e.g. cisplatin, carboplatin, oxaliplatin)

• Anthracyclines (e.g. Doxorubicin)

• Inhibitors of Bcl-2 family proteins e.g. ABT263 (navitoclax), a Bcl-2/Bcl-extra large (Bcl-xL) inhibitor

• Cytarabine

Belinostat One particular combination comprises a compound according to any one of Embodiments 1 .1 to 1 .77 together with a PARP inhibitor, such as niraparib, olaparib, rucaparib or talazoparib.

A further particular combination comprises a compound according to any one of Embodiments 1 .1 to 1 .77 together with a DNA damaging agent, such as gemcitabine. cisplatin, oxaliplatin, carboplatin, cyclophosphamide, chlorambucil or temozolomide.

Yet a further particular combination a compound according to any one of Embodiments 1 .1 to 1 .77 together with an immune checkpoint inhibitor, e.g. a PD-1 or a PD-L1 inhibitor, such as nivolumab, pembrolizumab, avelumab, durvalumab or atezolizumab.

The compounds may also be administered in conjunction with radiotherapy.

Posoloqy

The compounds may be administered over a prolonged term to maintain beneficial therapeutic effects or may be administered for a short period only. Alternatively, they may be administered in a pulsatile or continuous manner.

The compounds of the invention will be administered in an effective amount, i.e. an amount which is effective to bring about the desired therapeutic effect. For example, the "effective amount" can be a quantity of compound which, when administered to a subject suffering from cancer, slows tumour growth, ameliorates the symptoms of the disease and/or increases longevity.

The amount of Wee1 inhibitor compound of the invention administered to the subject will depend on the type and severity of the disease or condition and on the characteristics of the subject, such as general health, age, sex, body weight and tolerance to drugs. The skilled person will be able to determine appropriate dosages depending on these and other factors.

The compounds are generally administered to a subject in need of such administration, for example a human or animal subject (patient), preferably a human.

A typical daily dose of the compound of any of Embodiments 1.1 to 1 .77 can be in the range from 100 picograms to 100 milligrams per kilogram of body weight, more typically 5 nanograms to 25 milligrams per kilogram of body weight, and more usually 10 nanograms to 15 milligrams per kilogram of body weight (e.g. 10 nanograms to 10 milligrams, and more typically 1 microgram per kilogram to 20 milligrams per kilogram, for example 1 microgram to 10 milligrams per kilogram) although higher or lower doses may be administered where required. The compound can be administered on a daily basis or on a repeat basis every 2, or 3, or 4, or 5, or 6, or 7, or 10 or 14, or 21 , or 28 days for example.

In one particular dosing schedule, a patient will be given an infusion of a compound for periods of one hour daily for up to ten days in particular up to five days for one week, and the treatment repeated at a desired interval such as two to four weeks, in particular every three weeks.

More particularly, a patient may be given an infusion of a compound for periods of one hour daily for 5 days and the treatment repeated every three weeks.

In another particular dosing schedule, a patient is given an infusion over 30 minutes to 1 hour followed by maintenance infusions of variable duration, for example 1 to 5 hours, e.g. 3 hours.

In a further particular dosing schedule, a patient is given a continuous infusion for a period of 12 hours to 5 days, and in particular a continuous infusion of 24 hours to 72 hours.

Ultimately, however, the quantity of compound administered and the type of composition used will be commensurate with the nature of the disease or physiological condition being treated and will be at the discretion of the physician.

Methods of Diagnosis

Prior to administration of a compound of any one of Embodiments 1.1 to 1.77, a patient may be screened to determine whether a disease or condition from which the patient is or may be suffering is one which would be susceptible to treatment with a compound having activity against Wee1 or PLK1 kinase. Such patient can then be treated according to the methods described above.

For example, a biological sample taken from a patient may be analysed to determine whether a condition or disease, such as cancer, that the patient is or may be suffering from is one which is characterised by a genetic abnormality or abnormal protein expression which leads to up-regulation of Wee1/PLK1 kinase or to sensitisation of a pathway to normal Wee1/PLK1 kinase activity or to over-expression of Wee1/PLK1 kinase. The term up-regulation includes elevated expression or over-expression, including gene amplification (i.e. multiple gene copies) and increased expression by a transcriptional effect, and hyperactivity and activation, including activation by mutations. Thus, the patient may be subjected to a diagnostic test to detect a marker characteristic of up-regulation of Wee1/PL1 kinase. The term diagnosis includes screening. By marker we include genetic markers including, for example, the measurement of DNA composition to identify mutations of Wee1/PLK1 . The term marker also includes markers which are characteristic of up-regulation of Wee1/PLK1 , including enzyme activity, enzyme levels, enzyme state (e.g. phosphorylated or not) and mRNA levels of the aforementioned proteins.

Tumours with upregulation of Wee1/PLK1 kinase may be particularly sensitive to Wee1/PLK1 inhibitors. Tumours may preferentially be screened for upregulation of Wee1/PLK1. Thus, the patient may be subjected to a diagnostic test to detect a marker characteristic of up-regulation of Wee1/PL1 . The diagnostic tests are typically conducted on a biological sample selected from tumour biopsy samples, blood samples (isolation and enrichment of shed tumour cells), stool biopsies, sputum, chromosome analysis, pleural fluid, peritoneal fluid,

Methods of identification and analysis of mutations and up-regulation of proteins are known to a person skilled in the art. Screening methods could include, but are not limited to, standard methods such as reverse-transcriptase polymerase chain reaction (RT-PCR) or in-situ hybridisation.

In screening by RT-PCR, the level of mRNA in the tumour is assessed by creating a cDNA copy of the mRNA followed by amplification of the cDNA by PCR. Methods of PCR amplification, the selection of primers, and conditions for amplification, are known to a person skilled in the art. Nucleic acid manipulations and PCR are carried out by standard methods, as described for example in Ausubel, F.M. et al., eds. Current Protocols in Molecular Biology, 2004, John Wiley & Sons Inc., or Innis, M.A. etal., eds. PCR Protocols: a guide to methods and applications, 1990, Academic Press, San Diego. Reactions and manipulations involving nucleic acid techniques are also described in Sambrook et al., 2001 , 3 rd Ed, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press. Alternatively a commercially available kit for RT-PCR (for example Roche Molecular Biochemicals) may be used, or methodology as set forth in United States patents 4,666,828; 4,683,202; 4,801 ,531 ; 5,192,659, 5,272,057, 5,882,864, and 6,218,529 and incorporated herein by reference.

An example of an in-situ hybridisation technique for assessing mRNA expression would be fluorescence in-situ hybridisation (FISH) (see Angerer, 1987 Meth. Enzymol., 152: 649).

Generally, in situ hybridization comprises the following major steps: (1) fixation of tissue to be analyzed; (2) pre-hybridization treatment of the sample to increase accessibility of target nucleic acid, and to reduce nonspecific binding; (3) hybridization of the mixture of nucleic acids to the nucleic acid in the biological structure or tissue; (4) post-hybridization washes to remove nucleic acid fragments not bound in the hybridization, and (5) detection of the hybridized nucleic acid fragments. The probes used in such applications are typically labelled, for example, with radioisotopes or fluorescent reporters. Preferred probes are sufficiently long, for example, from about 50, 100, or 200 nucleotides to about 1000 or more nucleotides, to enable specific hybridization with the target nucleic acid(s) under stringent conditions. Standard methods for carrying out FISH are described in Ausubel, F.M. et al., eds. Current Protocols in Molecular Biology, 2004, John Wiley & Sons Inc and Fluorescence In Situ Hybridization: Technical Overview by John M. S. Bartlett in Molecular Diagnosis of Cancer, Methods and Protocols, 2nd ed.; ISBN: 1 - 59259-760-2; March 2004, pps. 077-088; Series: Methods in Molecular Medicine.

Alternatively, the protein products expressed from the mRNAs may be assayed by immunohistochemistry of tumour samples, solid phase immunoassay with microtiter plates, Western blotting, 2-dimensional SDS-polyacrylamide gel electrophoresis, ELISA, flow cytometry and other methods known in the art for detection of specific proteins. Detection methods would include the use of site-specific antibodies. The skilled person will recognize that all such well-known techniques for detection of up-regulation of Wee1 kinase could be applicable in the present case.

EXAMPLES

EXAMPLES 1 TO 57

The compounds of Examples 1 to 57 in Table 1 below are illustrative of the invention.

Analytical data 1H NMR spectra were recorded on a Bruker 400 machine.

LCMS methods: LCMS analysis was carried using the following method(s): LCMS method A:

LCMS was carried out on a UPLC AQUITY device with Photodiode Array (PDA) detector and QDA Mass Detector using an X-BRIDGE C18, 50 x 2.1 mm, 1 .6 gm column. Column flow was 0.8 mL/min and the mobile phase used was: (A) 0.1% Formic acid in milli-Q water (B) 0.1% formic acid in milli-Q water: acetonitrile (10:90). The following gradient was applied:

LCMS method B: LCMS was carried out on a UPLC AQUITY device with Photodiode Array (PDA) detector and QDA Mass Detector using an X-BRIDGE C18, 50 x 2.1 mm, 1 .6 gm column. Column flow was 0.8 mL/min and the mobile phase used was: (A) 0.1% Formic acid in milli-Q water (B) 0.1% formic acid in milli-Q water: acetonitrile (10:90). The following gradient was applied:

LCMS method C:

LCMS was carried out on a UPLC AQUITY device with Photodiode Array (PDA) detector and QDA Mass Detector using an X-BRIDGE C18, 50 x 4.6 mm, 3.5 pm column. Column flow was 1 .0 mL/min and the mobile phase used was: (A) 0.1% Formic acid in milli-Q water (B) 100% acetonitrile. The following gradient was applied:

LCMS method D:

LCMS was carried out on a Waters Alliance 2690 device coupled with 996 PDA and Micromass ZQ detectors using a C18, 50 x 4.6 mm, 3.5 pm column. Column flow was 1 .2 mL/min and the mobile phase used was: (A) 10mM Ammonium Bicarbonate in Milli-Q water and (B) Methanol. The following gradient was applied:

LCMS method E:

The same conditions were used as for LCMS method D except that the following gradient was used for the elution:

LCMS method F:

LCMS was carried out on a UPLC AQUITY device with Photodiode Array (PDA) detector and QDA Mass Detector using an X-BRIDGE C18, 50 x 4.6 mm, 5 pm column. Column flow was 1 .0 mL/min and the mobile phase used was: (A) 5mM Ammonium Bicarbonate in Water (B) Methanol. The following gradient was applied:

LCMS method G: The same conditions were used as for LCMS method D except that the following gradient was used for the elution:

LCMS method H:

LCMS was carried out on a YMC 50 x 2.9 mm, 1 .9 micron column. Column flow was 1.2 mL/min and the mobile phase used was as follows: (A) 0.1% Formic acid in milli-Q water (B) 0.1% Formic acid in milli-Q water: Acetonitrile (10:90). The following gradient was applied: LCMS method I: LCMS was carried out using a Waters Alliance 2690 device with linked 996 PDA detector and Micro mass ZQ. The C18column employed measured 50 x 4.6 mm, with 3.5 micron particle size. Column flow was 1 .0 mL/min and mobile phase used was as follows: (A) 5mM ammonium bicarbonate in Milli-Q water and (B) Methanol. The following gradient was used for elution:

LCMS method J:

LCMS was carried out on an ACQUITY UPLC device using a BEH 2.1 x 50mm, 1 .6 micron, C18 column. Column flow was 1.2 mL/min and mobile phase used was: (A) 0.1 % Formic acid in Milli Q water (pH= 2.70) (B) 0.1% formic acid in water: acetonitrile (10:90).

The following gradient was used for the elution.

Chiral analytical HPLC method A:

CHIRAL HPLC was carried out using a CHROMEGA CHIRAL CCO, 250 x 4.6 mm, 5 micron column. Flow rate was 1 .0 mL/min and solvents used were: (A) 0.1% DEA in heptane and (B) isopropyl alcohol: methanol (70:30). An isocratic method was used for the elution:- Mobile phase A: Mobile Phase B = 80:20 for 70 min.

Chiral analytical HPLC method B:

Chiral HPLC was carried out using a YMC cellulose SC 150 x 4.6 mm, 5 micron column. Flow rate was 1 .0 mL/min. Solvents used were: (A) 0.1% DEA in heptane and (B) Isopropyl alcohol: acetonitrile (80:20) as mobile phase. An isocratic method was used for the elution:- Mobile phase A: Mobile Phase B = 80:20 for 40 min.

Chiral analytical HPLC method C:

The same conditions were used as for Chiral analytical HPLC method B except that mobile phase B was comprised of Isopropyl alcohol: acetonitrile in the ratio 90:10 rather than 80:20.

Chiral analytical HPLC method D:

The same conditions were used as for Chiral analytical HPLC method B except that mobile phase B was comprised of Isopropyl alcohol: acetonitrile in the ratio 70:30 rather than 80:20.

Chiral analytical HPLC method E:

Chiral HPLC was carried out using a CCJ-250 x 4.6 mm, 5 micron, C18 column with flow rate of 1 .0 mL/min. The mobile phase used was as follows: (A) 0.1% TFA in heptane and (B) IPA: Methanol (70:30). An isocratic method was used for the elution:- Mobile phase A: Mobile Phase B = 55:45 for 40 min.

Chiral analytical HPLC method F:

Chiral HPLC was carried out using a CCJ-250 x 4.6 mm, 5 micron, C18 column with a flow rate of 1 .0 mL/min. The mobile phase used was as follows: (A) 0.1% TFA in heptane and (B) IPA: acetonitrile (90:10). An isocratic method was used for the elution:- Mobile phase A: Mobile Phase B = 60:40 for 40 min.

Chiral analytical HPLC method G:

Chiral HPLC was carried out using a YMC SC cellulose 250 x 4.6 mm, 5 micron column with a flow rate of 1 .0 mL/min. Solvents used were: (A) 0.1 % v/v DEA in MeOH (B) ACN. An isocratic method was used for the elution:- Mobile phase A: Mobile Phase B = 50:50 for 20 min.

Chiral analytical HPLC method H: The same conditions were used as for Chiral analytical HPLC method G except that mobile phase B was comprised of Isopropyl alcohol: methanol in the ratio 70:30 rather than 50:50.

Chiral analytical HPLC method I: The same conditions were used as for Chiral analytical HPLC method G with the following exceptions: i) mobile phase A was comprised of 0.1% v/v DEA in Heptane ii) mobile phase B was comprised of IPA: Methanol (70:30) iii) duration of the run was 35 min.

Chiral analytical HPLC method J: Chiral HPLC was carried out using an Agilent Technologies 1200 Series device, employing a Chiralpak IG 250 x 4.6 mm, 5 micron column. Flow rate was 1 .0 mL/min. Solvents used were: (A) 0.1% v/v DEA in Heptane, (B) IPA: ACN (70:30). An isocratic method was used for the elution:- Mobile phase A: Mobile Phase B = 50:50 for 50 min.

Chiral analytical HPLC method K: Chiral HPLC was carried out using a YMC SC cellulose 250 x 4.6 mm, 5 micron column, with a flow rate of 1 .0 mL/min. Solvents used were: (A) 0.1 % v/v DEA in Heptane, (B) IPA-ACN (70-30). An isocratic method was used for the elution:- Mobile phase A: Mobile Phase B = 50:50 for 50 min.

Chiral analytical HPLC method L:

Chiral analytical HPLC method M:

Chiral HPLC was carried out using a YMC cellulose SC 250 x 4.6 mm, 5 micron column. Flow rate was 1 .0 mL/min. Solvents used were: (A) 0.1% ammonia in heptane and (B) isopropyl alcohol: methanol (70:30) as mobile phase. The following gradient was used for the elution: Chiral analytical HPLC method N:

Chiral HPLC was carried out using a YMC cellulose SC 250 x 4.6 mm, 5 micron column. Flow rate was 1 .0 mL/min. Solvents used were: (A) 0.1% DEA in heptane and (B) isopropyl alcohol: methanol (70:30) as mobile phase. An isocratic method was applied, using 45% A and 55% B for 40 min. Chiral preparative HPLC method 1 :

Purification was carried out using (A) 0.1% v/v TFA in heptane and (B) IPA: MeOH (70:30) as mobile phase, using a CCO-250 x 19 mm column (5 micron particle size) with a flow rate of 27.0 mL/min. An isocratic method was applied, using 75% A and 25% B.

Chiral preparative HPLC method 2: Purification was carried out on a WATERS instrument using a CHIRALPAK IG SFC, 21 X 250 mm, 5pm column. Column flow was 12.0 mL/min and solvents used were: (A) 0.1% triethylamine in methanol and (B) 0.1% triethylamine in acetonitrile. An isocratic method was applied, using 98% A and 2% B for 180 min.

Preparative HPLC method A: Purification was carried out using (A) 0.05% v/v HCI in water and (B) ACN: MeOH: IPA (65:25:10) as mobile phase, using a SUNFIRE C18 250 x 19 mm column (5 micron particle size) with a flow rate of 19.0 mL/min and the following gradient: Preparative HPLC method B:

Purification was carried out using (A) 0.05% v/v HCI in water and (B) 100% Acetonitrile as mobile phase, using a SUNFIRE C18 250 x 19 mm column, (5 micron particle size) with a flow rate of 20.0 mL/min and with the following gradient:

Preparative HPLC method C:

Purification was carried out using a YMC-Actus Triart Prep C18-S column, size: 250 x 20 mm, 5 micron particle size. Column flow was 25.0 mL/min and solvents used were: (A) 0.05% v/v HCI in water; and (B) 100% acetonitrile using the following gradient:

Preparative HPLC method D:

Purification was carried out using a YMC-Actus Triart Prep C18-S column, size: 250 x 20 mm, 5 micron particle size. Column flow was 28.0 mL/min and solvents used were: (A) 0.05% v/v HCI in water; and (B) acetonitrile: methanol: IPA (65:25:10) using the following gradient:

Preparative HPLC method E:

Purification was carried out using a SUNFIRE PREP C18 OBD column, measuring 19 x 250 mm, 5pm particle size. Column flow was 13.0 mL/min and solvents used were: (A) 0.05% v/v HCI in water; and (B) 100% ACN applying the following gradient:

Preparative HPLC method F:

Purification was carried out using an Xbridge Prep C18, 30 x 250 mm column, 5pm particle size. Flow rate was 28.0 mL/min and solvents used were: (A) 0.05% v/v HCI in water; and (B) 100% acetonitrile. The following gradient was applied:

Preparative HPLC method G: Purification was carried out using a YMC-Actus Triart Prep C18-S column, size: 250 x 20 mm, 5 micron particle size. Column flow was 15.0 mL/min and solvents used were: (A) 0.05% v/v HCI in water; and (B) 100% acetonitrile using the following gradient: Preparative HPLC method H:

Purification was carried out using a YMC-Actus Triart Prep C18-S column, size: 250 x 20 mm, 5 micron particle size. Column flow was 25.0 mL/min and solvents used were: (A) 0.05% v/v HCI in water; and (B) acetonitrile: methanol: IPA (65:25:10) using the following gradient:

Preparative HPLC method I:

Purification was carried out using a YMC-Actus Triart Prep C18-S column, size: 250 x 20 mm, 5 micron particle size. Column flow was 13.0 mL/min and solvents used were: (A) 0.05% v/v HCI in water; and (B) 100% acetonitrile using the following gradient:

Preparative HPLC method J:

Purification was carried out using a SUNFIRE PREP C18 OBD column, measuring 19 x 250 mm, 5pm particle size. Column flow was 12.0 mL/min and solvents used were: (A) 0.05% v/v HCI in water; and (B) 100% ACN applying the following gradient:

Preparative HPLC method K:

Purification was carried out using a SUNFIRE PREP C18 OBD column, measuring 19 x 250 mm, 5pm particle size. Column flow was 12.0 mL/min and solvents used were: (A) 0.05% v/v HCI in water; and (B) 100% ACN applying the following gradient:

Preparative HPLC method L:

Purification was carried out using a SUNFIRE PREP C18 OBD column, measuring 19 x 250 mm, 5pm particle size. Column flow was 12.0 mL/min and solvents used were: (A) 0.05% v/v HCI in water; and (B) 100% ACN applying the following gradient:

Preparative HPLC method M:

Purification was carried out using a YMC C18 120GM, 50 MICRON column. Column flow was 12.0 mL/min and solvents used were: (A) 0.05% v/v HCI in water; and (B) 100% acetonitrile applying the following gradient:

Preparative HPLC method N:

Purification was carried out on a WATERS 2489 instrument using a REPACK C18,250X 21 2mm, 5 pm column. Column flow was 22.0 mL/min and solvents used were: (A) 5mM ammonium bicarbonate in water (B) acetonitrile:methanol:isopropyl alcohol (75:25:10) applying the following gradient: Preparative HPLC method O:

Purification was carried out on a SHIMADZU instrument using a WELCH ULTISIL XB- C18-(250 x 21 2mm), 5pm column. Column flow was 20.0 mL/min and solvents used were: (A) 0.05% HCI in water and (B) 100% acetonitrile applying the following gradient:

Preparative HPLC method P:

Purification was carried out on a WATERS instrument using a SUNFIRE Prep C18 OBD, 19 x 250 mm, 5pm column. Column flow was 16.0 mL/min and solvents used were: (A)

0.05% HCI in water and (B) 100% acetonitrile applying the following gradient:

General Schemes

The following schemes were used to synthesize the compound examples of this invention:

General scheme 1

X= B(OH)2 or Sn(Me)3 orBr A = CH or N

General scheme 2

Optional deprotection step step 3

X= B(OH)2 or Sn(Me)3 or Br A = CH or N

General scheme 3

X

X= B(0H)2 or Sn(Me)3 or Br A = CH or N

General scheme 4

X = B(0H)2 or Sn(Me)3 or Br A = CH or N

General scheme 5

X = B(0H)2 or Sn(Me)3 or Br 3 A = CH or N

EXAMPLE 1

7-[6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridyl1-2-[4-(4-methylpiperazin-1-yl)anili nolpyrrolo[2, 3- dlpyrimidine-5-carbonitrile

Step 1 : 2-[6-(5-bromo-2-chloro-pyrrolo[2,3-d]pyrimidin-7-yl)-2-pyrid yl]propan-2-ol

To a stirred solution of 5-bromo-2-chloro-7H-pyrrolo[2,3-d]pyrimidine (0.247 g, 1 .06 mmol, commercially available: Ark-Pharma, Cat. No. AK-24276) in chloroform (10 mL) was added pyridine (0.58 g, 0.59 mL, 7.33 mmol) followed by copper (II) acetate (0.19 g, 1 .05 mmol) at room temperature. The reaction mixture was purged with oxygen gas for 15 minutes at room temperature. 2-(6-trimethylstannyl-2-pyridyl)propan-2-ol (0.64 g, 2.13 mmol, see below for preparation) was then added at room temperature. The reaction mixture was stirred for a further 16h at room temperature under oxygen. The reaction mixture was poured into water (30 mL) and extracted with DCM (3 x 30 mL). The organic layers were combined, dried over anhydrous sodium sulphate, filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel eluting with 20% ethyl acetate in hexane to afford title product as a white solid (0.15 g, 39%).

Step 2: 2-[6-[5-bromo-2-[4-(4-methylpiperazin-1 -yl)anilino]pyrrolo[2,3-d]pyrimidin-7-yl]-2- pyridyl]propan-2-ol

To a solution of 2-[6-(5-bromo-2-chloro-pyrrolo[2,3-d]pyrimidin-7-yl)-2-pyrid yl]propan-2-ol (0.1 g, 0.272 mmol) and 4-(4-methylpiperazin-1-yl)aniline (0.052 g, 0.27 mmol, commercially available: from Combi-blocks, order code AN-1426) in THF (2 mL) was added a solution of Li-HMDS (1 M in THF) (0.82 mL, 0.82 mmol) under a nitrogen atmosphere at 0°C. The resulting reaction mixture was stirred at room temperature for 1 h followed by heating at 90°C for 14 h. The reaction mixture was allowed to cool to room temperature, then was poured into a sat. solution of NaHCC>3 (10 mL) and stirred at room temperature for 1 h. The aqueous layer was extracted with ethyl acetate (3 x 20 mL). The organic layers were combined, dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The residue was purified by flash column chromatography on neutral alumina eluting with 3% methanol in DCM to afford the title product as an off-white solid (0.03 g, 21%).

Step 3: 7-[6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridyl]-2-[4-(4-methylpiperazin-1 - yl)anilino]pyrrolo[2,3-d]pyrimidine-5-carbonitrile To a solution of 2-[6-[5-bromo-2-[4-(4-methylpiperazin-1 -yl)anilino]pyrrolo[2,3-d]pyrimidin- 7-yl]-2-pyridyl]propan-2-ol (0.030 g, 0.057 mmol) in DMF (0.8 ml.) was added Zn(CN) 2 (0.067 g, 0.57 mmol) at room temperature. The resulting reaction mixture was degassed with nitrogen for 15 min. Pd 2 (dba) 3 (0.002 g, 0.0022 mmol) and 1 ,1 '- Bis(diphenylphosphino)ferrocene (0.003 g, 0.0054 mmol) were added to the reaction mixture which was heated in a microwave reactor at 180°C for 30 min. The reaction mixture was allowed to cool to room temperature and was poured into water (15 ml_). The aqueous phase was extracted with ethyl acetate (3 x 10 ml_). The organic phases were combined, dried over anhydrous Na 2 SC>4, filtered and concentrated in vacuo. The residue material was purified by flash column chromatography on silica eluting with 12% ethyl acetate in hexane to afford the title product as a brown solid (0.015 g, 56%).

2-(6-trimethylstannyl-2-pyridyl)propan-2-ol was prepared as follows:

To a stirred solution of 2-(6-bromopyridin-2-yl)propan-2-ol (0.1 g, 0.47 mmol, commercially available CAS# 638218-78-7) in 1 ,4 dioxane (3 ml.) was added Hexamethylinditin (0.145 g, 0.55 mmol) at room temperature. The resulting reaction mixture was degassed by bubbling nitrogen through for 20 min. Pd(PPh 3 )2CI 2 (0.032 g, 0.46 mmol) was added and the reaction mixture was heated at 100°C for 1 h. The reaction mixture was allowed to cool to room temperature and was directly concentrated in vacuo. The residue was purified by flash column chromatography on neutral alumina eluting with 20% ethyl acetate in hexane to afford the title product as a colorless liquid (0.1 g, 72%). The reaction can be scaled up accordingly.

EXAMPLE 2

7-[6-( 1 -hvdroxy-1 -methyl-ethyl)-2-pyridyl1-2-[4-[[[(3S)-tetrahvdrofuran-3- yl1amino1methyl1anilinolPyrrolo[2.3-dlPyrimidine-5-carbonitr ile dihvdrochloride

The title product was prepared from 2-[6-(5-bromo-2-chloro-pyrrolo[2,3-d]pyrimidin-7-yl)- 2-pyridyl]propan-2-ol (0.35 g, 0.952 mmol) (refer to Example 1 for preparation) using the same method as described for Example 1 with the following exceptions: a) in step 2 tert- butyl N-[(4-aminophenyl)methyl]-N-[(3S)-tetrahydrofuran-3-yl]carba mate (see below for preparation) was used instead of 4-(4-methylpiperazin-1 -yl)aniline and the product of the step was purified by flash column chromatography on silica gel eluting with 35 % ethyl acetate in hexane b) following step 3, a final deprotection of the BOC group was carried out as follows (step 4): To a stirred solution of tert-butyl N-[[4-[[5-cyano-7-[6-(1 -hydroxy-1 -methyl-ethyl)-2- pyridyl]pyrrolo[2,3-d]pyrimidin-2-yl]amino]phenyl]methyl]-N- [(3S)-tetrahydrofuran-3- yl]carbamate (0.1 g, 0.16 mmol) in DCM (0.5 ml) was added 4N HCI in 1 ,4-dioxane (0.5 ml) dropwise at 0°C. The reaction mixture was then stirred at room temperature for 4 h. The resulting precipitate was filtered off, triturated with diethyl ether (2 x 10 ml), then filtered and dried in vacuo to afford the title product as a white solid (0.045 g, 52%). tert-butyl N-[(4-aminophenyl)methyl]-N-[(3S)-tetrahydrofuran-3-yl]carba mate was prepared as follows:

Step 1 : (3S)-N-[(4-nitrophenyl)methyl]tetrahydrofuran-3-amine

To a stirred solution of 4-nitrobenzaldehyde (1 .5 g, 9.92 mmol, commercially available from Spectrochem Cat. No.011414) in methanol (30 ml.) was added (S)-3- aminotetrahydrofuran (0.91 g, 10.45 mmol) and acetic acid (0.5 ml.) under nitrogen atmosphere. The reaction mixture was stirred at room temperature for 60 min. NaCNBH 3 (1 .24 g, 19.73 mmol) was added and the resulting reaction mixture was stirred at room temperature for 3 h. The resulting reaction mixture was concentrated in vacuo and was poured into water (150 ml_). The aqueous layer was extracted with ethyl acetate (3 x 100 ml_). The combined organic layers were dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The residue was purified by column chromatography on silica gel eluting with 100 % ethyl acetate to afford the title product as a colorless solid (0.88 g, 40%).

Step 2: tert-butyl N-[(4-nitrophenyl)methyl]-N-[(3S)-tetrahydrofuran-3-yl]carba mate

To a solution of (3S)-N-[(4-nitrophenyl)methyl]tetrahydrofuran-3-amine (0.87 g, 3.92 mmol) in THF (10 ml.) was added DIPEA (0.54 g, 0.73 ml_, 4.18 mmol) and cat. DMAP (0.01 g, 0.082 mmol) at room temperature under a nitrogen atmosphere. Di-ferf-butyl dicarbonate (2.15 g, 9.85 mmol) was added to the reaction mixture at room temperature. The reaction mixture was concentrated in vacuo and the residue was purified by flash column chromatography on silica gel eluting with 25% ethyl acetate in hexanes to afford title product as a yellow viscous liquid (1 .1 g, 87%).

Step 3: tert-butyl N-[(4-aminophenyl)methyl]-N-[(3S)-tetrahydrofuran-3-yl]carba mate

To a solution of tert-butyl N-[(4-nitrophenyl)methyl]-N-[(3S)-tetrahydrofuran-3- yl]carbamate (1 .1 g, 3.41 mmol) in MeOH (15 ml.) was added 10% Pd/C (0.2 g) under a nitrogen atmosphere at room temperature. The reaction mixture was then stirred under a hydrogen atmosphere at room temperature for 2 h. The reaction mixture was filtered through celite and the resulting filtrate concentrated in vacuo to afford title product as a brown viscous liquid (0.95 g, Yield: 95%).

EXAMPLE 3

7-[6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridyl1-2-[4-[[[(3R)-tetrahvdrofuran-3- yl1amino1methyl1anilinolpyrrolo[2,3-dlpyrimidine-5-carbonitr ile

The title product was prepared from 2-[6-(5-bromo-2-chloro-pyrrolo[2,3-d]pyrimidin-7-yl)- 2-pyridyl]propan-2-ol ((0.35 g, 0.952 mmol) as described in in Example 2 with the following exceptions: a) in step 2 tert-butyl N-[(4-aminophenyl)methyl]-N-[(3R)- tetrahydrofuran-3-yl]carbamate (see below for preparation) was used instead of tert-butyl N-[(4-aminophenyl)methyl]-N-[(3S)-tetrahydrofuran-3-yl]carba mate b) in step 3 the product after work-up was purified by flash column chromatography on neutral alumina eluting with 45 % ethyl acetate in hexane c) step 4 conditions were as follows: tert-butyl N-[[4-[[5-cyano-7-[6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridyl]pyrrolo[2,3-d]pyrimidin- 2-yl]amino]phenyl]methyl]-N-[(3R)-tetrahydrofuran-3-yl]carba mate (0.055 g, 0.097 mmol) was added to formic acid (3 mL) under a nitrogen atmosphere at 0°C. The reaction mixture was stirred at room temperature for 2 h. The reaction mixture was then poured into aqueous saturated NaHCC>3 solution (40 mL). The aqueous layer was extracted using ethyl acetate (3 x 30 mL). The organic layer was collected, dried over Na 2 SC> 4 , filtered and the filtrate concentrated in vacuo. The residue was purified by flash column chromatography on silica gel, eluting with 8% methanol in chloroform. The obtained solid was triturated with n-pentane (2 x 5 mL) to afford title product as a white solid (0.012 g, Yield: 27%). tert-butyl N-[(4-aminophenyl)methyl]-N-[(3R)-tetrahydrofuran-3-yl]carba mate was prepared as follows:

Step 1 : (3R)-N-[(4-nitrophenyl)methyl]tetrahydrofuran-3-amine

To a stirred solution of 4-nitrobenzaldehyde (1 .5 g, 9.92 mmol) in methanol (25 mL) was added (3R)-tetrahydrofuran-3-amine (1.03 g, 11 .82 mmol) and acetic acid (0.5 mL) under an atmosphere of nitrogen. The reaction mixture was stirred at room temperature for 30 min. NaCNBH 3 (1 .24 g, 19.73 mmol) was added and the reaction mixture was stirred at room temperature for 3 h. The reaction mixture was concentrated in vacuo and diluted with water (150 ml_). The aqueous layer was extracted with ethyl acetate (3 x 100 ml_). The organic phase was separated off, dried over anhydrous Na 2 SC> 4 , filtered and then concentrated in vacuo. The residue was purified by flash column chromatography on silica gel eluting with 30 % ethyl acetate in hexanes to afford the title product as a colourless solid (1.6 g, 73%).

Step 2: tert-butyl N-[(4-nitrophenyl)methyl]-N-tetrahydrofuran-3-yl-carbamate

To a solution of (3R)-N-[(4-nitrophenyl)methyl]tetrahydrofuran-3-amine (1.55 g, 6.97 mmol) in THF (25 ml.) was added DIPEA (0.95 g, 1 .28 ml_, 7.35 mmol) and cat. DMAP (0.010 g, 0.082 mmol) at room temperature under a nitrogen atmosphere. Di-tert-butyl dicarbonate (3.8 g, 17.41 mmol) was added to the reaction mixture at room temperature. The reaction mixture was stirred at room temperature for 12 h and was then poured into water (100 ml_). The aqueous layer was extracted with ethyl acetate (3 x 70 ml_). The combined organic layers were dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel eluting with 11% ethyl acetate in hexanes to afford the title product as a red solid (0.85 g, 38%).

Step 3: tert-butyl N-[(4-aminophenyl)methyl]-N-tetrahydrofuran-3-yl-carbamate

To a solution of tert-butyl N-[(4-nitrophenyl)methyl]-N-tetrahydrofuran-3-yl-carbamate (0.8 g, 2.48 mmol) in MeOH (20 ml.) was added Pd-C (10%) (0.08 g) under an atmosphere of nitrogen at room temperature. The reaction mixture was stirred under hydrogen at room temperature for 3 h. the reaction mixture was filtered and the filtrate concentrated in vacuo. The residue was purified by flash column chromatography on silica gel eluting with 25% ethyl acetate in hexane to afford the title product as a red semisolid (0.48 g, 66%).

EXAMPLE 4

2-[4-[(1 -ethyl-4-piperidyl)amino1anilino1-7-[6-(1 -hvdroxy-1 -methyl-ethyl)-2- pyridyllPyrrolo[2.3-dlPyrimidine-5-carbonitrile

The title product was prepared by following the same method as for Example 1 with the following exceptions: a) in step 2, N1 -(1-ethyl-4-piperidyl)benzene-1 ,4-diamine was used instead of 4-(4-methylpiperazin-1-yl)aniline (see below for preparation of N1 -(1 -ethyl-4- piperidyl)benzene-1 ,4-diamine). In addition, the product of step 2 was purified by flash column chromatography on silica gel eluting with 4.5% methanol in DCM b) in step 3 the product was purified by flash column chromatography on silica gel eluting with 5% methanol in DCM. Yield of step 3: 0.065 g.

N1 -(1 -ethyl-4-piperidvDbenzene-1 ,4-diamine was prepared as follows:

Step 1

1 -ethyl-N-(4-nitrophenvDpiperidin-4-amine

To a stirred solution of 1 -fluoro-4-nitrobenzene (1 .0 g, 7.09 mmol) in DMF (12 ml.) was added K 2 CO 3 (1 .95 g, 14.11 mmol). To that 1 -ethyl-piperidin-4-amine dihydrochloride (1 .42 g, 7.06 mmol) was added at room temperature. The resulting reaction mixture was heated at 100°C for 12 h. The reaction mixture was cooled to room temperature and poured onto ice water (50 ml). The resulting yellow precipitate was filtered and dried in vacuo to afford title product as a yellow solid (1 .28 g, 73%). This material was directly used for next step without any further purification.

Step 2

N1 -(1 -ethyl-4-piperidyl)benzene-1 .4-diamine

To a stirred solution of 1 -ethyl-N-(4-nitrophenyl)piperidin-4-amine (0.25 g, 1.00 mmol) in methanol (5 ml.) was added Pd/C (15%) (0.05 g, 20% w/w) at room temperature. The resulting reaction mixture was stirred under an atmosphere of hydrogen for 3 h. The reaction mixture was filtered through celite, washing through with methanol (15 ml_). The filtrate was dried over Na 2 SC> 4 and concentrated in vacuo to afford the title product which was taken forward in the crude state.

EXAMPLE 5

7-[6-(1 -hydroxy- 1 -methyl-ethyl)-2-pyridyl1-2-(4-piperazin-1 -ylanilino)pyrrolo[2.3- dlPyrimidine-5-carbonitrile

The title product was prepared by following the same method as for Example 1 with the following exceptions: a) in step 2, tert-butyl 4-(4-aminophenyl)piperazine-1 -carboxylate was used instead of 4-(4-methylpiperazin-1 -yl)aniline b) the product of step 3 was subject to a further BOC deprotection step (step 4) using conditions described below.

BOC deprotection conditions: To a stirred solution of tert-butyl 4-[4-[[5-cyano-7-[6-(1 -hydroxy-1 -methyl-ethyl)-2- pyridyl]pyrrolo[2,3-d]pyrimidin-2-yl]amino]phenyl]piperazine -1-carboxylate (0.04 g, 0.072 mmol) in DCM (5 ml.) was added 4N HCI in 1 ,4-dioxane (0.5 ml.) at 0°C. The reaction mixture was stirred at room temperature for 2 h. The reaction mixture was concentrated in vacuo to afford the residue which was further purified by flash column chromatography on silica gel eluting with 3% methanol in DCM. Yield: 0.010 g, (31%).

EXAMPLE 6

2-[6-[2-(3-chloro-4-piperazin-1-yl-anilino)pyrrolo[2 , 3-dlpyrimidin-7-yl1-2-pyridyllpropan-2-ol dihvdrochloride

Step 1 : 2-[6-(2-chloropyrrolo[2,3-d]pyrimidin-7-yl)-2-pyridyl]propan -2-ol

To a stirred mixture of 2-chloro-7H-pyrrolo[2,3-d]pyrimidine (1 .0 g, 6.51 mmol, commercially available CAS# 335654-06-3) and 2-(6-bromo-2-pyridyl)propan-2-ol (1.67 g, 7.73 mmol, commercially available CAS# 638218-78-7) in 1 ,4-dioxane (12 mL) was added K 3 PC> 4 (4.83 g, 22.75 mmol) under a nitrogen atmosphere at room temperature.

The resulting reaction mixture was purged by bubbling nitrogen through for 30 min. To the reaction mixture was added copper iodide (0.371 g, 1 .95 mmol) and trans-N,N’- dimethylcyclohexane-1 ,2-diamine (0.277 g, 0.307 mL, 1.95 mmol) at room temperature. The reaction mixture was then heated at 100°C for 12 h. The reaction mixture was allowed to cool to room temperature and was poured into water (300 mL). The aqueous layer was extracted with ethyl acetate (3 x 200 mL). The organic layers were combined, dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel eluting with 36% ethyl acetate in hexane to afford the title product as a yellow solid (1 .39 g, 74%).

Step 2: tert-butyl 4-[4-[[7-[6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridyl]pyrrolo[2, 3-d]pyrimidin-2- yl]amino]phenyl]piperazine-1-carboxylate

To a stirred mixture of 2-[6-(2-chloropyrrolo[2,3-d]pyrimidin-7-yl)-2-pyridyl]propan -2-ol (0.5 g, 1 .73 mmol) and tert-butyl 4-(4-aminophenyl)piperazine-1-carboxylate (0.53 g, 1 .91 mmol) in THF (10 mL) was added a solution of Li-HMDS (1 M in THF) (4.09 mL, 4.09 mmol) under a nitrogen atmosphere at room temperature. The resulting reaction mixture was stirred for 30 min at room temperature. The reaction mixture was then heated at 100°C for 12 h. The reaction mixture was allowed to cool to room temperature, poured into a saturated aqueous solution of NaHCC>3 (10 ml.) and stirred for 1 h. the resulting reaction mixture was poured into water (300 ml_). The aqueous layer was extracted with ethyl acetate (3 x 100 ml_). The organic layers were combined and dried over anhydrous Na 2 SC> 4 , filtered and then concentrated in vacuo. The obtained residue was purified by flash column chromatography on silica gel eluting with 35 % ethyl acetate in hexane to afford the title product as a yellow solid (0.9 g, 98%).

Step 3: tert-butyl 4-[2-chloro-4-[[7-[6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridyl]pyrrolo[2, 3- d]pyrimidin-2-yl]amino]phenyl]piperazine-1-carboxylate

To a stirred solution of tert-butyl 4-[4-[[7-[6-(1 -hydroxy-1 -methyl-ethyl)-2- pyridyl]pyrrolo[2,3-d]pyrimidin-2-yl]amino]phenyl]piperazine -1-carboxylate (0.85 g, 1.60 mmol) in DCM (16 ml.) was added N-chlorosuccinimide (0.257 g, 1 .92 mmol) at 0°C. The resulting reaction mixture was stirred at room temperature for 12 h. The reaction mixture was cooled to room temperature and poured into water (200 ml_). The aqueous phase was extracted with ethyl acetate (4 x 150 ml_). The organic phases were combined and dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel, eluting with 37% ethyl acetate in hexane and then further purified by trituration with n-pentane (2 x 10 ml.) and then diethyl ether (2 x 8 ml_). Title product was isolated as a dark solid (0.28 g, 31%).

Step 4: 2-[6-[2-(3-chloro-4-piperazin-1 -yl-anilino)pyrrolo[2,3-d]pyrimidin-7-yl]-2- pyridyl]propan-2-ol hydrochloride

To a solution of tert-butyl 4-[2-chloro-4-[[7-[6-(1 -hydroxy-1 -methyl-ethyl)-2- pyridyl]pyrrolo[2,3-d]pyrimidin-2-yl]amino]phenyl]piperazine -1-carboxylate (0.230 g, 0.401 mmol) in DCM (5 ml.) was added 4N HCI in 1 ,4-dioxane (2.3 ml.) under a nitrogen atmosphere at room temperature. The reaction mixture was stirred at room temperature for 1.5 h. The reaction mixture was concentrated in vacuo. The residue was purified by preparative HPLC method A to afford the title product as a light yellow solid (0.09 g,

41%).

2-(6-bromo-2-pyridyl)propan-2-ol was prepared as follows:

To a stirred solution of 1.6M n-butyl lithium in hexanes (53 ml_, 0.085 mol) in a 500 mL round bottom flask was added anhydrous THF (52 mL) at -78°C under nitrogen. The solution was stirred for 5 min. To this, a solution of 2,6-dibromo pyridine (20 g, 0.0844 mol) in THF (104 ml.) was added dropwise over a period of 30 minutes at -78°C. The reaction mixture was stirred for 30 minutes at the same temperature. After this, acetone (8.33 g, 10.63 ml_, 0.143 mol) was added dropwise to the reaction mixture over a period of 10 minutes and the resulting reaction was further stirred for 1 h at -78°C. After completion of the reaction, the reaction mixture was warmed to room temperature, poured into saturated aqueous NH CI solution and the aqueous was extracted with ethyl acetate (200 ml. x 3). The organic layers were combined, washed with brine, dried over anhydrous sodium sulphate, filtered and then concentrated in vacuo. The residue was purified by flash column chromatography using neutral silica gel eluting with 6% ethyl acetate in hexane to afford title product as a colourless liquid (15 g, 83%).

EXAMPLE 7

2-[6-[2-(4-piperazin-1 -ylanilino)pyrrolo[2 , 3-dlpyrimidin-7-yl1-2-pyridyllpropan-2-ol dihvdrochloride

The title product was prepared by following the same method as for Example 6 with the following exceptions: a) Step 3 was omitted b) The product of the BOC deprotection step (see example 26, step 4) was purified as follows: the reaction mixture was concentrated in vacuo then triturated with diethyl ether to afford the title product as a yellow solid (0.075 g, 79%).

EXAMPLE 8

2-f6-[5-fluoro-2-(4-piperazin-1 -ylanilino)pyrrolo[2 , 3-dlpyrimidin-7-yll-2-pyridyl]propan-2-ol dihydrochloride

Step 1 : 2-[6-(2-chloro-5-fluoro-pyrrolo[2,3-d]pyrimidin-7-yl)-2-pyri dyl]propan-2-ol

To a stirred solution of 2-chloro-5-fluoro-7H-pyrrolo[2,3-d]pyrimidine (0.2 g, 1 .2 mmol, commercially available from Astatech, CAS# 1638768-28-1 ) in 1 ,4-dioxane (8 mL) was added 2-(6-bromo-2-pyridyl)propan-2-ol (0.3 g, 1.4 mmol, refer to Example 6 for preparation) and K 3 PC> 4 (0.36 g, 1 .7 mmol). The reaction mixture was purged by bubbling nitrogen gas through for 15 minutes. Trans-N,N'-Dimethylcyclohexane-1 ,2-diamine (0.18 g, 0.20 mL, 1 .3 mmol) and Cul (0.22 g, 1 .2 mmol) were added to the reaction mixture at room temperature. The reaction mixture was heated at 100°C for 16h. The reaction mixture was allowed to cool to room temperature and poured into water (50 mL). The aqueous layer was extracted with ethyl acetate (2 x 50 mL). The combined organic phase was dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The obtained residue was purified by flash column chromatography on silica gel eluting with 7% ethyl acetate in hexane to afford title product as a yellow solid (0.35 g, 98%).

Step 2: tert-butyl 4-[4-[[5-fluoro-7-[6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridyl]pyrrolo[2, 3- d]pyrimidin-2-yl]amino]phenyl]piperazine-1-carboxylate

To a solution of 2-[6-(2-chloro-5-fluoro-pyrrolo[2,3-d]pyrimidin-7-yl)-2-pyri dyl]propan-2-ol (0.32 g, 1.04 mmol) and tert-butyl 4-(4-aminophenyl)piperazine-1-carboxylate (0.29 g,

1 .05 mmol) in THF (12 ml.) was added a 1 M solution of Li-HMDS in THF (3.2 ml.) under nitrogen. The resulting reaction mixture was stirred for 30 min at 0°C. The reaction mixture was then heated at 100°C for 2 h. The reaction mixture was allowed to cool to room temperature, poured into a saturated aqueous solution of NaFICO 3 (10 ml_). The aqueous layer was extracted with ethyl acetate (2 x 50 ml_). The organic layers were then combined and dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The obtained residue was purified by flash column chromatography on silica gel eluting with 31% ethyl acetate in hexane to afford the title product as an off-white solid (0.23 g, 40%).

Step 3: 2-[6-[5-fluoro-2-(4-piperazin-1 -ylanilino)pyrrolo[2,3-d]pyrimidin-7-yl]-2- pyridyl]propan-2-ol hydrochloride

To a solution of tert-butyl 4-[4-[[5-fluoro-7-[6-(1 -hydroxy-1 -methyl-ethyl)-2- pyridyl]pyrrolo[2,3-d]pyrimidin-2-yl]amino]phenyl]piperazine -1-carboxylate (0.18 g, 0.33 mmol) in DCM (10 ml.) was added 4N HCI in 1 ,4-dioxane (1 ml.) at 0°C. The reaction mixture was stirred at room temperature for 2 h. The reaction mixture was concentrated in vacuo. The obtained residue was triturated by diethyl ether (1 x 10 ml.) to afford the title product as a yellow solid (0.12 g, 70%).

EXAMPLE 9

7-[6-(1-hvdroxy-1-methyl-ethyl)-2-pyridyl1-2-[4-(4-piperi dylamino)anilinolPyrrolo[2.3- dlPyrimidine-5-carbonitrile dihvdrochloride

The title product was prepared by following the same method as for Example 5 with the following exceptions: a) tert-butyl 4-(4-aminoanilino)piperidine-1-carboxylate was used instead of tert-butyl 4-(4-aminophenyl)piperazine-1-carboxylate b) in step 4 the product was purified as follows: the reaction mixture was concentrated in vacuo and was triturated with n-pentane (3 x 5 mL) and diethyl ether (3 x 5 ml.) and then dried via lyophilization to afford title product: 0.055 g (96%).

EXAMPLE 10

1 -[4-[4-[[7-[6-( 1 -hvdroxy-1 -methyl-ethyl)-2-pyridyllpyrrolo[2,3-dlpyrimidin-2- yl1aminolphenyllpiperazin-1 -yl1ethanone

The title product was prepared by following the same method as for Example 7 with the following exceptions: a) in step 2, 1 -[4-(4-aminophenyl)piperazin-1 -yl]ethanone (commercially available) was used instead of tert-butyl 4-(4-aminophenyl)piperazine-1 - carboxylate b) the deprotection step (step 3) was omitted. Yield of step 2: 0.010 g (2%).

EXAMPLE 11

4-[[7-[6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridyllpyrrolo[2,3-dlpyrimidin-2-yl1amino1 -N-(1 - methyl-4-piperidyl)benzamide

The title product was prepared by following the same method as for Example 10 except that in step 2, 4-amino-N-(1 -methyl-4-piperidyl)benzamide was used instead of tert-butyl 4-(4-aminophenyl)piperazine-1 -carboxylate. Yield of step 2: brown solid, 0.1 g (30%).

4-amino-N-(1 -methyl-4-piperidyl)benzamide was prepared as follows:

Step 1 : N-(1 -methyl-4-piperidyl)-4-nitro-benzamide

To a stirred solution of 4-nitrobenzoic acid (3 g, 18.0 mmol) in DMA (30 mL) was added 1 - methylpiperidin-4-amine (2.05 g, 18.0 mmol) at room temperature. DIPEA (6.90 g, 9.30 mL, 53.4 mmol) and HATU (10.24 g, 26.9 mmol) were added to the reaction mixture. The reaction mixture was then stirred at room temperature for 4 h. The reaction mixture was poured into water (50 mL). The aqueous layer was extracted with ethyl acetate (2 x 80 mL). The combined organic phases were dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The obtained residue was purified by column chromatography on silica gel eluting with 8% MeOH in DCM to afford the title product as a yellow solid (6.0 g, >100%).

Step 2: 4-amino-N-(1 -methyl-4-piperidyl)benzamide To a stirred solution of N-(1 -methyl-4-piperidyl)-4-nitro-benzamide (1 .0 g, 3.8 mmol) in methanol (10 ml.) was added 10% Pd/C (0.2 g, 20 mol%) at room temperature. The reaction mixture was purged by bubbling hydrogen gas through at room temperature for 1 h. The reaction mixture was filtered through celite, washing through with methanol (30 ml.) at room temperature. The combined filtrate was concentrated in vacuo to afford the title product as an off-white solid (0.8 g, 90%).

EXAMPLE 12 tert-butyl 4-[4-[[5-fluoro-7-[6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridyllpyrrolo[2,3-dlpyrimidin-

2-yl1aminolphenyllpiperazine-1-carboxylate

Refer to Example 8, step 2 for synthesis of the title product.

EXAMPLE 13

2-[6-[2-[4-(4,7-diazaspiro[2.51octan-7-yl)anilino1-5-fluo ro-pyrrolo[2,3-dlpyrimidin-7-yl1-2- pyridyllpropan-2-ol dihvdrochloride

The title product was prepared by following the same method as for Example 8 with the following exceptions: a) in step 2, tert-butyl 7-(4-aminophenyl)-4,7-diazaspiro[2.5]octane- 4-carboxylate was used instead of tert-butyl 4-(4-aminophenyl)piperazine-1-carboxylate b) in step 3 the triturated product was further purified by preparative HPLC method C to afford the title product as a yellow solid (0.080 g, 37%). tert-butyl 7-(4-aminophenyl)-4,7-diazaspiro[2.5]octane-4-carboxylate was prepared as follows:

Step 1 : tert-butyl 7-(4-nitrophenyl)-4,7-diazaspiro[2.5]octane-4-carboxylate

To a stirred solution of 1 -fluoro-4-nitrobenzene (0.73 g, 5.2 mmol) and tert-butyl 4,7- diazaspiro[2.5]octane-4-carboxylate (1.0 g, 4.7 mmol, commercially available) in DMF (12 mL) was added K 2 CO 3 (1 .94 g, 14.0 mmol,) at room temperature. The reaction mixture was heated at 100°C for 5 h. The reaction mixture was allowed to cool to room temperature and was poured into ice-water (50 mL) then stirred for 15 min. The precipitated solid was filtered off then dried in vacuo to afford the title product as a yellow solid (1.74 g, 92.40%).

Step 2: tert-butyl 7-(4-aminophenyl)-4,7-diazaspiro[2.5]octane-4-carboxylate To a stirred solution of tert-butyl 7-(4-nitrophenyl)-4,7-diazaspiro[2.5]octane-4-carboxylate (1 .74 g, 5.2 mmol) in methanol (60 ml.) was added 10% Pd/C (0.3 g) at room temperature. The reaction mixture was purged by bubbling hydrogen gas through at room temperature for 1 h. The reaction mixture was filtered through celite, washing through with methanol (2 x 20 ml_). The combined filtrate was concentrated in vacuo to afford title product as a brown liquid (1 .2 g, 76%).

EXAMPLE 14

2-[6-[5-fluoro-2-[4-(tetrahvdropyran-4-ylmethylamino)anil inolpyrrolo[2 , 3-dlpyrimidin-7-yl1-

2-pyridyllpropan-2-ol hydrochloride

The title product was prepared by following the same method as for Example 13 with the following exceptions: a) in step 2, tert-butyl N-(4-aminophenyl)-N-(tetrahydropyran-4- ylmethyl)carbamate (see below for preparation) was used instead of tert-butyl 4,7- diazaspiro[2.5]octane-4-carboxylate b) in step 3 the triturated product was further purified by preparative HPLC method D to afford the title product as a yellow solid (0.090 g, 34%). tert-butyl N-(4-aminophenyl)-N-(tetrahydropyran-4-ylmethyl)carbamate was prepared as follows:

Step 1 : 4-nitro-N-(tetrahydropyran-4-ylmethyl)aniline

To a stirred mixture of 1-fluoro-4-nitro-benzene (0.5 g, 3.54 mmol) and tetrahydropyran-4- ylmethanamine (0.4 g, 3.47 mmol) in DMF (10 mL) was added K 2 CO 3 (0.97 g, 7.02 mmol) at room temperature. The reaction mixture was heated at 100°C for 16h with stirring. The reaction mixture was allowed to cool to room temperature and was then poured into ice- cold water (50 mL). The precipitate was filtered, washed with water (3 x 25 mL) and dried under vacuum to afford the title product as a yellow solid (0.7 g, 84%).

Step 2: tert-butyl N-(4-nitrophenyl)-N-(tetrahydropyran-4-ylmethyl)carbamate

To a stirred solution of 4-nitro-N-(tetrahydropyran-4-ylmethyl)aniline (0.7 g, 2.96 mmol) in THF (14 mL) was added a 1.0M solution of Li-HMDS in THF (5.9 mL, 5.9 mmol). Di-tert- butyl dicarbonate (0.97 g, 4.44 mmol) was added at room temperature. The reaction mixture was stirred at 70°C for 16 h. The reaction mixture was allowed to cool to room temperature and was diluted with ethyl acetate (50 mL) and was washed with saturated aqueous NaFICC>3 solution (2 x 50 mL).The organic phase was then washed with brine solution (50 ml_), dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel eluting with 20-25% ethyl acetate in hexane to afford title product as a yellow solid (1 .0 g, 100%).

Step 3: tert-butyl N-(4-aminophenyl)-N-(tetrahydropyran-4-ylmethyl)carbamate

To a solution of tert-butyl N-(4-nitrophenyl)-N-(tetrahydropyran-4-ylmethyl)carbamate (1 .0 g, 2.97 mmol) in methanol (100 ml.) was added 10% Pd/C (0.2 g, 50% wet with water) at room temperature. The reaction mixture was stirred under a hydrogen atmosphere at room temperature for 2 h. The reaction mixture was filtered through celite, washing through with methanol (2 x 20 ml_). The combined filtrate was concentrated in vacuo to afford the title product as yellow solid (0.6 g, 66%).

EXAMPLE 15

N-[4-[[5-fluoro-7-[6-(1-hvdroxy-1-methyl-ethyl)-2-pyridyl lpyrrolo[2,3-dlpyrimidin-2- yllaminolphenylltetrahvdropyran-4-carboxamide hydrochloride

The title product was prepared by following the same method as for Example 14 with the following exceptions: a) in step 2, N-(4-aminophenyl)tetrahydropyran-4-carboxamide was used instead of tert-butyl N-(4-aminophenyl)-N-(tetrahydropyran-4-ylmethyl)carbamate b) in step 2 the product after work-up was purified by flash column chromatography on silica gel eluting with 50% ethyl acetate in hexane. The product was further purified by preparative HPLC method E to afford the title product as a yellow solid (0.080 g, 9%)

Step 3 was omitted.

N-(4-aminophenyl)tetrahydropyran-4-carboxamide was prepared as follows:

Step 1 : N-(4-nitrophenyl)tetrahydropyran-4-carboxamide

To a stirred solution of tetrahydropyran-4-carboxylic acid (2.5 g, 19.2 mmol) and 4- nitroaniline (2.64 g, 19.1 mmol) in DCM (75 mL) was added pyridine (15.11 g, 15.39 mL, 191 .0 mmol) at 0°C. POCI 3 (20.49 g, 12.49 mL, 134 mmol) was then added at 0°C. The reaction mixture was stirred at room temperature for 2 h. The reaction mixture was poured into ice cold water (200 mL) and the aqueous layer was extracted with DCM (2 x 100 mL). The organic layers were combined and dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The obtained residue was purified by flash column chromatography on silica gel using 40% ethyl acetate in hexane to afford the title product as a yellow solid (3.2 g, 67%).

Step 2: N-(4-aminophenyl)tetrahydropyran-4-carboxamide

To a stirred solution of N-(4-nitrophenyl)tetrahydropyran-4-carboxamide (3.0 g, 12.0 mmol) in methanol (30 ml.) was added 10% Pd/C (0.6 g, 20% w/w) at room temperature. The reaction mixture was purged by bubbling hydrogen gas through at room temperature for 4 h. The reaction mixture was filtered through celite, washing through with methanol (2 x 20 ml.) at room temperature. The combined filtrate was concentrated in vacuo to afford the title product (2.3 g, 87%).

EXAMPLE 16

2-[6-[5-fluoro-2-[4-(tetrahvdropyran-4-ylamino)anilinolpy rrolo[2 , 3-dlpyrimidin-7-yl1-2-

Pyridyllpropan-2-ol hydrochloride

The title product was prepared by following the same method as for Example 15 with the following exceptions: a) in step 2, N1 -tetrahydropyran-4-ylbenzene-1 ,4-diamine was used instead of N-(4-aminophenyl)tetrahydropyran-4-carboxamide and preparative HPLC method F was used instead of method E. Yield: brown solid (0.100 g, 12%).

EXAMPLE 17

7-f6-[[dimethyl(oxo)-lambda6-sulfanylidenelaminol-2-pyrid vn-5-fluoro-N-f4-(4- methylpiperazin-1 -yl)phenynpyrrolo[2,3-d]pyrimidin-2-amine dihydrochloride

Step 1 : [6-(2-chloro-5-fluoro-pyrrolo[2,3-d]pyrimidin-7-yl)-2-pyridy l]imino-dimethyl-oxo- Iambda6-sulfane

To stirred solution of 2-chloro-5-fluoro-7H-pyrrolo[2,3-d]pyrimidine (0.5 g, 2.91 mmol) in 1 ,4-dioxane (10 mL) was added (6-bromo-2-pyridyl)imino-dimethyl-oxo-lambda6-sulfane (1 .3 g, 5.22 mmol) and K 3 PO 4 (1 .76 g, 8.29 mmol). The reaction mixture was purged by bubbling nitrogen gas through for 15 min. Trans-N,N'-Dimethylcyclohexane-1 ,2-diamine (0.083g, 0.092 mL, 0.58 mmol) and Cul (0.11 g, 0.58 mmol) were added to the reaction mixture at room temperature. The resulting reaction mixture stirred at 100°C for 2 h. The reaction mixture was cooled to room temperature and poured into ice-cold water (20 mL). The aqueous layer was extracted with ethyl acetate (2 x 25 mL). The organic layers were combined, dried over sodium sulphate, filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel eluting with 48% ethyl acetate in hexane to afford the title product (0.4 g, 41%).

(6-bromo-2-pyridyl)imino-dimethyl-oxo-lambda6-sulfane was prepared as follows:

To a stirred solution of 2,6-dibromopyridine (1 .0 g, 4.22 mmol) in toluene (10 ml.) was added S,S-dimethyl sulfoximine (0.39 g, 4.19 mmol), BINAP (0.078 g, 0.13 mmol) and sodium tert-butoxide (0.4 g, 4.16 mmol). The reaction mixture was purged by bubbling nitrogen gas through for 15 min at room temperature. Pd 2 (dba) 3 (0.11 g, 0.12 mmol) was added at room temperature. The resulting reaction mixture was heated at 120°C for 1 h in a microwave reactor. The reaction mixture was cooled to room temperature and poured into ice-cold water (100 ml_). The aqueous layer was extracted with ethyl acetate (4 x 100 ml.) and the combined organic layers were separated off, dried over sodium sulphate, filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel, eluting with 48% ethyl acetate in hexane to afford the title product (1 .4 g, >100%).

Step 2: 7-[6-[[dimethyl(oxo)-lambda6-sulfanylidene]amino]-2-pyridyl] -5-fluoro-N-[4-(4- methylpiperazin-1 -yl)phenyl]pyrrolo[2,3-d]pyrimidin-2-amine

To a stirred solution of [6-(2-chloro-5-fluoro-pyrrolo[2,3-d]pyrimidin-7-yl)-2-pyridy l]imino- dimethyl-oxo-lambda6-sulfane (0.3 g, 0.85 mmol) and 4-(4-methylpiperazin-1 -yl)aniline (0.16 g, 0.84 mmol, commercially available CAS# 16153-81 -4) in THF (3 ml.) was added a solution of 1 M Li-HMDS in THF (2.6 ml, 2.6 mmol) at 0°C under nitrogen. The resulting reaction mixture was stirred at 0°C for 1 h. The resulting reaction mixture was poured into water (20 ml.) and was extracted with ethyl acetate (4 x 20 ml_). The combined organic layers were dried over sodium sulphate, filtered and concentrated in vacuo. The obtained residue was purified by preparative HPLC method G to afford title product as a yellow solid (0.020 g, 4%).

EXAMPLE 18

2-[6-[2-(3-chloro-4-piperazin-1-yl-anilino)-5-fluoro-pyrr olof2.3-dlPyrimidin-7-yl1-2- pyridyllpropan-2-ol dihvdrochloride

The title product was prepared by following the same method as for Example 8 except that in step 2, tert-butyl 4-(4-amino-2-chloro-phenyl)piperazine-1 -carboxylate was used instead of tert-butyl 4-(4-aminophenyl)piperazine-1 -carboxylate. Yield of step 3: yellow solid, 0.020 g (21%). tert-butyl 4-(4-amino-2-chloro-phenyl)piperazine-1-carboxylate was prepared as follows:

Step 1 : tert-butyl 4-(2-chloro-4-nitro-phenyl)piperazine-1 -carboxylate

To a stirred solution of 2-chloro-1 -fluoro-4-nitrobenzene (2.5 g, 14.2 mmol) and tert-butyl piperazine-1 -carboxylate (3.44 g, 18.5 mmol) in DMF (25 ml.) was added K2CO3 (9.82 g, 71 .1 mmol) at room temperature. The reaction mixture was stirred with heating at 120°C for 4h. The reaction mixture was allowed to cool to room temperature then was poured into cold water (200 ml_). The precipitated solid was filtered off then dried in vacuo to afford title product (5.0 g, >100%).

Step 2: tert-butyl 4-(4-amino-2-chloro-phenyl)piperazine-1 -carboxylate

To a stirred solution of tert-butyl 4-(2-chloro-4-nitro-phenyl)piperazine-1 -carboxylate (5.0 g, 14.6 mmol) in methanol (50 ml.) was added NH4CI (3.92 g, 73.3 mmol), iron filings (4.09 g, 73.2 mmol) and water (5 ml) at room temperature. The reaction mixture was heated at 70°C for 12 h. The reaction mixture was filtered through celite, washing through with methanol (2 x 50 ml.) at room temperature. The combined filtrate was concentrated in vacuo and the residue purified by manual flash column chromatography using 10% ethyl acetate in hexane to afford title product as a brown solid (2.0 g, 44%).

EXAMPLE 19

2-[6-[2-[3-chloro-4-(4-ethylpiperazin-1 -yl)anilino1-5-fluoro-pyrrolo[2.3-dlPyrimidin-7-yl1-2- pyridyllpropan-2-ol dihvdrochloride

The title product was prepared by following the same method as for Example 18 with the following exceptions: a) in step 2, 3-chloro-4-(4-ethylpiperazin-1 -yl)aniline was used instead of tert-butyl 4-(4-amino-2-chloro-phenyl)piperazine-1 -carboxylate and in the same step the product after flash column purification was further purified by preparative HPLC method H c) step 3 (Boc deprotection) was omitted. Yield of step 2: yellow solid, 0.080 g (8%).

3-chloro-4-(4-ethylpiperazin-1 -yl)aniline was prepared in an analogous way to tert-butyl 4- (4-amino-2-chloro-phenyl)piperazine-1 -carboxylate (see example 42 for preparation) except that 1 -ethylpiperazine was used instead of tert-butyl piperazine-1 -carboxylate in step 1 .

EXAMPLE 20

2-[6-[2-[4-(4-ethylpiperazin-1-vDanilino1-5-fluoro-pyrrol o[2,3-dlpyrimidin-7-yl1-2-

Pyridyllpropan-2-ol dihvdrochloride

The title product was prepared by following the same method as for Example 19 with the following exceptions: a) in step 2, 4-(4-ethylpiperazin-1-yl)aniline was used instead of 3- chloro-4-(4-ethylpiperazin-1-yl)aniline. Yield of step 2: yellow solid, 0.080 g (9%).

4-(4-ethylpiperazin-1-yl)aniline was prepared in an analogous way to 3-chloro-4-(4- ethylpiperazin-1 -yl)aniline except that 1-fluoro-4-nitro-benzene was used instead of 2- chloro-1 -fluoro-4-nitrobenzene.

EXAMPLE 21

2-[6-[2-[4-[(1 R,4R)-2,5-diazabicvclo[2.2.1lheptan-2-vl]anilinol-5-fluoro-p vrrolo[2,3- dlpyrimidin-7-vn-2-pyridyllpropan-2-ol dihvdrochloride

The title product was prepared by following the same method as for Example 8 with the following exceptions: a) in step 2, tert-butyl (1 R,4R)-5-(4-aminophenyl)-2,5- diazabicyclo[2.2.1]heptane-2-carboxylate was used instead of tert-butyl 4-(4- aminophenyl)piperazine-1 -carboxylate b) in step 3, following trituration, the product was further purified by preparative HPLC method I to afford the title product as an orange solid. tert-butyl (1 R,4R)-5-(4-aminophenyl)-2,5-diazabicyclo[2.2.1]heptane-2-car boxylate was prepared as follows:

Step 1 : tert-butyl (1 R,4R)-5-(4-nitrophenyl)-2,5-diazabicyclo[2.2.1]heptane-2-car boxylate

To a stirred solution of 1 -fluoro-4-nitrobenzene (1 .00 g, 7.09 mmol) and tert-butyl (1 R,4R)-2,5-diazabicyclo[2.2.1]heptane-2-carboxylate (1.40 g, 7.06 mmol, commercially available) in ACN (10 mL) was added DIPEA (3.64 mL, 2.70 g, 20.9 mmol) at room temperature. The reaction mixture was then heated at 100°C for 16 h. The reaction mixture was allowed to cool to room temperature then was poured into ice cold water (50 mL) with stirring for 15 min. The precipitated solid was filtered and dried under vacuum to afford title product as a yellow solid (1 .5 g, 67%).

Step 2: tert-butyl (1 R,4R)-5-(4-aminophenyl)-2,5-diazabicyclo[2.2.1]heptane-2- carboxylate

To a stirred solution of tert-butyl (1 R,4R)-5-(4-nitrophenyl)-2,5- diazabicyclo[2.2.1]heptane-2-carboxylate (1 .5 g, 4.7 mmol) in methanol (20 mL) was added 10% Pd/C (0.3 g, 20% w/w) at room temperature. The reaction mixture was purged by bubbling hydrogen gas through at room temperature for 4 h. The reaction mixture was filtered through celite, washing through with methanol (2 x 20 mL) at room temperature. The combined filtrate was concentrated in vacuo and triturated with pentane (40 mL) to afford title product (1 .3 g, 96%).

EXAMPLE 22

2-[6-[2-[4-[(1S,4S)-2,5-diazabicvclo[2.2.11heptan-2-yl1an ilino1-5-fluoro-pyrrolo[2,3- dlPyrimidin-7-yl1-2-pyridyllpropan-2-ol dihvdrochloride

The title product was produced by an analogous method to Example 21 except that tert- butyl (1 S,4S)-5-(4-aminophenyl)-2,5-diazabicyclo[2.2.1]heptane-2-car boxylate was used instead of tert-butyl (1 R,4R)-5-(4-aminophenyl)-2,5-diazabicyclo[2.2.1]heptane-2- carboxylate in step 2. Yield of step 3: orange solid, 0.090 g (32%).

EXAMPLE 23

2-[6-[5-fluoro-2-[4-(4-piperidyloxy)anilinolPyrrolo[2.3-d lPyrimidin-7-yl1-2-pyridyllpropan-2- ol dihvdrochloride

The title product was produced by following the same method as for Example 21 with the following exceptions: a) in step 2, tert-butyl 4-(4-aminophenoxy)piperidine-1 -carboxylate was used instead of tert-butyl (1 R,4R)-5-(4-aminophenyl)-2,5-diazabicyclo[2.2.1]heptane- 2-carboxylate b) in step 3, following trituration, the product was further purified by preparative HPLC method J to afford the title product as an yellow solid, 0.08 g (37%). tert-butyl 4-(4-aminophenoxy)piperidine-1 -carboxylate was prepared as follows:

Step 1 : tert-butyl 4-(4-nitrophenoxy)piperidine-1 -carboxylate To a stirred solution of tert-butyl 4-hydroxypiperidine-1-carboxylate (7.1 g, 35.5 mmol, commercially available) in THF (50 ml.) was added potassium tert-butoxide (5.9 g, 52.6 mmol) at room temperature. The reaction mixture was stirred at room temperature for 15 min and 1-fluoro-4-nitrobenzene (5.0 g, 35.5 mmol) was added at room temperature. The reaction mixture was stirred at room temperature for 4 h. The reaction mixture was poured into water (50 ml_). The aqueous layer was extracted with ethyl acetate (2 x 80 ml_). The combined organic phase was dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel eluting with 12% EtOAc in hexane to afford the title product as a yellow solid (9.5 g, 83%).

Step 2: tert-butyl 4-(4-aminophenoxy)piperidine-1-carboxylate

To a solution of tert-butyl 4-(4-nitrophenoxy)piperidine-1-carboxylate (3.0 g, 9.3 mmol) in methanol (50 ml.) was added 10% Pd/C (0.6 g, 20% w/w) at room temperature. The reaction mixture was purged by bubbling hydrogen through at room temperature for 3 h. The reaction mixture was filtered through celite, washing through with with methanol (30 ml.) at room temperature. The combined filtrate was concentrated in vacuo to afford the title product as a black solid (2.5 g, 92%).

EXAMPLE 24

2-[6-[2-[4-[(1-ethyl-4-piperidyl)oxy1anilino1-5-fluoro-py rrolo[2.3-dlPyrimidin-7-yl1-2- pyridyllpropan-2-ol dihvdrochloride

The title product was produced by following the same method as for Example 19 with the following exceptions: a) in step 2, 4-((1 -ethylpiperidin-4-yl)oxy)aniline was used instead of

3-chloro-4-(4-ethylpiperazin-1-yl)aniline and in the same step the product after flash column purification was further purified by preparative HPLC method K to afford the title product as an orange solid: 0.07 g (6%).

4-((1-ethylpiperidin-4-yl)oxy)aniline was prepared by an analogous method to tert-butyl 4- (4-aminophenoxy)piperidine-1-carboxylate (example 47) except that 1 -ethylpiperidin-4-ol was used instead of tert-butyl 4-hydroxypiperidine-1-carboxylate.

EXAMPLE 25 2-[6-[5-fluoro-2-[4-[[(3S)-3-piperidylloxylanilinolpyrrolo[2 ,3-dlpyrimidin-7-yll-2- pyridyllpropan-2-ol dihydrochloride

The title product was prepared by an analogous method to that used for Example 23 with the following exceptions: a) in step 2, tert-butyl (S)-3-(4-aminophenoxy)piperidine-1- carboxylate was used instead of tert-butyl 4-(4-aminophenoxy)piperidine-1-carboxylate b) in step 3 preparative HPLC method L was used to afford the title product as a yellow solid, 0.08 g (42%). tert-butyl (S)-3-(4-aminophenoxy)piperidine-1-carboxylate was prepared as follows:

Step 1 : tert-butyl (S)-3-(4-nitrophenoxy)piperidine-1-carboxylate

To a solution of tert-butyl (S)-3-hydroxypiperidine-1-carboxylate (7.8 g, 38.8 mmol) in THF (50 mL) was added NaH (2.8 g, 70 mmol, 60% dispersion in mineral oil) at 0°C under nitrogen. The resulting reaction mixture was stirred at 0°C for 10 min and 1 -fluoro-4- nitrobenzene (5.0 g, 35.5 mmol) was added. The reaction mixture was further stirred at room temperature for 16 h. The reaction mixture was diluted with methanol (30 mL) and poured into ice-cold water (150 mL). The aqueous layer was extracted with ethyl acetate (4 x 150 mL). The combined organic layers were dried over sodium sulphate, filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica eluting with 15% ethyl acetate in hexane to afford the title product (12 g, >100%).

Step 2: tert-butyl (S)-3-(4-aminophenoxy)piperidine-1-carboxylate

The title product was prepared by following the method described in step 2 of the synthesis of tert-butyl 4-(4-aminophenoxy)piperidine-1-carboxylate (refer to example 47). Yield: 1.4 g (77%).

EXAMPLE 26

2-[6-[5-fluoro-2-[4-[[(3R)-3-DiDeridylloxylanilinolDyrrol of2.3-dlDyrimidin-7-yll-2- pyridyllprcipan-2-ol dihvdrochloride

The title product was prepared by an analogous method to that used for Example 25 except that in step 2, tert-butyl (R)-3-(4-aminophenoxy)piperidine-1-carboxylate was used instead of tert-butyl (R)-3-(4-aminophenoxy)piperidine-1-carboxylate.

EXAMPLE 27 1 -[4-[[5-fluoro-7-[6-(1 -hvdroxy-1 -methyl-ethyl)-2-pyridyllpyrrolo[2,3-dlpyrimidin-2- yllaminolphenyllpiperazin-2-one dihvdrochloride

The title product was prepared by following the same method as for Example 13 with the following exceptions: a) in step 2, tert-butyl 4-(4-aminophenyl)-3-oxopiperazine-1 - carboxylate was used instead of tert-butyl 4,7-diazaspiro[2.5]octane-4-carboxylate b) in step 3 the triturated product was further purified by preparative HPLC method J to afford the title product as a brown solid (0.01 g, 11%). tert-butyl 4-(4-aminophenyl)-3-oxopiperazine-1 -carboxylate was prepared as follows:

To a stirred solution of 4-bromoaniline (1 .0 g, 5.8 mmol) in 1 ,4-dioxane (7 ml.) was added tert-butyl 3-oxopiperazine-1 -carboxylate (1 .39 g, 6.9 mmol, commercially available) and K 2 CC> 3 (2.40 g, 17.4 mmol). The reaction mixture was purged by bubbling nitrogen through for 15 minutes. trans-N,N'-dimethylcyclohexane-1 ,2-diamine (0.998 ml_, 0.900 g, 6.3 mmol) and Cul (1.10 g, 5.8 mmol) were added to the reaction mixture at room temperature. The reaction mixture was heated at 120°C for 16 h. The reaction mixture was allowed to cool to room temperature and poured into water (100 ml_). The aqueous layer was extracted with ethyl acetate (3 x 50 ml_). The combined organic phase was dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The residue was purified by flash column chromatography using neutral alumina eluting with 40% ethyl acetate in hexane to afford the title product, 1 .0 g, 59%).

EXAMPLE 28

1 -[4-[2-chloro-4-[[5-fluoro-7-[6-(1 -hvdroxy-1 -methyl-ethyl)-2-DyridyllDyrrolo[2.3- dlPyrimidin-2-yl1aminolphenyllpiperazin-1 -yl1ethanone dihvdrochloride

The title product was prepared from the compound of Example 18 as follows:

To an stirred solution of 2-[6-[2-(3-chloro-4-piperazin-1 -yl-anilino)-5-fluoro-pyrrolo[2,3- d]pyrimidin-7-yl]-2-pyridyl]propan-2-ol hydrochloride (0.30 g, 0.54 mmol) in pyridine (7 ml.) at 0°C was added acetic anhydride (2.5 ml_). The reaction mixture was stirred at 0°C for 10 min. The reaction mixture was poured into sat. aqueous NaHCC>3 solution (30 ml.) and was extracted with ethyl acetate (3 x 20 ml_). The combined organic layer was separated and washed with solution of 10% citric acid in water (20 ml_). The organic layer was then dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by preparative HPLC using HCI buffer to afford the title product as a yellow solid (0.09 g, 30%).

EXAMPLE 29

1 -[4-[2-chloro-4-[[5-fluoro-7-[6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridyllpyrrolo[2,3- dlpyrimidin-2-yl1aminolphenyllpiperazin-1 -yllpropan-1 -one dihydrochloride

The title product was prepared by an analogous method to that used for Example 28 except that propionyl chloride was used instead of acetic anhydride. Yield: yellow solid, 0.13 g (29%).

EXAMPLE 30

[4-[2-chloro-4-[[5-fluoro-7-[6-(1-hvdroxy-1-methyl-ethyl) -2-pyridyllpyrrolo[2,3-dlpyrimidin-

2-yl1aminolphenyllpiperazin-1-yl1-cvclopropyl-methanone dihydrochloride

The title product was prepared by an analogous method to that used for Example 28 except that cyclopropanecarbonyl chloride was used instead of acetic anhydride. Yield: an off-white solid, 0.15 g (33%).

EXAMPLE 31

N-(cvclopropylmethyl)-4-[[5-fluoro-7-[6-(1-hvdroxy-1-meth yl-ethyl)-2-pyridyllPyrrolo[2.3- dlPyrimidin-2-yl1amino1benzamide dihydrochloride

Step 1 : 4-[[5-fluoro-7-[6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridyl]pyrrolo[2,3-d]pyrimidin-2- yl]amino]benzoic acid

To a stirred solution of 2-[6-(2-chloro-5-fluoro-pyrrolo[2,3-d]pyrimidin-7-yl)-2- pyridyl]propan-2-ol (0.75 g, 2.5 mmol, refer to example 29, step 1) and 4-aminobenzoic acid (0.36 g, 2.6 mmol) in THF (10 mL) was added a solution of 1 M Li-HMDS in THF (7.2ml, 7.2 mmol) under nitrogen. The reaction mixture was stirred at 0°C for 30 min then at 100°C for 6 h. The reaction mixture was allowed to cool to room temperature, then was poured into water (100mL). The aqueous layer was directly concentrated under lyophilization. The residue was purified by flash column chromatography on silica gel eluting with 7% MeOH in DCM to afford the title product as an off white solid (2.0 g, 50%). Step 2: N-(cyclopropylmethyl)-4-[[5-fluoro-7-[6-(1 -hydroxy-1 -methyl-ethyl)-2- pyridyl]pyrrolo[2,3-d]pyrimidin-2-yl]amino]benzamide dihydrochloride

To a stirred solution of 4-[[5-fluoro-7-[6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridyl]pyrrolo[2, 3- d]pyrimidin-2-yl]amino]benzoic acid (0.2 g, 0.49 mmol) and cyclopropylmethanamine (0.061 ml_, 0.05 g, 0.70 mmol) in THF (8 ml.) was added TEA (0.2 ml_, 1 .43 mmol) under nitrogen. The resulting mixture was stirred at 0°C for 15 min. A 50% solution of propylphosphonic anhydride in ethyl acetate was added (0.93 ml_, 1 .56 mmol) was added. The reaction mixture was stirred at room temperature for 16 h. The reaction mixture was poured into water (50mL). The aqueous layer was extracted with ethyl acetate (2 x 50 ml_). The organic layers were combined and dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel eluting with 5% MeOH in DCM. The crude was further purified by preparative HPLC using HCI buffer to afford the title product as an off white solid (0.03 g, 12%).

EXAMPLE 32

4-ff5-fluoro-7-f6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridynpynOlo[2 , 3-dlpyrimidin-2-vnaminol-

N-tetrahydropyran-4-yl-benzamide dihydrochloride

The title product was prepared by an analogous method to that used for Example 31 except that in step 2, tetrahydro-2H-pyran-4-amine hydrochloride was used instead of cyclopropylmethanamine. Yield of final step: white solid, 0.04 g (10%).

EXAMPLE 33

4-[[5-fluoro-7-[6-(1 -hvdroxy-1 -methyl-ethyl)-2-pyridyllPyrrolo[2.3-dlPyrimidin-2-yl1amino1 -

N-phenyl-benzamide dihvdrochloride

The title product was prepared by an analogous method to that used for Example 31 except that in step 2, aniline was used instead of cyclopropylmethanamine. Yield of final step: yellow solid, 0.055 g (14%).

EXAMPLE 34

4-ff5-fluoro-7-f6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridynpyrrolo[2,3-dlpyrimidin-2-vnaminol-

N-(1 -methyl-4-piperidyl)benzamide dihydrochloride The title product was prepared by an analogous method to that used for Example 31 except that in step 2, 1 -methylpiperidin-4-amine was used instead of cyclopropylmethanamine. Yield of final step: off-white solid, 0.049 g (12%).

EXAMPLE 35

N-(1 -ethyl-4-piperidyl)-4-[[5-fluoro-7-[6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridyllpyrrolo[2,3- dlpyrimidin-2-yl1amino1benzamide dihydrochloride

The title product was prepared by an analogous method to that used for Example 34 except that in step 2, 1 -ethyl-4-piperidin amine dihydrochloride was used instead of 1 - methylpiperidin-4-amine. Yield of final step: off-white solid, 0.04 g (9%).

EXAMPLE 36

2-fluoro-4-[[7-[6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridyllpyrrolo[2,3-dlpyrimidin-2-yl1amino1 -

N-(1 -methyl-4-pipehdvDbenzamide dihydrochloride

The title product was prepared by an analogous method to that used for Example 31 with the following exceptions: a) in step 1 , 2-[6-(2-chloropyrrolo[2,3-d]pyrimidin-7-yl)-2- pyridyl]propan-2-ol was used instead of 2-[6-(2-chloro-5-fluoro-pyrrolo[2,3-d]pyrimidin-7- yl)-2-pyridyl]propan-2-ol; and 4-amino-2-fluorobenzoic acid was used instead of 4- aminobenzoic acid b) in step 2, 1 -methylpiperidin-4-amine was used instead of cyclopropylmethanamine. Yield of final step: yellow solid, 0.025 g (15%).

EXAMPLE 37

2-chloro-4-[[5-fluoro-7-[6-(1 -hydroxy- 1 -methyl-ethyl)-2-pyridynpyrrolo[2,3-d]pyrimidin-2- yl]aminol-N-(1 -methyl-4-piperidyl)benzamide dihydrochloride

The title product was prepared by an analogous method to that used for Example 36 except that 4-amino-2-chlorobenzoic acid was used instead of 4-amino-2-fluorobenzoic acid in step 1 . Yield of final step: off-white solid, 0.016 g (5%).

EXAMPLE 38

2-[6-[5-fluoro-2-[4-(9-methyl-3.9-diazaspiro[5.51undecan- 3-yl)anilinolPyrrolof2.3- dlPyrimidin-7-yl1-2-pyridyllpropan-2-ol dihydrochloride The title product was prepared by following the same method as for Example 20 except that in step 2, 4-(9-methyl-3,9-diazaspiro[5.5]undecan-3-yl)aniline was used instead of 4- (4-ethylpiperazin-1-yl)aniline. Yield of final step: yellow solid, 0.112 g (11%).

4-(9-methyl-3,9-diazaspiro[5.5]undecan-3-yl)aniline was prepared as follows: Step 1 : 9-methyl-3-(4-nitrophenyl)-3,9-diazaspiro[5.5]undecane

To a stirred solution of 1 -fluoro-4-nitrobenzene (0.50 g, 3.55 mmol) and 3-methyl-3,9- diazaspiro[5.5]undecane (0.85 g, 3.52 mmol, commercially available) in DMSO (10 ml.) was added K 2 CO 3 (1.46 g, 10.6 mmol) at room temperature. The reaction mixture was heated at 120°C for 2 h. The reaction mixture was allowed to cool to room temperature and was poured into ice cold water (50 ml.) with stirring for 15 min. The precipitated solid was filtered off and dried in vacuo to afford title product as a yellow solid (1 .3 g, 64%).

Step 2: 4-(9-methyl-3,9-diazaspiro[5.5]undecan-3-yl)aniline

To a stirred solution of 9-methyl-3-(4-nitrophenyl)-3,9-diazaspiro[5.5]undecane (1 .3 g, 4.5 mmol) in methanol (20 ml.) was added 10% Pd/C (0.13 g, 10% w/w) at room temperature. The reaction mixture was purged by bubbling hydrogen through at room temperature for 2 h. The reaction mixture was filtered through celite, washing through with methanol (2 x 20 ml.) at room temperature. The combined filtrate was concentrated in vacuo to afford title product as a pink solid (1 .0 g, 86%).

EXAMPLES 39-45 Examples 39-45 were prepared via acetylation of compounds already described herein by analogous method to that described for Example 28. The table below describes the starting material and product formed.

EXAMPLE 46

2-fluoro-4-[[5-fluoro-7-[6-(1 -hvdroxy-1 -methyl-ethyl)-2-pyridyllPyrrolo[2.3-dlpyrimidin-2- yllaminol-N-d -methyl-4-DiDeridyl)benzamide dihvdrochloride Example 46 was prepared by an analogous method to that used for Example 36 except that in step 1 , 2-[6-(2-chloro-5-fluoro-pyrrolo[2,3-d]pyrimidin-7-yl)-2-pyri dyl]propan-2-ol was used instead of 2-[6-(2-chloropyrrolo[2,3-d]pyrimidin-7-yl)-2-pyridyl]propan -2-ol. Yield: off-white solid, 0.022 g (31%). EXAMPLE 47

5-fluoro-7-[6-(3-fluorc)Oxetan-3-vD-2-pyridyl1-N-[4-(4-me thylpiperazin-1- vDphenyllpyrrolo[2,3-dlpyrimidin-2-amine

Step 1 : 2-chloro-5-fluoro-7-[6-(3-fluorooxetan-3-yl)-2-pyridyl]pyrro lo[2,3-d]pyrimidine

To a stirred mixture of 2-chloro-5-fluoro-7H-pyrrolo[2,3-d]pyrimidine (0.5 g, 2.92 mmol) and 2-bromo-6-(3-fluorooxetan-3-yl)pyridine (0.81 g, 3.51 mmol, see below for preparation) in 1 ,4-dioxane (5 mL) was added K 3 PO 4 (1 .7 g, 8.01 mmol) at room temperature under nitrogen. The reaction mixture was degassed by bubbling nitrogen through for 30 min. Copper iodide (0.111 g, 0.58 mmol) and trans-N,N'-Dimethyl-1 ,2- cyclohexanediamine (0.083 g, 0.58 mmol) were added to the reaction mixture at room temperature. The reaction mixture was heated at 100°C for 3 h. The reaction mixture was allowed to cool to room temperature and was poured into water (50 mL). The aqueous layer was extracted with ethyl acetate (2 x 100 mL). The combined organic layers were dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel eluting with 10% ethyl acetate in hexane to afford title product as green solid (0.38 g, 40%).

Step 2: 5-fluoro-7-[6-(3-fluorooxetan-3-yl)-2-pyridyl]-N-[4-(4-methy lpiperazin-1 - yl)phenyl]pyrrolo[2,3-d]pyrimidin-2-amine

To a stirred mixture of 2-chloro-5-fluoro-7-[6-(3-fluorooxetan-3-yl)-2-pyridyl]pyrro lo[2,3- d]pyrimidine (0.3 g, 0.93 mmol) and 4-(4-methylpiperazin-1-yl)aniline (0.17 g, 0.89 mmol) in THF (6 mL) was added dropwise a 1 M solution of Li-HMDS in THF (4.65 mL, 4.65 mmol) at 0°C under nitrogen. The reaction mixture was stirred at room temperature for 6h. The reaction mixture was quenched by adding a saturated aqueous solution of sodium bicarbonate (20 mL) which was then extracted with ethyl acetate (2 x 50 mL). The combined organic layers were dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The obtained residue was purified by flash column chromatography on basic silica eluting with 10% MeOH in DCM to afford title product as an off white solid (0.090 g, 21%)

2-bromo-6-(3-fluorooxetan-3-yl)pyridine was prepared as follows:

Step 1 : 3-(6-bromo-2-pyridyl)oxetan-3-ol To a stirred solution of 2,6-dibromopyridine (5.0 g, 21.1 mmol) in DCM (50 ml.) was added dropwise a 2.5M solution of n-BuLi in hexane (9.4 ml_, 23.50 mmol) under nitrogen at -60°C. The reaction mixture was allowed to stir at -60°C for 15 min. Oxetan-3-one (1 .2 ml_, 22.7 mmol, commercially available: CAS: 6704-31 -0) was added dropwise to the reaction mixture at -60°C under nitrogen. The reaction mixture was quenched by adding a saturated aqueous solution of ammonium chloride (20 ml.) which was then extracted with DCM (2 x 50 ml_). The organic layers were combined and dried over anhydrous Na 2 S0 4 , filtered and concentrated in vacuo. The obtained residue was purified by flash column chromatography on neutral alumina eluting with 30% ethyl acetate in hexane to afford title product as an off white solid (4.8g, 93%).

Step 2: 2-bromo-6-(3-fluorooxetan-3-yl)pyridine

To a stirred solution of 3-(6-bromopyridin-2-yl)oxetan-3-ol (2.0 g, 8.73 mmol) in DCM (80 ml.) was added dropwise (Diethylamino)sulfur trifluoride (DAST) (2.3 ml_, 17.4 mmol) at - 10°C. The reaction mixture was stirred at -10°C for 30 min. The reaction mixture was quenched by adding a saturated aqueous solution of sodium bicarbonate (20 ml.) which was then extracted with DCM (2 x 50 ml_). The combined organic layers were dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The obtained residue was purified by trituration using n-pentane (2 x 20 ml_); the pentane was decanted off and the solid was dried under high vacuum to afford title product as an off white solid (1 .8 g, 89%).

EXAMPLE 48

7-[6-[[dimethyl(oxo)-lambda6-sulfanylidene1amino1-2-pyrid yl1-N-[4-(4-ethylpiperazin-1 - yl)phenyl1-5-fluoro-pyrrolo[2.3-dlPyrimidin-2-amine dihvdrochloride

The title product was prepared by an analogous method to that used for Example 17 except that in step 2, 4-(4-ethylpiperazin-1 -yl)aniline was used instead of 4-(4- methylpiperazin-1 -yl)aniline.

EXAMPLE 49

1 -[7-[4-[[7-[6-[[dimethyl(oxo)-lambda6-sulfanylidene1amino1-2 -pyridyl1-5-fluoro-pyrrolof2.3- dlPyrimidin-2-yl1aminolphenyl1-4.7-diazaspiro[2.51octan-4-yl 1ethanone The title product was prepared by an analogous method to that used for Example 41 with the exception that in step 1 , [6-(2-chloro-5-fluoro-pyrrolo[2,3-d]pyrimidin-7-yl)-2- pyridyl]imino-dimethyl-oxo-lambda6-sulfane (refer to Example 17, step 1 for preparation) was used instead of 2-[6-(2-chloro-5-fluoro-pyrrolo[2,3-d]pyrimidin-7-yl)-2-pyri dyl]propan- 2-ol.

EXAMPLE 50

7-[6-[[dimethyl(oxo)-lambda6-sulfanylidene1amino1-2-pyrid yl1-5-fluoro-N-[4-(9-methyl-3 , 9- diazaspiro[5.51undecan-3-yl)phenyllpyrrolo[2,3-dlpyrimidin-2 -amine

The title product was prepared by an analogous method to that used for Example 17 except that in step 2, 4-(9-methyl-3,9-diazaspiro[5.5]undecan-3-yl)aniline (refer to example 62 for preparation) was used instead of 4-(4-methylpiperazin-1 -yl)aniline.

EXAMPLE 51

N4-[7-[6-[[dimethyl(oxo)-lambda6-sulfanylidene1amino1-2-p yridyl1-5-fluoro-pyrrolof2.3- dlPyrimidin-2-yl1-N1 -tetrahvdropyran-4-yl-benzene-1 .4-diamine

The title product was prepared by an analogous method to that used for Example 17 except that in step 2, N1 -tetrahydropyran-4-ylbenzene-1 ,4-diamine (refer to Example 16) was used instead of 4-(4-methylpiperazin-1 -yl)aniline.

EXAMPLE 52

7-[6-[[dimethyl(oxo)-A 6 -sulfanylidene1amino1-2-pyridyl1-5-fluoro-N-[4-[[[(3S) - tetrahvdrofuran-3-yl1amino1methyllphenyllPyrrolo[2.3-dlPyrim idin-2-amine dihvdrochloride

The title compound was prepared by coupling together [6-(2-chloro-5-fluoro-pyrrolo[2,3- d]pyrimidin-7-yl)-2-pyridyl]imino-dimethyl-oxo-A 6 -sulfane (refer to Example 17 for preparation) with tert-butyl N-[(4-aminophenyl)methyl]-N-[(3S)-tetrahydrofuran-3- yl]carbamate (refer to Example 2 for preparation) using the following conditions:

Step 1 : tert-butyl N-[[4-[[7-[6-[[dimethyl(oxo)-A 6 -sulfanylidene]amino]-2-pyridyl]-5-fluoro- pyrrolo[2,3-d]pyrimidin-2-yl]amino]phenyl]methyl]-N-[(3S)-te trahydrofuran-3-yl]carbamate

To a stirred solution of [6-(2-chloro-5-fluoro-pyrrolo[2,3-d]pyrimidin-7-yl)-2-pyridy l]imino- dimethyl-oxo-A 6 -sulfane (0.300 g, 0.883 mmol) and tert-butyl N-[(4-aminophenyl)methyl]- N-[(3S)-tetrahydrofuran-3-yl]carbamate (0.206 g, 0.705 mmol) in 1 ,4-dioxane (10 ml.) was added CS2CO3 (0.575 g, 1 .76 mmol). The reaction mixture was purged with nitrogen gas for 20 min. Xantphos (0.120 g, 0.207 mmol) and Pd(OAc)2 (0.060 g, 0.267 mmol) were added and the reaction mixture was stirred in a microwave reactor at 100°C for 1 h. The reaction mixture was then poured into water (100 ml.) and extracted with ethyl acetate (2 x 50 ml_). The combined organic layer was dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The residue was purified by flash column chromatography on basic alumina, eluting with 7% methanol in DCM to afford the title product as a yellow solid (0.440 g, >100% * ). trapped solvent

Step 2: 7-[6-[[dimethyl(oxo)-A 6 -sulfanylidene]amino]-2-pyridyl]-5-fluoro-N-[4-[[[(3S) - tetrahydrofuran-3-yl]amino]methyl]phenyl]pyrrolo[2,3-d]pyrim idin-2-amine dihydrochloride

To a stirred solution of tert-butyl N-[[4-[[7-[6-[[dimethyl(oxo)-A 6 -sulfanylidene]amino]-2- pyridyl]-5-fluoro-pyrrolo[2,3-d]pyrimidin-2-yl]amino]phenyl] methyl]-N-[(3S)- tetrahydrofuran-3-yl]carbamate (0.440 g, 0.739 mmol) in DCM (10 ml.) was added a solution of 4 M HCI in dioxane (2 ml.) at 0°C. The reaction mixture was stirred at room temperature for 24 h. The reaction mixture was concentrated in vacuo and triturated with diethyl ether (10 ml_). The solid material was filtered off and dried in vacuo then further purified by preparative HPLC using Method M. The resultant fractions were lyophilized to afford title product as a yellow solid (0.170 g, 41%).

EXAMPLE 53

7-f6-[(ethyl-methyl-oxo-A 6 -sulfanylidene)aminol-2-pvridyl]-5-fluoro-N-f4-(4- methvlpiperazin-1 -vl)phenvnpvrrolo[2,3-d]pvrimidin-2-amine

The title compound was prepared from 2-chloro-5-fluoro-7H-pyrrolo[2,3-d]pyrimidine (0.6 g, 3.50 mmol) by the same method as described for Example 17 except that in Step 1 , (6-bromo-2-pyridyl)imino-ethyl-methyl-oxo-A 6 -sulfane (1 .07, 4.07 mmol) was used instead of (6-bromo-2-pyridyl)imino-dimethyl-oxo- A 6 -sulfane. The final product was purified by preparative HPLC using Method N to afford the title product as a cream solid, 0.029 g (11%).

Preparation of (6-bromo-2-pyridyl)imino-ethyl-methyl-oxo-A 6 -sulfane:

To a solution of 2, 6-dibromopyridine (2.20 g, 9.3 mmol) in toluene (25 mL) was added ethyl-imino-methyl-oxo-A 6 -sulfane (1 .0 g, 9.3 mmol) and sodium tert-butoxide (0.89 g, 9.3 mmol) at room temperature. The reaction mixture was purged with nitrogen gas for 20 min at room temperature. Tris(dibenzylideneacetone)dipalladium(0) (0.16 g, 0.18 mmol) and rac-BINAP (0.34 g, 0.55 mmol) were added to the reaction mixture at room temperature. The reaction mixture was further stirred at 120°C for 4h. The reaction mixture was cooled to room temperature, poured into ice-cold water (100 ml.) and then extracted with ethyl acetate (2 x 150 ml_). The combined organic layer was dried over sodium sulphate, filtered and concentrated in vacuo. The residue was purified by flash chromatography on silica gel eluting with 20% ethyl acetate in Hexane to afford the title product (1.1 g, 45%).

EXAMPLE 54

7-[6-[(cvclopropyl-methyl-oxo-A 6 -sulfanylidene)amino1-2-pyridyl1-5-fluoro-N-[4-(4- methylpiperazin-1 -yl)phenyllpyrrolo[2,3-dlpyrimidin-2-amine dihvdrochloride

The title compound was prepared from 2-chloro-5-fluoro-7H-pyrrolo[2,3-d]pyrimidine (0.55 g, 3.22 mmol) by the same method as described for Example 17 except that in Step 1 , (6-bromo-2-pyridyl)imino-cyclopropyl-methyl-oxo-A 6 -sulfane (1 .05 g, 3.82 mmol) was used instead of (6-bromo-2-pyridyl)imino-dimethyl-oxo- A 6 -sulfane. The final product was purified by preparative HPLC using method O to afford the title product as a cream solid, 0.07 g (22%).

Preparation of (6-bromo-2-pyridyl)imino-cyclopropyl-methyl-oxo-A 6 -sulfane:

To a solution of 2,6-dibromopyridine (1 .98 g, 8.36 mmol) in toluene (25 mL) was added cyclopropyl-imino-methyl-oxo-A 6 -sulfane (1 .0 g, 8.39 mmol) followed by sodium tert- butoxide (0.80 g, 8.32 mmol) at room temperature. The reaction mixture was purged with nitrogen gas for 20 min at room temperature. Tris(dibenzylideneacetone)dipalladium(0) (0.15 g, 0.164 mmol) and rac-BINAP (0.31 g, 0.498 mmol) were added to the reaction mixture at room temperature. The reaction mixture was heated at 120°C for 4h. The reaction mixture was then cooled to room temperature, poured into ice-cold water (100 mL) and extracted with ethyl acetate (2 x 150 mL). The combined organic layer was dried over sodium sulphate, filtered and concentrated in vacuo. The residue was purified by flash chromatography on silica gel, eluting with 22% ethyl acetate in hexane to afford the title product (1.1 g, 48%).

EXAMPLE 55 N-[3-chloro-4-(4-methylpiperazin-1-yl)phenyl1-7-[6-[fdimethy l(oxo)-A 6 - sulfanylidene1amino1-2-pyridyl1-5-fluoro-pyrrolo[2,3-dlpyrim idin-2-amine dihydrochloride

The title compound was prepared by coupling together [6-(2-chloro-5-fluoro-pyrrolo[2,3- d]pyrimidin-7-yl)-2-pyridyl]imino-dimethyl-oxo-A 6 -sulfane (refer to Example 17 for preparation) with 3-chloro-4-(4-methylpiperazin-1-yl)aniline using the following conditions:

To a mixture of [6-(2-chloro-5-fluoro-pyrrolo[2,3-d]pyrimidin-7-yl)-2-pyridy l]imino-dimethyl- oxo-A 6 -sulfane (0.3 g, 0.88 mmol) and 3-chloro-4-(4-methylpiperazin-1-yl)aniline (0.15 g, 0.665 mmol) in DMF (5 ml.) was added K 3 PO 4 (0.37 g, 1 .74 mmol) at room temperature under nitrogen. The resulting reaction mixture was purged with nitrogen for 30 min at room temperature. Bis(tri-tert-butylphosphine)palladium(0) (0.026 g, 0.0509 mmol) was added to the reaction mixture. The reaction mixture was stirred at 140°C for 2 h under nitrogen. The reaction mixture was then allowed to cool to room temperature and poured into ice cold water (20 ml_). The aqueous layer was extracted with ethyl acetate (2 x 100 ml_). The combined organic layer was dried over anhydrous Na 2 SC> 4 , filtered and concentrated in vacuo. The residue was purified by flash chromatography on silica, eluting with 5% Methanol in DCM. The residue was further purified by prep. HPLC purification using Method P, followed by lyophilization to afford the title product as a green solid (0.07 g, 17%).

3-chloro-4-(4-methylpiperazin-1-yl)aniline was prepared as follows:

Step 1 : 1-(2-chloro-4-nitro-phenyl)-4-methyl-piperazine

To a solution of 2-chloro-1-fluoro-4-nitrobenzene (5.0 g, 28.48 mmol) in DMF (50 ml.) was added potassium carbonate (19.63 g, 142.05 mmol). N-methylpiperazine (4.86 g, 5.38 ml_, 48.52 mmol) was added at room temperature. The reaction mixture was stirred at 120°C for 16h. The reaction mixture was cooled to room temperature and poured into water (80 ml_). The precipitated solid was filtered off and washed with water (100 ml.) under vacuum. The solid was dried well under vacuum to afford the title product as a yellow solid (5.0g, 69%).

Step 2: 3-chloro-4-(4-methylpiperazin-1-yl)aniline

To a stirring solution of 1-(2-chloro-4-nitrophenyl)-4-methylpiperazine (2.5 g, 9.80 mmol) in methanol: water (30: 3) was added ammonium chloride (5.2 g, 97.2 mmol) followed by iron powder (2.72 g, 48.7 mmol) at room temperature. The reaction mixture was stirred at 70°C for 16 h. The reaction mixture was filtered through celite and the filtrate was concentrated in vacuo to afford the title product (1 .5g, 68%).

EXAMPLE 56

7-[6-[(ethyl-methyl-oxo-A 6 -sulfanylidene)amino1-2-pyridyl1-5-fluoro-N-[4-(4- methylpiperazin-1 -yl)phenyllpyrrolo[2,3-dlpyrimidin-2-amine (enantiomer 1 )

Yield: cream solid, 0.07 g.

EXAMPLE 57

7-[6-[(ethyl-methyl-oxo-A 6 -sulfanylidene)amino1-2-pyridyl1-5-fluoro-N-[4-(4- methylpiperazin-1 -yl)phenyllpyrrolo[2,3-dlpyrimidin-2-amine (enantiomer 2) Yield: cream solid, 0.07 g.

The enantiomers of Examples 56 and 57 were each isolated following separation of the racemate, example 53, using chiral prep. HPLC method 2. Example 56 was the first- eluting isomer (retention time 14.913 min) whereas Example 57 was the second-eluting isomer (retention time 16.630 min). Absolute configuration of the enantiomers has not been determined.

ANALYTICAL DATA FOR EXAMPLES 1 TO 57

BIOLOGICAL ACTIVITY

(a) Determination of HUMAN Wee1 inhibitory activity

The ability of compounds of the invention to inhibit human Wee1 kinase may be determined using the protocol below. Test compounds are prepared to 50x final assay concentration in 100% DMSO. This working stock of the compound is added to the assay well as the first component in the reaction, followed by the remaining components as detailed in the general assay protocols below.

Method: Kinases are diluted to the appropriate concentration in the following buffer prior to addition to the reaction mix: 20 mM TRIS, 0.2 mM EDTA, 0.1% b-mercaptoethanol,

0.01% Brij-35, 5% glycerol.

Wee1 (h) kinase assay:

Wee1 (h) is incubated with 20 mM Tris/HCI pH 8.5, 0.2 mM EDTA, 500 mM LSNLYHQGKFLQTFCGSPLYRRR, 10 mM magnesium acetate and [y- 33 P]-ATP (specific activity and concentration as required). The reaction is initiated by the addition of the Mg/ATP mix. After incubation for 40 minutes at room temperature, the reaction is stopped by the addition of phosphoric acid to a concentration of 0.5%. 10 pL of the reaction is then spotted onto a P30 filtermat and washed four times for 4 minutes in 0.425% phosphoric acid and once in methanol prior to drying and scintillation counting.

The Wee1 inhibitory activity of compounds of the invention was tested using the protocol described above and the results are shown in the table below.

(b) Determination of HUMAN PLK1 inhibitory activity

The ability of compounds of the invention to inhibit human PLK1 kinase may be determined using the protocol below. Test compounds are prepared to 50x final assay concentration in 100% DMSO. This working stock of the compound is added to the assay well as the first component in the reaction, followed by the remaining components as detailed in the general assay protocols below.

Method: Kinases are diluted to the appropriate concentration in the following buffer prior to addition to the reaction mix: 20 mM MOPS, 1 mM EDTA, 0.01% Brij-35, 5% Glycerol, 0.1% b-mercaptoethanol, 1 mg/ml_ BSA.

PLK1 (h) kinase assay: PLK1 (h) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 20 mM DTT, 1 .25 mg/ml casein, 10 mM Magnesium acetate and [g- 33 R]-ATR (specific activity and concentration as required). The reaction is initiated by the addition of the Mg/ATP mix. After incubation for 40 minutes at room temperature, the reaction is stopped by the addition of phosphoric acid to a concentration of 0.5%. 10 mI_ of the reaction is then spotted onto a P30 filtermat and washed four times for 4 minutes in 0.425% phosphoric acid and once in methanol prior to drying and scintillation counting.

The Plk1 inhibitory activity of compounds of the invention was tested using the protocol described above and the results are shown in the table below. 10 (c) Determination of IC50 values in OVCAR3 cells following treatment with test compounds

1 . OVCAR3 cells are seeded at a concentration of 5000 cells per well in 75 mI_ media into 96 well flat-bottomed tissue culture plates which are then placed in an incubator at 37°C, 5% CO2 overnight for the cells to adhere.

2. Cells are then treated with relevant concentrations of test compound in 25 mI_ to give final concentrations of 10, 3, 1 , 0.3, 0.1 , 0.03, 0.01 , 0.003 and 0.001 uM for 72 hours.

3. After incubation, plates are removed from the incubator and viability established using the Cell Titer 96 Aqueous One solution Assay. Percentage viability can be calculated against the mean of the DMSO treated control samples, and IC 50 values for inhibition of cell growth can be calculated using GraphPad Prism software by nonlinear regression (4 parameter logistic equation) with bottom and top constraints at 0 and 100%, respectively.

The OVCAR3 anti-proliferation activity of compounds of the invention was tested using the protocol described above and the results are shown in the table below.

(d) Determination of values in HT29 cells following treatment with test compounds 1 . HT29 cells are seeded at a concentration of 2500 cells per well in 75 mI_ media into 96 well flat-bottomed tissue culture plates and then placed in an incubator at 37°C, 5% CO2 overnight for the cells to adhere.

2. Cells are then treated with relevant concentrations of test compound in 25 mI_ to give final concentrations of 10, 3, 1 , 0.3, 0.1 , 0.03, 0.01 , 0.003 and 0.001 uM for 72 hours.

3. After incubation, plates are removed from the incubator and viability established using the Cell Titer 96 Aqueous One solution Assay.

Percentage viability can be calculated against the mean of the DMSO treated control samples, and IC 5 o values for inhibition of cell growth can be calculated using GraphPad Prism software by nonlinear regression (4 parameter logistic equation) with bottom and top constraints at 0 and 100%, respectively.

The HT29 anti-proliferation activity of compounds of the invention was tested using the protocol described above and the results are shown in the table below (e) In vivo pharmacokinetics: Evaluation of brain. CSF and plasma concentrations in an in vivo cassette mouse model

Compounds of the invention were evaluated in an in vivo mouse model to determine brain and plasma concentrations following oral dosing. This is an industry-standard and recognised means to assess brain penetration of small molecules. It is recognised that higher brain concentrations (and higher ratios of brain : plasma concentration) lead to greater exposure in the brain - this is clearly advantageous in order to treat cancers of the brain. In addition, concentration in Cerebrospinal Fluid (CSF) gives an indication of the amount of compound in the CNS, free of protein binding.

Experimental method:

A two-arm study was used: the first arm was intravenous (IV) dose whereas the second arm was oral (PO) dose. Study design was as follows:

Results:

Example 17 was found to be highly orally bioavailable (>90%) with good levels of exposure observed in plasma, brain and CSF from the oral dose (see the table below). A brain: plasma ratio of greater than 0.5 was observed based on AUC. These data support the proposition that compounds of this invention can penetrate the brain effectively, which is a pre-requisite to treating cancers of the brain such as Medulloblastoma, DIPG and GBM.

PHARMACEUTICAL FORMULATIONS

(0 Tablet Formulation

A tablet composition containing a compound of the formula (1) as defined in any one of Embodiments 1 .1 to 1 .76 may be prepared by mixing 50 mg of the compound with 197 mg of lactose (BP) as diluent, and 3 mg magnesium stearate as a lubricant and compressing to form a tablet in known manner.

(ii) Capsule Formulation

A capsule formulation is prepared by mixing 100 mg of a compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1 .76 with 100 mg lactose and filling the resulting mixture into standard opaque hard gelatin capsules.

(iii) Injectable Formulation I

A parenteral composition for administration by injection can be prepared by dissolving a compound of the formula (1 ) as defined in any one of Embodiments 1.1 to 1.76 in water containing 10% propylene glycol to give a concentration of active compound of 1 .5 % by weight. The solution is then sterilised by filtration, filled into an ampoule and sealed.

(iv) Injectable Formulation

A parenteral composition for injection is prepared by dissolving in water a compound of the formula (1 ) as defined in any one of Embodiments 1 .1 to 1 .76 (2 mg/ml) and mannitol (50 mg/ml), sterile filtering the solution and filling into sealable 1 ml vials or ampoules. v) Injectable formulation III

A formulation for i.v. delivery by injection or infusion can be prepared by dissolving the compound of formula (1 ) as defined in any one of Embodiments 1.1 to 1 .76 (e.g. in a salt form) in water at 20 mg/ml. The vial is then sealed and sterilised by autoclaving. vi) Injectable formulation IV A formulation for i.v. delivery by injection or infusion can be prepared by dissolving the compound of formula (1 ) as defined in any one of Embodiments 1.1 to 1 .76 (e.g. in a salt form) in water containing a buffer (e.g. 0.2 M acetate pH 4.6) at 20mg/ml. The vial is then sealed and sterilised by autoclaving. (vii) Subcutaneous Injection Formulation

A composition for sub-cutaneous administration is prepared by mixing a compound of the formula (1 ) as defined in any one of Embodiments 1 .1 to 1.76 with pharmaceutical grade corn oil to give a concentration of 5 mg/ml. The composition is sterilised and filled into a suitable container. viii) Lyophilised formulation

Aliquots of formulated compound of formula (1) as defined in any one of Embodiments 1 .1 to 1 .76 are put into 50 ml vials and lyophilized. During lyophilisation, the compositions are frozen using a one-step freezing protocol at {-45 e C). The temperature is raised to - 10 e C for annealing, then lowered to freezing at -45 e C, followed by primary drying at +25 e C for approximately 3400 minutes, followed by a secondary drying with increased steps if temperature to 50 e C. The pressure during primary and secondary drying is set at 80 millitor.

Equivalents

The foregoing examples are presented for the purpose of illustrating the invention and should not be construed as imposing any limitation on the scope of the invention. It will readily be apparent that numerous modifications and alterations may be made to the specific embodiments of the invention described above and illustrated in the examples without departing from the principles underlying the invention. All such modifications and alterations are intended to be embraced by this application.