WO2021074251A1 - Dérivés de pyrrolo[2,3-d]pyrimidine et leur utilisation dans le traitement du cancer - Google Patents

Dérivés de pyrrolo[2,3-d]pyrimidine et leur utilisation dans le traitement du cancer Download PDF

Info

Publication number
WO2021074251A1
WO2021074251A1 PCT/EP2020/078960 EP2020078960W WO2021074251A1 WO 2021074251 A1 WO2021074251 A1 WO 2021074251A1 EP 2020078960 W EP2020078960 W EP 2020078960W WO 2021074251 A1 WO2021074251 A1 WO 2021074251A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
mmol
compound according
reaction mixture
hydrogen
Prior art date
Application number
PCT/EP2020/078960
Other languages
English (en)
Inventor
Robert George Boyle
David Winter Walker
Meriel Ruth Major
Original Assignee
Sentinel Oncology Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sentinel Oncology Limited filed Critical Sentinel Oncology Limited
Publication of WO2021074251A1 publication Critical patent/WO2021074251A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • 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.
  • 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 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.
  • Cdk1 complexes with Cyclin B into order to induce 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.
  • ATR ataxia-telangiectasia-related
  • 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.
  • 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.
  • 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).
  • DIPG Diffuse Intrinsic Pontine Glioma
  • Ependymoma see Kool et al, “Therapeutic Targeting of Ependymoma as Informed by Oncogenic Enhancer Profiling”, Nature. 2018 January 04; 553(7686):
  • 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);
  • Uterine Serous Carcinoma USC
  • 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).
  • a further chemotherapeutic agent e.g. gemcitabine or carboplatin.
  • ADME Absorption, Distribution, Metabolism, Excretion
  • 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.
  • 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.
  • 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.
  • 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.
  • GAP GTPase-activating protein
  • 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.
  • GEF Guanine Nucleotide Exchange Factor
  • PLK1 Polo- Like Kinase 1
  • 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
  • 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:
  • 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
  • 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.
  • 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.
  • R 2 is selected from hydrogen, fluorine, chlorine, C1-3 alkyl, C1-3 alkoxy, difluoromethyl and trifluoromethyl.
  • R 2 is selected from hydrogen, fluorine, chlorine, methyl, ethyl, methoxy, trifluoromethyl and difluoromethyl.
  • R c is selected from hydrogen, C1-3 alkyl, C3-4 cycloalkyl and cyclopropylmethyl.
  • a 5- to 7-membered monocyclic, heterocyclic group e.g. a non-aromatic group
  • a 7- to 11 - membered bicyclic heterocyclic group e.g. a non-aromatic group
  • 1 or 2 heteroatom ring members for example 2 nitrogen heteroatom ring members
  • Cyc 1 is selected from:
  • a 5- to 7-membered monocyclic, heterocyclic group e.g. a non-aromatic group
  • a 5- to 7-membered monocyclic, heterocyclic group e.g. a non-aromatic group
  • 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
  • a non-aromatic group such as a non-aromatic bridged bicyclic or spiro-bicyclic group
  • 1 or 2 nitrogen heteroatom ring members for example 2 nitrogen heteroatom ring members
  • Cyc 1 is selected from:
  • 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 ;
  • 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 ;
  • bicyclic non-aromatic group heterocyclic group e.g. a bridged bicyclic or spiro-bicyclic group
  • R 7 substituents selected from R 7 ;
  • bicyclic non-aromatic group heterocyclic group e.g. a bridged bicyclic or spiro-bicyclic group
  • R 7 substituents selected from R 7 ;
  • 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.
  • 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.
  • 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.
  • 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.
  • R a is selected from hydrogen, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, cyclopropylmethyl and f-butyl.
  • R b is selected from hydrogen, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, cyclopropylmethyl, methylcyclopropyl and f-butyl.
  • 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:
  • R 6 is selected from hydrogen, halogen, C1-4 alkyl, C1-2 alkoxy and C1-2 haloalkyl.
  • 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.
  • R 6 is selected from hydrogen, fluorine and chlorine.
  • references to “carbocyclic” and “heterocyclic” groups as used herein shall, unless the context indicates otherwise, include both aromatic and non -aromatic ring systems.
  • 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.
  • aryl refers to a carbocyclic group having aromatic character and the term “heteroaryl” is used herein to denote a heterocyclic group having aromatic character.
  • 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.
  • non-aromatic group refers to unsaturated ring systems without aromatic character, partially saturated and fully saturated carbocyclic and heterocyclic ring systems.
  • 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.
  • hydrocarbyl refers to aliphatic, alicyclic and aromatic groups having an all-carbon backbone and consisting of carbon and hydrogen atoms, except where otherwise stated.
  • 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.
  • one or more, but not all, of the carbon atoms of the hydrocarbyl group may be replaced by another atom or group of atoms.
  • alkylene refers to an alkanediyl group, i.e. a divalent saturated acyclic straight chain or branched chain hydrocarbon group.
  • straight chain alkanediyl groups include methylene (CH 2 ), ethylene (CH 2 CH 2 ) and propylene ((CH 2 CH 2 CH 2 ).
  • branched chain alkanediyl groups include CH(CH 3 ), CH 2 CH(CH 3 )CH2 and CH 2 (CH 3 )CH 2 CH2.
  • haloalkyl 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.
  • Embodiments 1.1 to 1 .71 may be presented in the form of salts.
  • the salts referred to above 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.
  • 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 may be formed with a wide variety of acids, both inorganic and organic.
  • 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- 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, unde
  • 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.
  • 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 may form N-oxides.
  • a compound contains several amine functions
  • one or more than one nitrogen atom may be oxidised to form an N-oxide.
  • 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.
  • MCPBA m-chloroperoxybenzoic acid
  • 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.
  • tautomeric forms and references to the compounds of formula (1 ) as defined in Embodiments 1 .1 to 1 .77 include all such forms.
  • Optical Isomers 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.
  • 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.
  • 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.
  • 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.
  • chiral acids such as (+)-tartaric acid, (-)-pyroglutamic acid, (-)-di-toluoyl-L-tartaric acid, (+)-mandelic acid, (-)-malic acid, and (-)-camphorsulphonic
  • 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).
  • 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).
  • 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.
  • a reference to hydrogen includes within its scope 1 H, 2 H (D), and 3 H (T).
  • 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.
  • the compounds contain no radioactive isotopes. Such compounds are preferred for therapeutic use.
  • the compound may contain one or more radioisotopes. Compounds containing such radioisotopes may be useful in a diagnostic context.
  • 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).
  • a non-toxic pharmaceutically acceptable solvent referred to below as the solvating solvent.
  • 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.
  • TGE thermogravimetric analysis
  • DSC differential scanning calorimetry
  • X-ray crystallography X-ray crystallography
  • the solvates can be stoichiometric or non-stoichiometric solvates.
  • solvates are hydrates, and examples of hydrates include hemihydrates, monohydrates and dihydrates.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.).
  • the prodrug may be a sugar derivative or other glycoside conjugate, or may be an amino acid ester derivative.
  • complexes e.g. inclusion complexes or clathrates with compounds such as cyclodextrins, or complexes with metals
  • compounds such as cyclodextrins, or complexes with metals
  • 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).
  • 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.
  • the invention provides:
  • 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.
  • 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.
  • proliferative disorders e.g. cancers
  • proliferative disorders 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
  • proliferative disorders e.g. cancers
  • GBM glioblastoma
  • medulloblastoma medulloblastoma
  • ependymoma e.g. cancers
  • 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.
  • DIPG Diffuse Intrinsic Pontine Glioma
  • Uterine Serous Carcinoma USC
  • Uterine Carcinosarcoma Uterine Carcinosarcoma
  • 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:
  • DIPG Diffuse Intrinsic Pontine Glioma
  • 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).
  • 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).
  • 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).
  • 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).
  • 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).
  • mAb monoclonal antibody
  • the invention also provides
  • a further therapeutic agent e.g. a further therapeutic agent described herein
  • 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).
  • a further therapeutic agent e.g. a further therapeutic agent described herein.
  • a method for the diagnosis and treatment of a cancer which is characterised by mutant or deficient tumour protein p53 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.
  • 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 .
  • a method of treating a subject suffering from a cancer which is sensitive to Wee1 inhibition 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.
  • 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 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.
  • 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.
  • 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 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.
  • 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 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.
  • 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.
  • the invention also provides:
  • 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.
  • a method of treating a subject suffering from a cancer which is sensitive to PLK1 inhibition 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.
  • 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 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.
  • 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 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.
  • a method for the diagnosis and treatment of a disease state or condition mediated by PLK1 kinase 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.
  • 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 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.
  • 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 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.
  • 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.
  • a method of treating a subject suffering from a cancer which is sensitive to Wee1 and PLK1 inhibition 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.
  • 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 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.
  • 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 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.
  • a method for the diagnosis and treatment of a disease state or condition mediated by Wee1 and PLK1 kinase 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.
  • 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 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.
  • 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.
  • 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”.
  • 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.
  • IC 5 o against PLK1 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.
  • the invention provides:
  • 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.
  • 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).
  • 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.
  • 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.
  • 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.
  • a polar solvent such as dimethyl formamide (DMF) or dioxane
  • 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 ).
  • Hal is a halogen (e.g. chlorine)
  • the reaction between the compounds of formulae (12) and (13) typically takes place in the presence of a coupling catalyst.
  • 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.
  • a non-nucleophilic base such as pyridine
  • 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.
  • 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).
  • a cyanide salt e.g. Zn(CN) 2
  • a palladium catalyst such as Pd 2 (dba)3
  • 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.
  • 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)).
  • 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).
  • an acylating agent e.g. an anhydride of a carboxylic acid or an acyl chloride
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • compositions of the invention can be in any form suitable for oral, parenteral, topical, intranasal, intrabronchial, ophthalmic, otic, rectal, intra-vaginal, or transdermal administration.
  • 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.
  • the invention provides:
  • 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.
  • Embodiment 4.8 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.
  • 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.
  • Embodiment 4.12 A pharmaceutical composition according to Embodiment 4.11 which is a solution or suspension for injection or infusion.
  • compositions 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.
  • tablet compositions 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
  • effervescent agents such as citrate/bicarbonate mixtures.
  • Capsule formulations 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 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.
  • a protective film coating e.g. a wax or varnish
  • the coating e.g. a Eudragit TM type polymer
  • the coating can be designed to release the active component at a desired location within the gastro -intestinal tract.
  • 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.
  • 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.
  • 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.
  • 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 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.
  • 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.
  • the powdered formulations typically comprise the active compound together with an inert solid powdered diluent such as lactose.
  • 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.
  • 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.
  • 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)
  • Topoisomerase II inhibitors e.g. etoposide
  • mTOFt inhibitors e.g. Everolimus
  • PI3K pathway inhibitors e.g. PI3K, PDK1
  • 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)
  • hypoxia triggered DNA damaging agents e.g. Tirapazamine
  • 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
  • PARP inhibitors e.g. Olaparib
  • chemotherapeutic agents that may be co-administered with a compound as defined in any one Embodiments 1.1 to 1.77 include:
  • 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
  • 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 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.
  • a DNA damaging agent such as gemcitabine.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • a patient 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.
  • 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.
  • 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.
  • the patient may be subjected to a diagnostic test to detect a marker characteristic of up-regulation of Wee1/PL1 kinase.
  • diagnosis includes screening.
  • 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.
  • 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,
  • Screening methods could include, but are not limited to, standard methods such as reverse-transcriptase polymerase chain reaction (RT-PCR) or in-situ hybridisation.
  • RT-PCR reverse-transcriptase polymerase chain reaction
  • telomere amplification 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.
  • FISH fluorescence in-situ hybridisation
  • 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.
  • 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.
  • 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 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 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 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 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:
  • 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.
  • 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).
  • 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 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 :
  • 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) 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:
  • 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:
  • Step 1 2-[6-(5-bromo-2-chloro-pyrrolo[2,3-d]pyrimidin-7-yl)-2-pyridyl]propan-2-ol
  • 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
  • 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
  • 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.
  • Step 2 tert-butyl N-[(4-nitrophenyl)methyl]-N-[(3S)-tetrahydrofuran-3-yl]carbamate
  • 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]carbamate
  • Step 2 tert-butyl N-[(4-nitrophenyl)methyl]-N-tetrahydrofuran-3-yl-carbamate
  • Step 3 tert-butyl N-[(4-aminophenyl)methyl]-N-tetrahydrofuran-3-yl-carbamate
  • 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).
  • step 2 was purified by flash column chromatography on silica gel eluting with 4.5% methanol in DCM
  • 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 2 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.
  • Step 1 2-[6-(2-chloropyrrolo[2,3-d]pyrimidin-7-yl)-2-pyridyl]propan-2-ol
  • the resulting reaction mixture was purged by bubbling nitrogen through for 30 min.
  • copper iodide 0.77 g, 0.307 mL, 1.95 mmol
  • trans-N,N’- dimethylcyclohexane-1 ,2-diamine 0.277 g, 0.307 mL, 1.95 mmol
  • 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
  • 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
  • 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
  • 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%).
  • Step 1 2-[6-(2-chloro-5-fluoro-pyrrolo[2,3-d]pyrimidin-7-yl)-2-pyridyl]propan-2-ol
  • 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
  • Step 3 2-[6-[5-fluoro-2-(4-piperazin-1 -ylanilino)pyrrolo[2,3-d]pyrimidin-7-yl]-2- pyridyl]propan-2-ol hydrochloride
  • 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%).
  • step 2 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%).
  • step 2 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%).
  • Step 1 N-(1 -methyl-4-piperidyl)-4-nitro-benzamide
  • Step 2 4-amino-N-(1 -methyl-4-piperidyl)benzamide
  • N-(1 -methyl-4-piperidyl)-4-nitro-benzamide (1 .0 g, 3.8 mmol) in methanol (10 ml.)
  • 10% Pd/C 0.2 g, 20 mol%
  • 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%).
  • step 2 for synthesis of the title product Refer to Example 8, step 2 for synthesis of the title product.
  • 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
  • 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
  • Step 2 tert-butyl 7-(4-aminophenyl)-4,7-diazaspiro[2.5]octane-4-carboxylate
  • 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%).
  • 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
  • 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
  • Step 2 tert-butyl N-(4-nitrophenyl)-N-(tetrahydropyran-4-ylmethyl)carbamate
  • 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
  • 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
  • Step 1 [6-(2-chloro-5-fluoro-pyrrolo[2,3-d]pyrimidin-7-yl)-2-pyridyl]imino-dimethyl-oxo- Iambda6-sulfane
  • 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
  • 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
  • Step 2 tert-butyl 4-(4-amino-2-chloro-phenyl)piperazine-1 -carboxylate
  • 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 .
  • step 2 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.
  • 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
  • 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-carboxylate was prepared as follows:
  • Step 1 tert-butyl (1 R,4R)-5-(4-nitrophenyl)-2,5-diazabicyclo[2.2.1]heptane-2-carboxylate
  • Step 2 tert-butyl (1 R,4R)-5-(4-aminophenyl)-2,5-diazabicyclo[2.2.1]heptane-2- carboxylate
  • 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
  • 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_).
  • 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.
  • tert-butyl (S)-3-(4-aminophenoxy)piperidine-1- carboxylate was used instead of tert-butyl 4-(4-aminophenoxy)piperidine-1-carboxylate
  • 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
  • tert-butyl 4-(4-aminophenyl)-3-oxopiperazine-1 - carboxylate was used instead of tert-butyl 4,7-diazaspiro[2.5]octane-4-carboxylate
  • 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:
  • 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%).
  • Step 1 4-[[5-fluoro-7-[6-(1 -hydroxy-1 -methyl-ethyl)-2-pyridyl]pyrrolo[2,3-d]pyrimidin-2- yl]amino]benzoic acid
  • 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
  • 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%).
  • 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%).
  • 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 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-pyridyl]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
  • Step 1 2-chloro-5-fluoro-7-[6-(3-fluorooxetan-3-yl)-2-pyridyl]pyrrolo[2,3-d]pyrimidine
  • Step 2 5-fluoro-7-[6-(3-fluorooxetan-3-yl)-2-pyridyl]-N-[4-(4-methylpiperazin-1 - yl)phenyl]pyrrolo[2,3-d]pyrimidin-2-amine
  • 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%)
  • Step 1 3-(6-bromo-2-pyridyl)oxetan-3-ol
  • 2,6-dibromopyridine 5.0 g, 21.1 mmol
  • DCM DCM
  • n-BuLi n-BuLi in hexane
  • 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.
  • 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%).
  • 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%).
  • 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.
  • 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)-tetrahydrofuran-3-yl]carbamate
  • 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]pyrimidin-2-amine dihydrochloride
  • 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%).
  • 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%).
  • 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%).
  • 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%).
  • 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%).
  • 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:
  • Step 1 1-(2-chloro-4-nitro-phenyl)-4-methyl-piperazine
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • concentration in Cerebrospinal Fluid (CSF) gives an indication of the amount of compound in the CNS, free of protein binding.
  • IV intravenous
  • PO oral
  • 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 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.
  • BP lactose
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • Subcutaneous Injection Formulation e.g. 0.2 M acetate pH 4.6
  • 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.
  • 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.

Abstract

L'invention concerne des composés de formule (1) : ou un sel ou un tautomère de ceux-ci, A, R1, R2, R3, R4, R5 et R6 étant tels que définis dans la description. Les composés sont des inhibiteurs de la kinase Wee1 et/ou PLK1 et sont envisagés pour être utiles dans le traitement de cancers.
PCT/EP2020/078960 2019-10-15 2020-10-14 Dérivés de pyrrolo[2,3-d]pyrimidine et leur utilisation dans le traitement du cancer WO2021074251A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB1914910.3 2019-10-15
GB201914910A GB201914910D0 (en) 2019-10-15 2019-10-15 Pharmaceutical compounds

Publications (1)

Publication Number Publication Date
WO2021074251A1 true WO2021074251A1 (fr) 2021-04-22

Family

ID=68619675

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2020/078960 WO2021074251A1 (fr) 2019-10-15 2020-10-14 Dérivés de pyrrolo[2,3-d]pyrimidine et leur utilisation dans le traitement du cancer

Country Status (2)

Country Link
GB (1) GB201914910D0 (fr)
WO (1) WO2021074251A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022228509A1 (fr) * 2021-04-30 2022-11-03 微境生物医药科技(上海)有限公司 Dérivé de pyrrolopyrimidine, son procédé de préparation et son utilisation
WO2022228512A1 (fr) * 2021-04-30 2022-11-03 微境生物医药科技(上海)有限公司 Dérivé de pyrrolopyrimidine utilisé comme inhibiteur de wee -1
WO2023030388A1 (fr) * 2021-08-31 2023-03-09 微境生物医药科技(上海)有限公司 Composé de 5-fluoro-7h-pyrrolo[2,3-d]pyrimidines servant d'inhibiteur de wee-1

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009098236A1 (fr) * 2008-02-06 2009-08-13 Novartis Ag Composés hétérocycliques
WO2015038417A1 (fr) * 2013-09-10 2015-03-19 Asana Biosciences, Llc Composés permettant de réguler les voies fak et/ou src
WO2018098561A1 (fr) * 2016-12-01 2018-06-07 Aptose Biosciences Inc. Composés de pyrimidine fusionnés en tant qu'inhibiteurs doubles de brd4 et de jak2 et leurs procédés d'utilisation
CN110467615A (zh) * 2018-05-10 2019-11-19 四川科伦博泰生物医药股份有限公司 吡咯并嘧啶类化合物、包含其的药物组合物及其制备方法和用途
CN111718348A (zh) * 2019-03-22 2020-09-29 首药控股(北京)有限公司 Wee1抑制剂及其制备和用途

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009098236A1 (fr) * 2008-02-06 2009-08-13 Novartis Ag Composés hétérocycliques
WO2015038417A1 (fr) * 2013-09-10 2015-03-19 Asana Biosciences, Llc Composés permettant de réguler les voies fak et/ou src
WO2018098561A1 (fr) * 2016-12-01 2018-06-07 Aptose Biosciences Inc. Composés de pyrimidine fusionnés en tant qu'inhibiteurs doubles de brd4 et de jak2 et leurs procédés d'utilisation
CN110467615A (zh) * 2018-05-10 2019-11-19 四川科伦博泰生物医药股份有限公司 吡咯并嘧啶类化合物、包含其的药物组合物及其制备方法和用途
CN111718348A (zh) * 2019-03-22 2020-09-29 首药控股(北京)有限公司 Wee1抑制剂及其制备和用途

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
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] *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022228509A1 (fr) * 2021-04-30 2022-11-03 微境生物医药科技(上海)有限公司 Dérivé de pyrrolopyrimidine, son procédé de préparation et son utilisation
WO2022228512A1 (fr) * 2021-04-30 2022-11-03 微境生物医药科技(上海)有限公司 Dérivé de pyrrolopyrimidine utilisé comme inhibiteur de wee -1
WO2023030388A1 (fr) * 2021-08-31 2023-03-09 微境生物医药科技(上海)有限公司 Composé de 5-fluoro-7h-pyrrolo[2,3-d]pyrimidines servant d'inhibiteur de wee-1

Also Published As

Publication number Publication date
GB201914910D0 (en) 2019-11-27

Similar Documents

Publication Publication Date Title
US20230062022A1 (en) Heterocyclic compounds as kinase inhibitors
JP6789239B2 (ja) Krasの縮合三環系インヒビターおよびその使用の方法
JP6869947B2 (ja) 置換キナゾリン化合物ならびにそのg12c変異kras、hrasおよび/またはnrasタンパク質の阻害剤としての使用
EP2880035B9 (fr) Nouveaux composés de pyrrolopyrimidine utilisés en tant qu'inhibiteurs des protéines kinases
KR20200119824A (ko) 축합 고리 화합물
US20170224689A1 (en) Novel pyrrolopyrimidine compounds as inhibitors of protein kinases
CA3150689A1 (fr) Composes heterocycliques en tant qu'inhibiteurs de kinase
KR20180005178A (ko) 치환된 퀴나졸린 화합물 및 이의 사용방법
WO2021074251A1 (fr) Dérivés de pyrrolo[2,3-d]pyrimidine et leur utilisation dans le traitement du cancer
WO2015192658A1 (fr) Nouvel inhibiteur de la tyrosine kinase de bruton
KR20160076519A (ko) Kras g12c 억제제
CN114057771B (zh) 大环化合物及其制备方法和应用
EA034558B1 (ru) 4-ИМИДАЗО[1,5-a]ПИРИДАЗИН-1-ИЛ-БЕНЗАМИДЫ В КАЧЕСТВЕ Btk-ИНГИБИТОРОВ
JP2020502065A (ja) 8,9−ジヒドロイミダゾール[1,2−a]ピリミド[5,4−e]ピリミジン−5(6H)−ケトン類化合物
US9586965B2 (en) Pyrrolo[2,3-d]pyrimidine compounds as inhibitors of protein kinases
EP3993802A1 (fr) Composés hétérocycliques en tant qu'inhibiteurs de kinase
JP2022539208A (ja) チロシンキナーゼ非受容体1(tnk1)阻害剤およびその使用
WO2017162611A1 (fr) Composés quinoline-3-carboxamide et leur utilisation pour traiter le cancer
AU2020356348A1 (en) Pharmaceutical compounds
WO2016115869A1 (fr) Nouvel inhibiteur de la kinase flt3 et utilisation de ce dernier
KR20240019241A (ko) Setd2 억제제를 사용한 조합 요법
EP3896059B1 (fr) Inhibiteur de kinase pan-kit ayant une structure quinoléine et son utilisation
US20220281859A1 (en) Heterocyclic compounds as kinase inhibitors
CN111138426B (zh) 吲唑类激酶抑制剂及其用途
RU2789405C2 (ru) Ингибитор киназы pan-kit, имеющий структуру хинолина, и его применение

Legal Events

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

Ref document number: 20793310

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20793310

Country of ref document: EP

Kind code of ref document: A1