WO2015000959A1 - 4-amino-6-aryl[2,3-d]pyrimidines for the inhibition of egfr tyrosine kinase - Google Patents

4-amino-6-aryl[2,3-d]pyrimidines for the inhibition of egfr tyrosine kinase Download PDF

Info

Publication number
WO2015000959A1
WO2015000959A1 PCT/EP2014/064067 EP2014064067W WO2015000959A1 WO 2015000959 A1 WO2015000959 A1 WO 2015000959A1 EP 2014064067 W EP2014064067 W EP 2014064067W WO 2015000959 A1 WO2015000959 A1 WO 2015000959A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
oci
compound
nhco
hal
Prior art date
Application number
PCT/EP2014/064067
Other languages
French (fr)
Inventor
Bård HOFF
Svein KASPERSEN
Steffen Bugge
Eirik SUNDBY
Jin Han
Original Assignee
Norwegian University Of Science And Technology (Ntnu)
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
Priority claimed from GB201311943A external-priority patent/GB201311943D0/en
Priority claimed from GB201403809A external-priority patent/GB201403809D0/en
Application filed by Norwegian University Of Science And Technology (Ntnu) filed Critical Norwegian University Of Science And Technology (Ntnu)
Publication of WO2015000959A1 publication Critical patent/WO2015000959A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Definitions

  • Epidermal growth factor receptor tyrosine kinase is one of the more important targets in small molecular cancer therapy. These transmembrane enzyme-receptor complexes contain a binding site for external messenger molecules like epidermal growth factors (EGF), and an intracellular kinase domain. Binding of EGF induces dimerization and thereby activation of the kinase domain. Monoclonal antibodies such as Cetuximab and Panitumumab are efficient in blocking the entrance of EGF to EGFR and some monoclonal antibodies might, in addition, trigger immune responses.
  • EGF epidermal growth factor receptor tyrosine kinase
  • ATP competitive inhibitors on the marked includes Gefitinib, Erlotinib, Lapatinib and Vandetanib, which all are based on a central quinazoline core, see scheme 1. Both classes of inhibitors have the effect of down regulating downstream signalling events and thereby the growth and survival of the tumor.
  • pyrrolo-, thieno-, and furo[2,3- djpyrimidines as the central aromatic core with specific substitution patterns as inhibitors of EGFR-TK.
  • Pyrrolopyrimidines have been shown to affect a broad range of biological targets with applications as antiviral, anti-inflammatory, antifungal agents and so on.
  • Pyrrolopyrimidines have also been design to act as inhibitors of different kinases including JAK3 (Zhang et al. Bioorg.Med.Chem.Lett. (2007) 17, 1250-1253), Aurora kinase (Warner et al. Mol. Cancer Ther.
  • WO97/02266 examples include AEE-788 being a dual EGFR/VEGFR inhibitor (Traxler et al. Cancer Res (2004) 64,4931-4941), and Kawakita, et al.
  • Thienopyrimidines have also in general become an interesting structural element in development of pharmaceutical compounds and have, among others, been evaluated as cGMP phosphodiesterase inhibitors, anti-viral agents, but also as kinase inhibitors and potential anti-cancer agents.
  • Research at Glaxo SmitfiKline (Rheault et al. Bioorg.Med.Chem.Lett. (2009) 19, 817-820) on thienopyrimidine analogues of Lapatinib such as I (scheme 1), revealed low nanomolar potency towards EGFR, while Becker et al. Bioorg.Med.Chem. (2012) 20, 125-136 prepared compound II with low nM activity towards EGFR-TK.
  • irreversible thienopyrimidine based EGFR inhibitors have been developed in Wu et al.
  • Furopyrimidines have also been investigated as anti-cancer agents by kinase inhibition as discussed, inter alia, in WO2002/092603.
  • the inventors Based on flexible synthetic routes, the inventors have synthesised a series of new chiral pyrrolo[2,3-d]pyrimidines, thieno[2,3-d]pyrimidines, and some related furo[2,3-d]pyrimidine analogues, and evaluated their efficiency as EGFR-TK inhibitors. The inventors have therefore found a series of new and highly efficacious anti-cancer molecules. Whilst similar molecules are known in the art, the structure of the molecules of the invention is new and leads to improved activity.
  • the inventors also envisage that some compounds of the invention may have utility in the combined inhibition of EGFR and other kinases such as HER2, HER4, FGR, LYN A and LYN B.
  • compounds of the invention may have utility in combined inhibition of EGFR and other kinases such as ABL1, CSF1R, HER2, HER4, FGR, LYN A and LYN B, SRC and YES1.
  • the maximum potency of the compounds suggested by Bugge on EGFR-TK has been found to be 35 nM.
  • the invention provides a compound of formula (I) or (I')
  • X is S, O, or NH
  • each R is independently H or Ci_ 6 alkyl
  • each n is independently 1 to 3;
  • Re is H, Ci_6 alkyl, C 6 _io aryl, C7-12 arylalkyl, Hal, OR, SR * , N0 2 , NR 2 ;
  • R 7 is H, linear Ci_ 6 alkyl, (CH 2 ) n OR * , C0 2 R, CONR 2 , CH 2 NR 2 , CH 2 Hal, CH 2 SR, (CH 2 )nC0 2 R * or (CH 2 )nCONR * 2 ;
  • R 8 is H, Hal, Ci_ 6 alkyl, OCi_ 6 alkyl, OH, (CH 2 ) n OR, or CF 3 ;
  • R 9 is H, Hal, Ci_ 6 alkyl, OCi_ 6 alkyl, OH, (CH 2 ) n OR; or CF 3
  • Rio is H, Hal, Ci_ 6 alkyl, or (CH 2 ) n OR;
  • R11 is H, Hal, Ci_ 6 alkyl, OCi_ 6 alkyl, OH, (CH 2 ) n OR, or CF 3 ;
  • Ri 2 is H, Hal, Ci_ 6 alkyl, OCi_ 6 alkyl, OH, (CH 2 ) n OR, or CF 3;
  • X is S, O, or NH
  • Y is (CH 2 )nOR"
  • each R' is independently H or Ci_ 6 alkyl
  • R4 is H, OH, OCi_6 alkyl, Hal, CN, Ci_ 6 alkyl, SR * , CHO, NR * 2 , COR * , C0 2 R,
  • each n is independently 1 to 3;
  • Re is H, Ci_6 alkyl, C 6 _io aryl, C7-12 arylalkyl, Hal, OR, SR * , N0 2 , NR 2 ;
  • R 8 is H, Hal, Ci_ 6 alkyl, OCi_ 6 alkyl, OH, (CH 2 ) n OR, or CF 3 ;
  • R 9 is H, Hal, Ci_ 6 alkyl, OCi_ 6 alkyl, OH, (CH 2 ) n OR; or CF 3;
  • Rio is H, Hal, Ci_ 6 alkyl, or (CH 2 ) n OR;
  • R11 is H, Hal, Ci_ 6 alkyl, OCi_ 6 alkyl, OH, (CH 2 ) n OR, or CF 3 ;
  • Ri 2 is H, Hal, Ci_ 6 alkyl, OCi_ 6 alkyl, OH, (CH 2 ) n OR, or CF 3;
  • CH CH-CH 2 -NR' 2 , OCF 3 , OCF 2 H, CHF 2 , CH 2 F, (CH 2 ) n OR * , CH 2 NR 2 , SO 3 R or CH(OH)-R; and
  • R" is H or C i_6 alkyl, preferably Me or ethyl
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising compound as hereinbefore defined and at least
  • the invention provides a compound as hereinbefore defined for use in medicine.
  • the invention provides a compound as hereinbefore defined for use in the treatment of cancer.
  • the invention provides use of a compound as hereinbefore defined for the manufacture of a medicament for the treatment of cancer.
  • the invention provides a method of treating cancer in a patient comprising administering to a patient in need thereof, an effective compound as hereinbefore defined.
  • Ci_ 6 alkyl group is preferably a linear Ci_ 6 alkyl group. It is preferably a
  • Ci_3 alkyl group such as a linear Ci_ 3 alkyl group, especially methyl, ethyl or n- propyl.
  • Hal which stand for halide, is preferably CI or F, ideally F.
  • arylalkyl covers the combination of an aryl group and alkyl group. That may be bound to the atom in question either via the aryl portion or alkyl portion of the moiety, i.e. the term covers tolyl or benzyl for example. It is preferred if the arylalkyl unit links via the aryl portion of the moiety.
  • n is preferably 1 or 2, especially 1.
  • R' is preferably H or linear Ci_ 6 alkyl, such as Ci_3 alkyl, such as a linear Ci_ 3 alkyl group, preferably H or Me.
  • This invention relates to new pyrimidines as EGFR tyrosine kinase inhibitors.
  • the compounds preferably contain a substituted unit of formula:
  • the compounds of the invention are of formula (I) and comprise at least two non hydrogen substituents on phenyl ring 1. Note that one ortho position on phenyl ring 1 carries hydrogen by definition. Our molecules cannot therefore be substituted in the 2, 4 and 6 positions.
  • the compounds of the invention are of formula (II) and therefore carry a (CH 2 ) n OR" group at the equivalent of the R 7 position on the molecule.
  • substitution on the phenyl ring 1 e.g. only one non hydrogen substituent or even no substituent on phenyl ring 1. It is again preferred however, if there are two or more non hydrogen substituents on phenyl ring 1 in the compounds of formula (II).
  • X is S, O or NH, preferably S or NH, more preferably NH.
  • Ri is H, OH, -(CH 2 ) consultOH, CHO, CH 2 OCi_ 6 alkyl or
  • Ri is H, OH or OCi_ 6 alkyl, especially OMe. Ri can also be H, especially if there are non hydrogen substituents at positions R 2 , R 3 and/or R4.
  • Rj 3 is H, OH, -(CH 2 ) n OH, CHO, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl. More preferably Ri 3 is H, OH or OCi_ 6 alkyl, especially H or OMe, most especially H.
  • R 7 is H, linear Ci_ 6 alkyl, (CH 2 ) n OCi_ 2 alkyl or (CH 2 ) n OH, especially CH 2 OH, CH 3 , ethyl, C0 2 H, or CH 2 OMe. Most especially R 7 is H, Me or CH 2 OH, such as Me or CH 2 OH.
  • Rn is H, F, or CI, most preferably H.
  • phenyl ring 2 is unsubstituted in any compound of the invention. If it contains a non H substituent, it is preferred if that is positioned in the Rio position, then Rs.
  • stereochemistry of the molecule can be important. Whilst compounds of the invention could be used as a racemic mixture, it is preferred that the stereogenic centre of the amine part connected to C-4 of the pyrimidine is in the (R)- configuration in the Cahn Ingold Prelog system, when N is priority 1 , the Ph ring is priority 2 and the R 7 group is priority three. It is preferably in the (S)-configuration where the R 7 group is priority two and the Ph ring priority 3. In compounds of formula (II) the chiral carbon atom is ideally in the ( ⁇ -configuration as N is priority 1 , the CH 2 OH type group priority 2, and Ph priority 3.
  • Ri is H, OH, -(CH 2 ) complicatOH, CHO, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl;
  • R 2 is H, OH, -(CH 2 ) n OH, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl;
  • R 3 is H, OH, CHO, -(CH 2 ) n OH, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl;
  • R 4 is H, OH, CHO, -(CH 2 ) n OH, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl;
  • R 7 is H, linear Ci_ 6 alkyl, (CH 2 ) n OCi_ 2 alkyl, (CH 2 ) n OH; or C0 2 H;
  • Y is the group (CH 2 )nOR" which represents (CH 2 )nOH or (CH 2 )nOMe; n is 1 or 2;
  • Ri 3 is H or OMe
  • the invention relates to a compound of formula (V) or (V)
  • R 2 is H, OH, -(CH 2 ) n OH, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl;
  • R 4 is H, OH, CHO, -(CH 2 ) n OH, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl;
  • n 1 or 2;
  • Ri- R 4 are not H; rmula (VI) or (VF)
  • Ri is H, OH, -(CH 2 ) complicatOH, CHO, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl;
  • R 2 is H, OH, -(CH 2 ) n OH, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl;
  • R4 is H, OH, CHO, -(CH 2 ) n OH, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl; the group (CH 2 )nOR" represents (CH 2 )nOH or (CH 2 )nOMe, n is 1 or 2;
  • Ri 3 is H or OMe; and Rio position is F or H.
  • Ri is OH, -(CH 2 ) complicatOH, CHO, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl;
  • R 2 is H, OH, -(CH 2 ) n OH, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl;
  • R 3 is H, OH, CHO, -(CH 2 ) n OH, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl;
  • R4 is H, OH, CHO, -(CH 2 ) n OH, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl.
  • n 1 or 2;
  • Ri is H, OH, -(CH 2 ) complicatOH, CHO, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl;
  • R 2 is H, OH, -(CH 2 ) n OH, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl;
  • R 3 is H, OH, F, CHO, -(CH 2 ) n OH, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl;
  • n 1 or 2;
  • phenyl ring 1 is represented by the group:
  • Ri is OH, -(CH 2 ) n OH, CHO, CH 2 OCi_ 6 alkyl or OCi_ 6 alkyl;
  • the most preferred compounds of the invention offer EGFR IC50 values of 5 nM or less, such as 3 nM or less, especially 1 nM or less. Some compounds even possess inhibition values of 0.5 nM or less. As there are structural similarties in bio macro molecules and especially kinases binding ATP, having a high selectivity towards the target (EGFR) will reduce the chance of off-target toxicity. The most potent of the prepared compounds have a high selectivity for inhibiting EGFR in competition with other kinases. Kinase inhibition data are measured using a proprietary Invitrogen assay, details of which are given below.
  • the compounds of the invention can be synthesised using known chemistry in high yields and with few steps or side products.
  • the target thienopyrimidines were constructed as shown in scheme 2 below based on Bugge et al. Tetrahedron (2012) 68, 9226-9233. A benefit of this process is that a large number of compounds can be prepared cheaply and easily in high amounts.
  • R 7 CH 2 OH
  • R 10 H (40)
  • R 7 CH 2 OH
  • R 10 F (41 )
  • such molecules can be made by a different route as presented by Martin-Kohler, et al. Helv. Chim. Acta (2004) 87, 956-975.
  • the compounds of the invention target cancer, in particular breast, lung, and head and neck cancers, and other cancers where EGFR receptors are over expressed or mutated.
  • Other cancers of interest are hematologic and gastric cancers. It is also possible however that the compounds of the invention will have utility in the treatment of protozoan linked conditions, leprosy and malaria.
  • the compounds of the invention may also be used for the treatment of pain, especially neuropathic pain.
  • compositions pharmaceutically acceptable compositions.
  • pharmaceutically acceptable refers to molecular entities and other ingredients of such compositions that are
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia for use in mammals, and more particularly in humans.
  • carrier applied to pharmaceutical compositions of the invention refers to a diluent, excipient, or vehicle with which an active compound is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water, saline solutions, aqueous dextrose solutions, aqueous glycerol solutions, and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • Suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences” by E.W. Martin, 18th Edition, incorporated by reference.
  • Particularly preferred for the present invention are carriers suitable for immediate-release, i.e., release of most or all of the active ingredient over a short period of time, such as 60 minutes or less, and make rapid absorption of the drug possible.
  • the compounds of the invention can be administered in salt, solvate, prodrug or ester form, especially salt form.
  • a pharmaceutical acceptable salt may be readily prepared by using a desired acid.
  • the salt may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent.
  • an aqueous solution of an acid such as hydrochloric acid may be added to an aqueous suspension of a compound of formula (I) and the resulting mixture evaporated to dryness (lyophilised) to obtain the acid addition salt as a solid.
  • a compound of formula (I) may be dissolved in a suitable solvent, for example an alcohol such as isopropanol, and the acid may be added in the same solvent or another suitable solvent.
  • the resulting acid addition salt may then be precipitated directly, or by addition of a less polar solvent such as diisopropyl ether or hexane, and isolated by filtration.
  • Suitable addition salts are formed from inorganic or organic acids which form non-toxic salts and examples are hydrochloride, hydrobromide, hydroiodide, sulphate, bisulphate, nitrate, phosphate, hydrogen phosphate, acetate,
  • trifluoroacetate maleate, malate, fumarate, lactate, tartrate, citrate, formate, gluconate, succinate, pyruvate, oxalate, oxaloacetate, trifluoroacetate, saccharate, benzoate, alkyl or aryl sulphonates (eg methanesulphonate, ethanesulphonate, benzenesulphonate or p-toluenesulphonate) and isethionate.
  • Representative examples include trifluoroacetate and formate salts, for example the bis or tris trifluoroacetate salts and the mono or diformate salts, in particular the tris or bis trifluoroacetate salt and the mono formate salt.
  • solvates complexes with solvents in which they are reacted or from which they are precipitated or crystallized.
  • solvates For example, a complex with water is known as a "hydrate”.
  • Solvates of the compounds of the invention are within the scope of the invention.
  • the salts of the compound of Formula (I) may form solvates (e.g. hydrates) and the invention also includes all such solvates.
  • prodrug means a compound which is converted within the body, e.g. by hydrolysis in the blood, into its active form that has medical effects.
  • the compounds of the invention are proposed for use in the treatment of, inter alia, cancer.
  • treating or treatment is meant at least one of:
  • treatment is also used herein to cover prophylactic treatment, i.e. treating subjects who are at risk of developing a disease in question.
  • the compounds of the invention can be used on any animal subject, in particular a mammal and more particularly to a human or an animal serving as a model for a disease (e.g. mouse, monkey, etc.).
  • a mammal in particular a mammal and more particularly to a human or an animal serving as a model for a disease (e.g. mouse, monkey, etc.).
  • an “effective dose” means the amount of a compound that, when
  • the “effective dose” will vary depending on the compound, the disease and its severity and the age, weight, physical condition and
  • a compound of the invention may be administered as the bulk substance, it is preferable to present the active ingredient in a pharmaceutical formulation, for example, wherein the agent is in admixture with a pharmaceutically acceptable carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
  • carrier refers to a diluent, excipient, and/or vehicle with which an active compound is administered.
  • the pharmaceutical compositions of the invention may contain combinations of more than one carrier.
  • Such pharmaceutical carriers can be sterile liquids, such as water, saline solutions, aqueous dextrose solutions, aqueous glycerol solutions, and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water or aqueous solution saline solutions and aqueous dextrose and glycerol solutions are preferably employed as carriers, particularly for injectable solutions. Suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin, 18th Edition. The choice of pharmaceutical carrier can be selected with regard to the intended route of administration and standard
  • compositions may comprise as, in addition to, the carrier any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), and/or solubilizing agent(s).
  • compositions for use in accordance with the present invention may be in the form of oral, parenteral, transdermal, inhalation, sublingual, topical, implant, nasal, or enterally administered (or other mucosally administered) suspensions, capsules or tablets, which may be formulated in conventional manner using one or more pharmaceutically acceptable carriers or excipients.
  • composition/formulation requirements may be different depending on the different delivery systems.
  • the composition comprises more than one active component, then those components may be administered by the same or different routes.
  • the pharmaceutical formulations of the present invention can be liquids that are suitable for oral, mucosal and/or parenteral administration, for example, drops, syrups, solutions, injectable solutions that are ready for use or are prepared by the dilution of a freeze-dried product but are preferably solid or semisolid as tablets, capsules, granules, powders, pellets, pessaries, suppositories, creams, salves, gels, ointments; or solutions, suspensions, emulsions, or other forms suitable for administration by the transdermal route or by inhalation.
  • the compounds of the invention can be administered for immediate-, delayed-, modified-, sustained-, pulsed-or controlled-release applications.
  • oral compositions are slow, delayed or positioned release (e.g., enteric especially colonic release) tablets or capsules.
  • This release profile can be achieved without limitation by use of a coating resistant to conditions within the stomach but releasing the contents in the colon or other portion of the GI tract wherein a site has been identified or a delayed release can be achieved by a coating that is simply slow to disintegrate or the two (delayed and positioned release) profiles can be combined in a single formulation by choice of one or more appropriate coatings and other excipients.
  • Such formulations constitute a further feature of the present invention.
  • compositions can be prepared by mixing a therapeutically effective amount of the active substance with a pharmaceutically acceptable carrier that can have different forms, depending on the way of administration.
  • composition components include one or more of binders, fillers, lubricants, odorants, dyes, sweeteners, surfactants, preservatives, stabilizers and antioxidants.
  • compositions of the invention may contain from 0.01 to
  • the therapeutic doses will generally be between about 10 and 2000 mg/day and preferably between about 30 and 1500 mg/day. Other ranges may be used, including, for example, 50-500 mg/day, 50-300 mg/day, 100-200 mg/day.
  • Administration may be once a day, twice a day, or more often, and may be decreased during a maintenance phase of the disease or disorder, e.g. once every second or third day instead of every day or twice a day.
  • the dose and the administration frequency will depend on the clinical signs, which confirm maintenance of the remission phase, with the reduction or absence of at least one or more preferably more than one clinical signs of the acute phase known to the person skilled in the art.
  • a compound as described herein to be administered in combination with another pharmaceutical, e.g. another drug with known efficacy against the disease in question.
  • another pharmaceutical e.g. another drug with known efficacy against the disease in question.
  • the compounds of the invention may therefore be used in combination therapy.
  • the compounds of the invention may be used in combination with other kinase inhibitors with other targets, and other chemotherapeutic agents (cisplatin, taxol etc) used to treat cancerous diseases. .
  • ERBB2 (HER2) 50 57 PDGFRA (PDGFR alpha) ⁇ 1 ⁇ 1
  • ERBB4 (HER4) 63 30 PDGFRB (PDGFR beta) ⁇ 1 3
  • GSK3B (GSK3 beta) 8 7 RET 9 5
  • IKK beta 1 10 STK22B (TSSK2) 8 8
  • GSK3B (GSK3 beta) Km app -8 -1 -13 8 6 5 -4 -8
  • MAPK14 (p38 alpha) Km app 13 3 6 19 24 24 30 25
  • PDGFRA (PDGFR alpha) Km app 5 29 51 6 6 8 -3 -4
  • PRKCB 1 (PKC beta I) Km app -2 16 10 1 8 10 8 8
  • pyrrolopyrimidine based EGFR-TK inhibitors were evaluated in genetically modified model cells Ba/F3-EGFR L858R cells, and four human cancer cell lines including A-431 (epidermoid carcinoma), AU-565 (breast adenocarcinoma), K-562 (leukemia) and C-33A (cervix carcinoma). The results are shown in Table 7. Table 7. Summary of cell proliferation data (IC 50 ) of pyrrolopyrimidines towards
  • Ba/F3-EGFR L858R the cancer cell lines A-431, AU-565, C-33A, K-562 and the normal lung fibroblast cell line MRC-5.
  • Methyl 2-aminothiophene-3-carboxylate (1) (26.0 g, 165 mmol) was mixed with formamide (285 mL) and heated at 215 °C for 5 h. The reaction mixture was then cooled to rt. and water (500 mL) was added, followed by extraction with EtOAc (4 x 500 mL). The combined organic fractions were dried over Na 2 S0 4 , and
  • Ethyl 3-amino-3-iminopropanoate hydrochloride (23) (8.3 g, 50.0 mmol) and NaOEt (5.1 g, 75.0 mmol) were dissolved in abs. EtOH at 0 °C and stirred for 20 min. under argon. The mixture was heated to 60 °C, and 2-bromo-l-phenylethanone (24) (5.0 g, 25.0 mmol) was added portion wise over 5 min. After 1.5 h the mixture was cooled to 20 °C and the solvent was evaporated under reduced pressure. The residue was diluted with distilled water (20 mL) and extracted with EtOAc (3 X80 mL).
  • 6-Phenyl-7H-pyrrolo[2,3-d]pyrimidin-4-ol (26) (1.57 g, 7.43 mmol) and neat POCI 3 (13.5 mL) were mixed and reacted at 90 °C for 3 h.
  • the solution was cooled with an ice-salt bath, and water (60 mL) was added.
  • NaOH (8 M, 80 mL) was used to adjust the pH to 12.
  • the formed precipitate was isolated by filtration, washed with water and n-pentane and dried to give 1.49 g (6.49 mmol, 87%>) of a yellowish solid, mp. 255-257 °C.
  • the activity measurements of compounds towards ErbB 1 was performed by Invitrogen using their Z'-LYTE ® assay technology. 38 The test compounds were diluted in 1% DMSO solution, and subjected to a duplicate 10 points titration, using an ATP concentration equal to K m . The IC 50 values were calculated from activity data with a four parameter logistic model using SigmaPlot (Windows Version 11.0 from Systat Software, Inc.)
  • Transfected Ba/F3 cells containing expression vectors for the L858R and T790M EGFR mutants were a kind gift from Dr. Nikolas von Bubnoff at the Technical University of Kunststoff, Kunststoff, Germany.
  • the cells were cultured in RPMI 1640 (Gibco, Invitrogen) supplemented with 10% FCS (Gibco, Invitrogen), 1% L- glutamine (Gibco, Invitrogen) and 0.1% Gentamycin (Sanofi Aventis).
  • Erlotinib was purchased from LC Laboratories (Woburn, MA). All inhibitors were reconstituted in DMSO, and appropriate stock solutions were prepared using cell culture medium. The final percentage concentrations of DMSO were ⁇ 0.2%.
  • Cultures were maintained at 37 °C in a humidified atmosphere of 5% C0 2 and 95% air.
  • Compound, Erlotinib and Staurosporine (positive control) were all dissolved in DMSO in 10 mM stock.
  • Culture medium (10 ⁇ ) was added to each well of 384 well cell culture plates.
  • the compounds were diluted in a source plate in DMSO at 3 fold serial dilutions starting at 10 mM, total 10 doses.
  • the compounds (0.25 ⁇ ) were delivered from source plate to each well of the cell culture plates by Echo 550. Then, 15 ⁇ of culture medium containing 5000 cells of A-431 or K-562 were added to the wells of the cell culture plates.
  • the cells were incubated with the compounds at 37 °C, 5% C0 2 for 72 hours.
  • 25 ⁇ of Cell Titer Glo reagent (25 ⁇ ) was added to each well according to the instruction of the kit.
  • the contents were mixed on an orbital shaker for 2 minutes and incubated at room temperature for 10 minutes to stabilize luminescent signal.
  • Luminescence was recorded by Envision 2104 Multilabel Reader (PerkinElmer, Santa Clara, CA). The maximum luminescence for each cell line in the absence of test compound, but in the presence of 0.4% DMSO, was similarly recorded after incubation for 72 hours.
  • the number of viable cells in the culture was determined based on quantitation of the ATP present in each culture well.
  • NTRC Netherlands Translational Research Center B.V
  • All cell lines were licensed from the American Type Culture Collection (ATCC) Manassas, Virginia (US).
  • Master and working cell banks (MCB and WCB) were prepared by subculturing in ATCC-recommended media and freezing according to ATCC recommended protocols (www.atcc.org).
  • Cell line stocks for the assays were prepared from the WCB.
  • the MCB, WCBs and assay stocks were prepared within respectively 3, 6 and 9 passages of the ATCC vial.
  • Proliferation analysis Compounds were weighed on a calibrated balance and dissolved in 100% DMSO to a concentration of 10 mM. The samples were stored at room temperature.
  • the compound stock was diluted in 3.16 fold steps in 100% DMSO to obtain a 10-point dilution series. This was further diluted 31.6 times in 20 mM sterile Hepes buffer pH 7.4. A volume of 5 ml was transferred to the cells to generate a test concentration range from 3.16 x 10 "5 M to 3.16 ⁇ 10 "9 M in duplicate. The final DMSO concentration during incubation was 0.4% in all wells.
  • An assay stock was thawed and diluted in its ATCC recommended medium and dispensed in a 384-well plate, depending on the cell line used, at a concentration of 400 - 1600 cells per well in 45 ⁇ medium. For each used cell line the optimal cell density was used.
  • IC 50 values were calculated from % growth and compound concentration with a four parameter logistic model using SigmaPlot (Windows Version 12.0 from Systat Software, Inc.)
  • the compounds and the reference staurosporine (Sigma- Aldrich , Saint Louis, MI) were all dissolved in DMSO in 10 mM stock.
  • Cell Titer-Glo® Luminescent cell viability assay reagent was obtained from Promega (Madison, WI).
  • MRC-5 human normal lung fibroblast cell line was obtained from American Type Culture
  • MRC-5 cells were grown in DMEM medium

Abstract

This invention relates to certain new pyrrolo-, thieno-, and furo-[2,3- d]pyrimidine compounds, such as of general formula (I) These compounds are epidermal growth factor receptor tyrosine kinase inhibitors and therefore offer potential in the treatment of cancer.

Description

4-AMINO-6-ARYL[2,3-D]PYRIMIDINES FOR THE INHIBITION OF EGFR TYROSINE KINASE
This invention relates to certain new pyrrolo-, thieno-, and furo-[2,3- djpyrimidine compounds and in particular, a series of such new 4-amino-6-aryl [2,3- djpyrimidines. These compounds are epidermal growth factor receptor tyrosine kinase inhibitors and therefore offer potential in the treatment of cancer.
Their in vitro activity was found to depend strongly on the substitution pattern in the 6-aryl ring, the stereochemistry and the structure of the amine at C-4 of the pyrimidine. Optimization led to the discovery of several potent agents (IC50 < 6.5 nM, such as <5 nM), whereas the most potent, obtained by combination of "active" fragments, had EGFR IC50 of 0.3 nM or even 0.2 nM.
Background
Epidermal growth factor receptor tyrosine kinase (EGFR-TK) is one of the more important targets in small molecular cancer therapy. These transmembrane enzyme-receptor complexes contain a binding site for external messenger molecules like epidermal growth factors (EGF), and an intracellular kinase domain. Binding of EGF induces dimerization and thereby activation of the kinase domain. Monoclonal antibodies such as Cetuximab and Panitumumab are efficient in blocking the entrance of EGF to EGFR and some monoclonal antibodies might, in addition, trigger immune responses. Properly designed low molecular weight compounds are, on the other hand, attractive as inhibitors as, in contrast to monoclonal antibodies, they can act intracellularly. These compounds also have more moderate production costs. ATP competitive inhibitors on the marked includes Gefitinib, Erlotinib, Lapatinib and Vandetanib, which all are based on a central quinazoline core, see scheme 1. Both classes of inhibitors have the effect of down regulating downstream signalling events and thereby the growth and survival of the tumor.
Figure imgf000003_0001
Scheme 1. Structure of some potent EGFR inhibitors.
One problem with these medicines, e.g. Gefitinib and Erlotinib is that eventually patients who initially respond to treatment develop resistance to these drugs. Also, these established agents can have serious side effects.
There are strong reasons therefore for further drugs to be found in this field.
The present inventors now propose the use of pyrrolo-, thieno-, and furo[2,3- djpyrimidines as the central aromatic core with specific substitution patterns as inhibitors of EGFR-TK. Pyrrolopyrimidines have been shown to affect a broad range of biological targets with applications as antiviral, anti-inflammatory, antifungal agents and so on. Pyrrolopyrimidines have also been design to act as inhibitors of different kinases including JAK3 (Zhang et al. Bioorg.Med.Chem.Lett. (2007) 17, 1250-1253), Aurora kinase (Warner et al. Mol. Cancer Ther. (2006) 5, 1764-1773), Mpsl(TTK) kinase (Bursavich et al. Bioorg.Med.Chem.Lett. (2013) 23, 6829-6833), but also as EGFR-TK inhibitors as described, inter alia, in
WO97/02266. Examples include AEE-788 being a dual EGFR/VEGFR inhibitor (Traxler et al. Cancer Res (2004) 64,4931-4941), and Kawakita, et al.
Bioorg.Med.Chem. (2012) 20 6171-6180, 2012.
Thienopyrimidines have also in general become an interesting structural element in development of pharmaceutical compounds and have, among others, been evaluated as cGMP phosphodiesterase inhibitors, anti-viral agents, but also as kinase inhibitors and potential anti-cancer agents. Research at Glaxo SmitfiKline (Rheault et al. Bioorg.Med.Chem.Lett. (2009) 19, 817-820) on thienopyrimidine analogues of Lapatinib such as I (scheme 1), revealed low nanomolar potency towards EGFR, while Becker et al. Bioorg.Med.Chem. (2012) 20, 125-136 prepared compound II with low nM activity towards EGFR-TK. In addition, irreversible thienopyrimidine based EGFR inhibitors have been developed in Wu et al.
J.Med.Chem. (2010) 53, 7316-7326.
Furopyrimidines have also been investigated as anti-cancer agents by kinase inhibition as discussed, inter alia, in WO2002/092603.
Based on flexible synthetic routes, the inventors have synthesised a series of new chiral pyrrolo[2,3-d]pyrimidines, thieno[2,3-d]pyrimidines, and some related furo[2,3-d]pyrimidine analogues, and evaluated their efficiency as EGFR-TK inhibitors. The inventors have therefore found a series of new and highly efficacious anti-cancer molecules. Whilst similar molecules are known in the art, the structure of the molecules of the invention is new and leads to improved activity.
The inventors also envisage that some compounds of the invention may have utility in the combined inhibition of EGFR and other kinases such as HER2, HER4, FGR, LYN A and LYN B. The inventors have also envisage that compounds of the invention may have utility in combined inhibition of EGFR and other kinases such as ABL1, CSF1R, HER2, HER4, FGR, LYN A and LYN B, SRC and YES1.
In Bugge et al. Tetrahedron (2012) 68, 9226-9233, the synthesis of certain 6- aryl-N-(l-arylethyl)thienopyrimidine-4-amines is discussed using Suzuki chemistry and suggest their use in cancer therapy. The target compounds in Bugge are of formul
Figure imgf000004_0001
where Ar is Ph or
Figure imgf000005_0001
The maximum potency of the compounds suggested by Bugge on EGFR-TK has been found to be 35 nM.
The present inventors have now found however, that certain aromatic substituent patterns and/or the presence of a CH2OH type group in benzylic position of the chiral amine bound to the pyrimidine at C-4 improves inhibition of EGFR-TK and therefore offer better performance in terms of EGFR inhibition and thus cancer treatment.
Moreover, the synthesis of these new compounds remains simple.
These compounds may therefore offer an alternative to the likes of Erlotinib and Gefitinib or may offer a combination therapy with these or other chemical or protein based agents. Summary of Invention
Thus viewed from one aspect the invention provides a compound of formula (I) or (I')
Figure imgf000006_0001
wherein
X is S, O, or NH;
Ri is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR*, CHO, NR2, COR*, C02R*, CONR*2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-CH=CH- CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR, CH2NR* 2, SO3R or CH(OH)-
R*;
each R is independently H or Ci_6 alkyl;
R2 is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR*, CHO, NR* 2, COR*, C02R, CONR*2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-CH=CH- CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR*, CH2NR* 2, SO3R or CH(OH)-
R*;
R3 is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR*, CHO, NR* 2, COR*, C02R, CONR2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-CH=CH- CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR, CH2NR* 2, SO3R or CH(OH)-
R;
R4 is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR*, CHO, NR* 2, COR*, C02R, CONR2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-CH=CH- CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR, CH2NR* 2, SO3R or CH(OH)- R;
each n is independently 1 to 3;
R5 is H, Hal, Ci_6-alkyl, N02, NH2, NHCO-CH=CH-CH2-NR'2, or NHCO- CH=CH-R';
Re is H, Ci_6 alkyl, C6_io aryl, C7-12 arylalkyl, Hal, OR, SR*, N02, NR2;
NHCO-CH=CH-CH2-NR'2, NHCO-CH=CH-R' , or C02R' ;
R7 is H, linear Ci_6alkyl, (CH2)nOR*, C02R, CONR2, CH2NR2, CH2Hal, CH2SR, (CH2)nC02R* or (CH2)nCONR* 2;
R8 is H, Hal, Ci_6 alkyl, OCi_6 alkyl, OH, (CH2)nOR, or CF3;
R9 is H, Hal, Ci_6 alkyl, OCi_6 alkyl, OH, (CH2)nOR; or CF3
Rio is H, Hal, Ci_6 alkyl, or (CH2)nOR;
R11 is H, Hal, Ci_6 alkyl, OCi_6 alkyl, OH, (CH2)nOR, or CF3;
Ri2 is H, Hal, Ci_6 alkyl, OCi_6 alkyl, OH, (CH2)nOR, or CF3;
Ri3 is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR, CHO, NR* 2, COR*, C02R, CONR2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO- CH=CH-CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR*, CH2NR2, SO3R or CH(OH)-R;
or a salt, ester, solvate, N-oxide, or prodrug thereof;
with the proviso that at least two of Ri- R4 and R13 are not H.
Viewed from another aspect the invention provides a compound of formula (II)
Figure imgf000008_0001
preferably of formula (ΙΓ)
Figure imgf000008_0002
wherein
X is S, O, or NH;
Y is (CH2)nOR";
Ri is H, OH, OCi-6 alkyl, Hal, CN, Ci_6 alkyl, SR*, CHO, NR* 2, COR*, C02R, CONR2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-CH=CH- CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR', CH2NR* 2, S03R or CH(OH)-
R; each R' is independently H or Ci_6 alkyl;
R2 is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR*, CHO, NR* 2, COR*, C02R, CONR2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-CH=CH- CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR, CH2NR* 2, SO3R or CH(OH)- R;
R3 is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR*, CHO, NR* 2, COR*, C02R, CONR2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-CH=CH- CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR, CH2NR* 2, SO3R or CH(OH)-
R*;
R4 is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR*, CHO, NR* 2, COR*, C02R,
CONR2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-CH=CH- CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR, CH2NR* 2, SO3R or CH(OH)-
R;
each n is independently 1 to 3;
R5 is H, Hal, Ci_6-alkyl, N02, NH2, NHCO-CH=CH-CH2-NR'2, or NHCO-
CH=CH-R';
Re is H, Ci_6 alkyl, C6_io aryl, C7-12 arylalkyl, Hal, OR, SR*, N02, NR2;
NHCO-CH=CH-CH2-NR'2 NHCO-CH=CH-R', or C02R';
R8 is H, Hal, Ci_6 alkyl, OCi_6 alkyl, OH, (CH2)nOR, or CF3;
R9 is H, Hal, Ci_6 alkyl, OCi_6 alkyl, OH, (CH2)nOR; or CF3;
Rio is H, Hal, Ci_6 alkyl, or (CH2)nOR;
R11 is H, Hal, Ci_6 alkyl, OCi_6 alkyl, OH, (CH2)nOR, or CF3;
Ri2 is H, Hal, Ci_6 alkyl, OCi_6 alkyl, OH, (CH2)nOR, or CF3;
Ri3 is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR, CHO, NR* 2, COR*, C02R, CONR2, NR'CONR* 2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-
CH=CH-CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR*, CH2NR2, SO3R or CH(OH)-R; and
R" is H or C i_6 alkyl, preferably Me or ethyl;
or a salt thereof or a salt, ester, solvate, N-oxide, or prodrug thereof.
Viewed from another aspect the invention provides a pharmaceutical composition comprising compound as hereinbefore defined and at least
pharmaceutically acceptable carrier. Viewed from another aspect the invention provides a compound as hereinbefore defined for use in medicine.
Viewed from another aspect the invention provides a compound as hereinbefore defined for use in the treatment of cancer.
Viewed from another aspect the invention provides use of a compound as hereinbefore defined for the manufacture of a medicament for the treatment of cancer.
Viewed from another aspect the invention provides a method of treating cancer in a patient comprising administering to a patient in need thereof, an effective compound as hereinbefore defined.
Definitions
The following definitions apply to all claimed compounds.
Any Ci_6 alkyl group is preferably a linear Ci_6 alkyl group. It is preferably a
Ci_3 alkyl group, such as a linear Ci_3alkyl group, especially methyl, ethyl or n- propyl.
Any Hal, which stand for halide, is preferably CI or F, ideally F.
The term arylalkyl covers the combination of an aryl group and alkyl group. That may be bound to the atom in question either via the aryl portion or alkyl portion of the moiety, i.e. the term covers tolyl or benzyl for example. It is preferred if the arylalkyl unit links via the aryl portion of the moiety.
In any compound of the invention, n is preferably 1 or 2, especially 1.
In any compound of the invention R' is preferably H or linear Ci_6 alkyl, such as Ci_3 alkyl, such as a linear Ci_3alkyl group, preferably H or Me.
Detailed Description of Invention
This invention relates to new pyrimidines as EGFR tyrosine kinase inhibitors. The compounds preferably contain a substituted unit of formula:
Figure imgf000011_0001
In the discussion which follows, phenyl rings 1 and 2 are as shown above.
In a first embodiment, the compounds of the invention are of formula (I) and comprise at least two non hydrogen substituents on phenyl ring 1. Note that one ortho position on phenyl ring 1 carries hydrogen by definition. Our molecules cannot therefore be substituted in the 2, 4 and 6 positions.
In a second embodiment, the compounds of the invention are of formula (II) and therefore carry a (CH2)nOR" group at the equivalent of the R7 position on the molecule. In these compounds, there can be a wider variation of substitution on the phenyl ring 1 , e.g. only one non hydrogen substituent or even no substituent on phenyl ring 1. It is again preferred however, if there are two or more non hydrogen substituents on phenyl ring 1 in the compounds of formula (II).
In all compounds of the invention it is preferred if X is S, O or NH, preferably S or NH, more preferably NH.
In all compounds of the invention, it is preferred if there is at least one non hydrogen substituent on the phenyl ring 1. In compounds of formula (II), if there is only one substituent present, the location of that substituent is preferably in position R3, next preferred Ri, then R2. Alternatively, Ri, next preferred R3, then R2.
If two non hydrogen substituents are present in any compound of the invention, it is preferred if those are located ortho and para to the bond to the pyrimidine, i.e. the Ri and R3 positions on phenyl ring 1. Alternatively, substituents may be in the Ri and R2 positions or Ri and R4 positions. Another preferred option is in positions Ri and Ri3. It is also possible for phenyl ring 1 to contain 3 or 4 non hydrogen substituents. In a further preferred embodiment, phenyl ring 1 of the compounds of the invention carries at least one substituent comprising an oxygen atom. Preferably, phenyl ring 1 carries two substituents comprising an oxygen atom. Preferably all non H substituents contain an O atom on phenyl ring 1.
Thus, it is preferred if Ri is H, OH, -(CH2)„OH, CHO, CH2OCi_6 alkyl or
OCi_6 alkyl. More preferably Ri is H, OH or OCi_6 alkyl, especially OMe. Ri can also be H, especially if there are non hydrogen substituents at positions R2, R3 and/or R4.
R2 is preferably H, OH, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl, in particular R2 is -(CH2)nOH if not H. It is generally preferred if R2 is H.
It is preferred if R3 is H, OH, F, CHO, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl. It is preferred if R3 is H, F, OH, CHO, OCi_6 alkyl or CH2OH. R3 is also most preferably -CH2OH. R3 is also preferably H, especially if there are non H substituents at positions Ri .
It is preferred if R4 is H, OH, CHO, Hal -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl. It is most preferred however, if R4 is H, CH2OH, or CHO, especially H.
It is preferred if Rj3 is H, OH, -(CH2)nOH, CHO, CH2OCi_6 alkyl or OCi_6 alkyl. More preferably Ri3 is H, OH or OCi_6 alkyl, especially H or OMe, most especially H.
In a further preferred embodiment in compounds of formula (II), R1-R4 and
Ri3 are all H.
It will be appreciated that in compounds of formula (I) at least two of R1-R4 and Ri3 must not be H. Ideally two of R1-R4 are not H. In these compounds, the presence of two substituents improves activity.
It is preferred if R5 is H.
It is preferred if 5 is H.
It is preferred if R7 is H, linear Ci_6 alkyl, (CH2)nOCi_2 alkyl or (CH2)nOH, especially CH2OH, CH3, ethyl, C02H, or CH2OMe. Most especially R7 is H, Me or CH2OH, such as Me or CH2OH.
In formula (II), the group Y is preferably (CH2)nOH or (CH2)nOMe, i.e. R" is H or Me. In this group n is preferably 1 , 2 or 3 such as 1. Thus the unit Y is most preferably (CH2)OH or (CH2)OMe, most preferably (CH2)OH. It is preferred if Rs is H, F, or CI, most preferably H.
It is preferred if Rg is H, F, or CI, most preferably H.
It is preferred if Rn is H, F, or CI, most preferably H.
It is preferred if R12 is H, F, or CI, most preferably H.
It is preferred if the Rio position is CI, F or H, ideally H.
In a most preferred embodiment therefore, phenyl ring 2 is unsubstituted in any compound of the invention. If it contains a non H substituent, it is preferred if that is positioned in the Rio position, then Rs.
The stereochemistry of the molecule can be important. Whilst compounds of the invention could be used as a racemic mixture, it is preferred that the stereogenic centre of the amine part connected to C-4 of the pyrimidine is in the (R)- configuration in the Cahn Ingold Prelog system, when N is priority 1 , the Ph ring is priority 2 and the R7 group is priority three. It is preferably in the (S)-configuration where the R7 group is priority two and the Ph ring priority 3. In compounds of formula (II) the chiral carbon atom is ideally in the (^-configuration as N is priority 1 , the CH2OH type group priority 2, and Ph priority 3.
It is thus preferred if the compounds of the invention are of formula (III)
especially of formula
R"0(H2C)n
or especially of formula (III")
Figure imgf000014_0001
wherein Ri-Rio, R13, X, Y, n, R' and R" are as hereinbefore defined; formula (IV)

Figure imgf000015_0001
wherein Ri-R^ X and n are as hereinbefore defined.
Ideally, therefore in compounds of formula (III), (III') or (IV)/(IV);
X is S or NH;
Ri is H, OH, -(CH2)„OH, CHO, CH2OCi_6 alkyl or OCi_6 alkyl;
R2 is H, OH, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl;
R3 is H, OH, CHO, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl;
R4 is H, OH, CHO, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl;
R7 is H, linear Ci_6 alkyl, (CH2)nOCi_2 alkyl, (CH2)nOH; or C02H;
Y is the group (CH2)nOR" which represents (CH2)nOH or (CH2)nOMe; n is 1 or 2;
R8 is H, F or CI;
R9 is H, F or CI;
Rio position is F, CI or H;
Ri3 is H or OMe;
with the proviso that in compounds of formula (IV) , (IV) or (IV") at least two of Ri- R4 and Ri3 are not H.
In a further preferred embodiment therefore, the invention relates to a compound of formula (V) or (V)
Figure imgf000016_0001
(V) (V)
especially
Figure imgf000017_0001
(Va) or (Vb) wherein Ri-Rio, R13 and n are as hereinbefore defined, preferably
Ri is H, OH, -(CH2)„OH, CHO, CH2OCi_6 alkyl or OCi_6 alkyl;
R2 is H, OH, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6alkyl;
R3 is H, OH, CHO, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl;
R4 is H, OH, CHO, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl;
R7 is H, linear Ci_6 alkyl, (CH2)nOCi_2 alkyl or (CH2)nOH;
n is 1 or 2;
Rio position is F or H;
Figure imgf000017_0002
with the proviso that at least two of Ri- R4 are not H; rmula (VI) or (VF)
Figure imgf000018_0001
(VI) (VF) especially of formula
Figure imgf000018_0002
(Via) or (VIb) wherein R1-R 3, n and R" are as hereinbefore defined; preferably
Ri is H, OH, -(CH2)„OH, CHO, CH2OCi_6 alkyl or OCi_6 alkyl; R2 is H, OH, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl;
R3 is H, OH, CHO, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl;
R4 is H, OH, CHO, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl; the group (CH2)nOR" represents (CH2)nOH or (CH2)nOMe, n is 1 or 2;
Ri3 is H or OMe; and Rio position is F or H.
More preferred are compounds of formula (VII)
Figure imgf000019_0001
(VII) (VIII)
especially of formula (VIF) or (VIIF)
Figure imgf000019_0002
(VIF) (VIIF) wherein R -R4 and n are as hereinbefore defined, preferably
Ri is OH, -(CH2)„OH, CHO, CH2OCi_6 alkyl or OCi_6 alkyl;
R2 is H, OH, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl;
R3 is H, OH, CHO, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl; and
R4 is H, OH, CHO, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl.
n is 1 or 2;
with the proviso that in compounds of formula (VII) or (VIF) at least two of Ri- R4 are not H.
In particular, the compounds (IX) and (X) are
interesting:
Figure imgf000020_0001
(IX) (X) especially
Figure imgf000021_0001
(ΙΧ') (Χ') wherein R -R4 and n are as hereinbefore defined, preferably
Ri is H, OH, -(CH2)„OH, CHO, CH2OCi_6 alkyl or OCi_6 alkyl;
R2 is H, OH, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl;
R3 is H, OH, F, CHO, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl; and
R4 is H, OH, CHO, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl;
n is 1 or 2;
with the proviso that in compounds of formula (IX) and (IX') at least two of Ri- R4 are not H.
In any compound above, it is further preferred if phenyl ring 1 is represented by the group:
Figure imgf000021_0002
where Ri and R3 are as hereinbefore defined, preferably Ri is H, OH, -(CH2)nOH, CHO, CH2OCi_6 alkyl or OCi_6 alkyl; and R3 is H, OH, F, CHO, -(CH2)„OH, CH2OCi_6 alkyl or OCi_6 alkyl; preferably where
Ri is OH, -(CH2)nOH, CHO, CH2OCi_6 alkyl or OCi_6 alkyl; and
R3 is OH, F, CHO, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl; and n is 1 or 2.
A further preferred embodiment, therefore relates to a compound of formula (XI) or (XII)
Figure imgf000022_0001
(XI)
Figure imgf000022_0002
Ri is H, OH, -(CH2)„OH, CHO, CH2OCi_6 alkyl or OCi_6 alkyl; and
R3 is H, OH, F, CHO, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl n is 1 or 2;
where for compound (XII) and (ΧΙΓ) Ri and R3 are not H.
In a highly preferred embodiment, the compounds mentioned in the examples are preferred especially the compounds:
Figure imgf000023_0001
It will be appreciated that any compound of the invention such as those of formula (III) to (ΧΙΓ) and the four compounds above can be administered as a salt, ester, solvate, N-oxide, or prodrug thereof, in particular a salt thereof.
Lack of water solubility is a problem for a pharmaceutical and whilst our compounds are not water soluble, they are more polar than competing products which may enhance bioavailability.
The most preferred compounds of the invention offer EGFR IC50 values of 5 nM or less, such as 3 nM or less, especially 1 nM or less. Some compounds even possess inhibition values of 0.5 nM or less. As there are structural similarties in bio macro molecules and especially kinases binding ATP, having a high selectivity towards the target (EGFR) will reduce the chance of off-target toxicity. The most potent of the prepared compounds have a high selectivity for inhibiting EGFR in competition with other kinases. Kinase inhibition data are measured using a proprietary Invitrogen assay, details of which are given below.
Synthesis
The compounds of the invention can be synthesised using known chemistry in high yields and with few steps or side products. The target thienopyrimidines were constructed as shown in scheme 2 below based on Bugge et al. Tetrahedron (2012) 68, 9226-9233. A benefit of this process is that a large number of compounds can be prepared cheaply and easily in high amounts.
Formamide/
Figure imgf000025_0001
Figure imgf000025_0002
R7=H (5), Me (6), Et (7),
CH2OH (8), CH2OMe (9)
Figure imgf000025_0003
R7=H (10), Me (11 ), Et (12), R7=H (16), Me (17), Et (18), CH2OH (13), CH2OMe (14) CH2OH (19), CH2OMe (20)
Figure imgf000025_0004
Scheme 2. Synthesis of thienopyrimidines. The synthesis of the pyrrolopyrimidines were performed essentially by two different routes, the first route adopted from Kaspersen et al. Eur. J. Med Chem. (2011) 46, 6002-6014 and references therein, see scheme 3.
Figure imgf000026_0001
* Letter defines the 6-aryl group as shown in Scheme 2
Scheme 3. Route for preparation of the target pyrrolopyrimidines.
The second route of synthesis is shown in Scheme 4:
Figure imgf000027_0001
Figure imgf000027_0002
38-41
R7 = H R10 = H (38)
R7 = CH3 R10 = F (39) R7 = CH2OH R10 = H (40) R7 = CH2OH R10 = F (41 )
Figure imgf000027_0003
up as shown in Scheme 2
Scheme 4. Pyrrolopyrimidines synthesised by Suzuki coupling The furopyrimidines can be synthesised by a method adapted from Yang, B.
S.;Yang, K. M.; Kim, H. J.; Park, I. S.; Park, S. D.; Lee, J. H.; Kwon, H. M.; Woo, B. Y. WO 2005092896, 2005. (scheme 5).
Alternatively, such molecules can be made by a different route as presented by Martin-Kohler, et al. Helv. Chim. Acta (2004) 87, 956-975.
Figure imgf000028_0001
Figure imgf000028_0002
* Letter defines the 6-aryl group as shown in Scheme 2
Scheme 5. Synthetic route to fuoropyrimidines. Further experimental protocols are given in detail in the examples.
Applications
The compounds of the invention target cancer, in particular breast, lung, and head and neck cancers, and other cancers where EGFR receptors are over expressed or mutated. Other cancers of interest are hematologic and gastric cancers. It is also possible however that the compounds of the invention will have utility in the treatment of protozoan linked conditions, leprosy and malaria.
The compounds of the invention may also be used for the treatment of pain, especially neuropathic pain.
Formulation The compounds of the invention are preferably formulated as
pharmaceutically acceptable compositions. The phrase "pharmaceutically acceptable", as used in connection with compositions of the invention, refers to molecular entities and other ingredients of such compositions that are
physiologically tolerable and do not typically produce untoward reactions when administered to a mammal (e.g. human). Preferably, as used herein, the term
"pharmaceutically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia for use in mammals, and more particularly in humans.
The term "carrier" applied to pharmaceutical compositions of the invention refers to a diluent, excipient, or vehicle with which an active compound is administered. Such pharmaceutical carriers can be sterile liquids, such as water, saline solutions, aqueous dextrose solutions, aqueous glycerol solutions, and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin, 18th Edition, incorporated by reference. Particularly preferred for the present invention are carriers suitable for immediate-release, i.e., release of most or all of the active ingredient over a short period of time, such as 60 minutes or less, and make rapid absorption of the drug possible.
The compounds of the invention can be administered in salt, solvate, prodrug or ester form, especially salt form. Typically, a pharmaceutical acceptable salt may be readily prepared by using a desired acid. The salt may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent. For example, an aqueous solution of an acid such as hydrochloric acid may be added to an aqueous suspension of a compound of formula (I) and the resulting mixture evaporated to dryness (lyophilised) to obtain the acid addition salt as a solid.
Alternatively, a compound of formula (I) may be dissolved in a suitable solvent, for example an alcohol such as isopropanol, and the acid may be added in the same solvent or another suitable solvent. The resulting acid addition salt may then be precipitated directly, or by addition of a less polar solvent such as diisopropyl ether or hexane, and isolated by filtration. Suitable addition salts are formed from inorganic or organic acids which form non-toxic salts and examples are hydrochloride, hydrobromide, hydroiodide, sulphate, bisulphate, nitrate, phosphate, hydrogen phosphate, acetate,
trifluoroacetate, maleate, malate, fumarate, lactate, tartrate, citrate, formate, gluconate, succinate, pyruvate, oxalate, oxaloacetate, trifluoroacetate, saccharate, benzoate, alkyl or aryl sulphonates (eg methanesulphonate, ethanesulphonate, benzenesulphonate or p-toluenesulphonate) and isethionate. Representative examples include trifluoroacetate and formate salts, for example the bis or tris trifluoroacetate salts and the mono or diformate salts, in particular the tris or bis trifluoroacetate salt and the mono formate salt.
Those skilled in the art of organic chemistry will appreciate that many organic compounds can form complexes with solvents in which they are reacted or from which they are precipitated or crystallized. These complexes are known as "solvates". For example, a complex with water is known as a "hydrate". Solvates of the compounds of the invention are within the scope of the invention. The salts of the compound of Formula (I) may form solvates (e.g. hydrates) and the invention also includes all such solvates.
The term "prodrug" as used herein means a compound which is converted within the body, e.g. by hydrolysis in the blood, into its active form that has medical effects.
The compounds of the invention are proposed for use in the treatment of, inter alia, cancer. By treating or treatment is meant at least one of:
(i) . preventing or delaying the appearance of clinical symptoms of the disease developing in a mammal;
(ii) . inhibiting the disease i.e. arresting, reducing or delaying the development of the disease or a relapse thereof or at least one clinical or subclinical symptom thereof, or (iii). relieving or attenuating one or more of the clinical or subclinical symptoms of the disease. The benefit to a subject to be treated is either statistically significant or at least perceptible to the patient or to the physician. In general a skilled man can appreciate when "treatment" occurs.
The word "treatment" is also used herein to cover prophylactic treatment, i.e. treating subjects who are at risk of developing a disease in question.
The compounds of the invention can be used on any animal subject, in particular a mammal and more particularly to a human or an animal serving as a model for a disease (e.g. mouse, monkey, etc.).
An "effective dose" means the amount of a compound that, when
administered to an animal for treating a state, disorder or condition, is sufficient to effect such treatment. The "effective dose" will vary depending on the compound, the disease and its severity and the age, weight, physical condition and
responsiveness of the subject to be treated and will be ultimately at the discretion of the attendant doctor.
While it is possible that, for use in the methods of the invention, a compound of the invention may be administered as the bulk substance, it is preferable to present the active ingredient in a pharmaceutical formulation, for example, wherein the agent is in admixture with a pharmaceutically acceptable carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
The term "carrier" refers to a diluent, excipient, and/or vehicle with which an active compound is administered. The pharmaceutical compositions of the invention may contain combinations of more than one carrier. Such pharmaceutical carriers can be sterile liquids, such as water, saline solutions, aqueous dextrose solutions, aqueous glycerol solutions, and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water or aqueous solution saline solutions and aqueous dextrose and glycerol solutions are preferably employed as carriers, particularly for injectable solutions. Suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin, 18th Edition. The choice of pharmaceutical carrier can be selected with regard to the intended route of administration and standard
pharmaceutical practice. The pharmaceutical compositions may comprise as, in addition to, the carrier any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), and/or solubilizing agent(s).
It will be appreciated that pharmaceutical compositions for use in accordance with the present invention may be in the form of oral, parenteral, transdermal, inhalation, sublingual, topical, implant, nasal, or enterally administered (or other mucosally administered) suspensions, capsules or tablets, which may be formulated in conventional manner using one or more pharmaceutically acceptable carriers or excipients.
There may be different composition/formulation requirements depending on the different delivery systems. Likewise, if the composition comprises more than one active component, then those components may be administered by the same or different routes.
The pharmaceutical formulations of the present invention can be liquids that are suitable for oral, mucosal and/or parenteral administration, for example, drops, syrups, solutions, injectable solutions that are ready for use or are prepared by the dilution of a freeze-dried product but are preferably solid or semisolid as tablets, capsules, granules, powders, pellets, pessaries, suppositories, creams, salves, gels, ointments; or solutions, suspensions, emulsions, or other forms suitable for administration by the transdermal route or by inhalation.
The compounds of the invention can be administered for immediate-, delayed-, modified-, sustained-, pulsed-or controlled-release applications.
In one aspect, oral compositions are slow, delayed or positioned release (e.g., enteric especially colonic release) tablets or capsules. This release profile can be achieved without limitation by use of a coating resistant to conditions within the stomach but releasing the contents in the colon or other portion of the GI tract wherein a site has been identified or a delayed release can be achieved by a coating that is simply slow to disintegrate or the two (delayed and positioned release) profiles can be combined in a single formulation by choice of one or more appropriate coatings and other excipients. Such formulations constitute a further feature of the present invention.
Pharmaceutical compositions can be prepared by mixing a therapeutically effective amount of the active substance with a pharmaceutically acceptable carrier that can have different forms, depending on the way of administration.
Typically composition components include one or more of binders, fillers, lubricants, odorants, dyes, sweeteners, surfactants, preservatives, stabilizers and antioxidants.
The pharmaceutical compositions of the invention may contain from 0.01 to
99% weight - per volume of the active material. The therapeutic doses will generally be between about 10 and 2000 mg/day and preferably between about 30 and 1500 mg/day. Other ranges may be used, including, for example, 50-500 mg/day, 50-300 mg/day, 100-200 mg/day.
Administration may be once a day, twice a day, or more often, and may be decreased during a maintenance phase of the disease or disorder, e.g. once every second or third day instead of every day or twice a day. The dose and the administration frequency will depend on the clinical signs, which confirm maintenance of the remission phase, with the reduction or absence of at least one or more preferably more than one clinical signs of the acute phase known to the person skilled in the art.
It is within the scope of the invention for a compound as described herein to be administered in combination with another pharmaceutical, e.g. another drug with known efficacy against the disease in question. The compounds of the invention may therefore be used in combination therapy.
In particular, the compounds of the invention may be used in combination with other kinase inhibitors with other targets, and other chemotherapeutic agents (cisplatin, taxol etc) used to treat cancerous diseases. .
Also within the scope of the invention is the combination of the compounds of the invention with monoclonal antibodies.
The invention will now be further described with reference to the following non limiting examples:
Examples
Examples marked "c" are outside the claims. Those marked * are within the claims.
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Erlotinib 0.4 101 0.6 0.5
EGFR- EGFR- EGFR EGFR native native L858R L861Q
Compound Structure IC50 [nm] % inhibition IC50 [nm] IC50 [nm]
#
[ATP] = Km 100 nm [ATP] = Km [ATP] = Km
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
The selectivity of compounds ( ?)-17v and (5)-19v as a kinase inhibitor was further evaluated towards a panel of kinases at a test concentration of 500 nM, see Table 5. Table 5. Inhibition of various kinases of compound ( ?)-17v and (5)-19v (500 nM), mean % of two measurements. ATP concentration was unless otherwise noticed equal to KM.
Kinase Inhibition (%) Kinase Inhibition (%)
(S)-19v (tf)-17v (5)-19v (tf)-17v
ABL1 22 7 LCK 6 1 1
AURKB (Aurora B) 5 <1 LYN A 40 61
AURKB (Aurora B) 10 <1 LYN B 56 57
BRAF 20 a) 1 MAP2K1 (MEK1) 18 a) 4
CHEK1 (CHK1) 15 <1 MAPK1 (ERK2) 7 <1
CSF1R (FMS) 3 2 MAPK14 (p38 alpha) 13 <1
CSK 5 7 MAPK8 (JNK1) 14 2
EGFR (ErbBl) 100 98 MAPKAPK2 8 <1
EPHA1 <1 <1 MET (cMet) 3 15
EPHB1 4 <1 NEK1 13 <1
ERBB2 (HER2) 50 57 PDGFRA (PDGFR alpha) <1 <1
ERBB4 (HER4) 63 30 PDGFRB (PDGFR beta) <1 3
FER 8 10 PIM2 1 <1
FGFR1 4 <1 PLK1 <1 <1
FGR 58 40 PRKACA (PKA) Not determined 2
FLT1 (VEGFR1) <1 4 PRKCB1 (PKC beta l) 4 <1
FLT4 (VEGFR3) <1 3 PRKCQ (PKC theta) <1 <1
FYN 12 <1 PTK2 (FAK) <1 6
GSK3B (GSK3 beta) 8 7 RET 9 5
HCK 20 7 SRC 9 10
IKBKB (IKK beta) 1 10 STK22B (TSSK2) 8 8
JAK2 9 7 SYK 10 8
KDR (VEGFR2) 12 12 TEK (Tie2) 13 <1
KIT <1 7 YES1 37 12
[ATP] = 100 μΜ
A low activity was seen towards most of the kinases. Activity was seen towards ERBB2 (HER2), EBB4 (HER4), LYN A and LYN B which might have clinical impact.
Also, the selectivity of compounds 28v (R)-2S\, (S)-3la, (5)-31d, (S)-31m, (iS)-31v and (S)-42\ as a kinase inhibitor was evaluated towards a panel of kinases at a test concentration of 500 nM, see Table 6. Table 6. Inhibition of various kinases of selected pyrrolopyrimidines and Erlotinib (500 nM), mean % of two measurements with [ATP] = Km. BRAF, MAP2K1 (MEK1), MAPK8 (JNK1) was profiled at [ATP] = 100 μΜ.
ATP
Kinase Erlotinib 28v (R)-29 (5)-31a (5)-31d (5)-31m (S)-31v (5)-42v
cons.
ABL1 Km app 75 77 75 65 50 84 68 73
AURKB (Aurora B) Km app 20 1 7 19 9 15 8 -3
AURKB (Aurora B) Km app 44 -1 4 13 5 24 4 1
BLK Km app 38 28 33 33 29 42 23 25
BRAF 100 μΜ 3 6 15 -2 4 3 19 18
BTK Km app 0 -4 -10 -1 1 -5 -3 -8
CHEK1 (CHK1 ) Km app -7 -8 -7 10 3 -1 -9 -9
CSF IR (FMS) Km app 38 92 87 88 79 90 82 75
CSK Km app 8 0 -5 9 6 16 10 7
EGFR (ErbB l ) Km app 93 99 100 96 100 98 96 95
EPHA1 Km app 3 1 0 19 15 39 44 10
EPHB 1 Km app 1 1 40 12 16 7 29 17 12
ERBB2 (HER2) Km app 52 58 76 53 53 48 64 59
ERBB4 (HER4) Km app 78 49 70 51 79 71 87 76
FER Km app -1 4 10 6 7 8 18 5
FGFR1 Km app -4 7 16 16 6 13 0 -6
FGR Km app 42 86 92 73 74 84 89 79
FLT1 (VEGFR1 ) Km app 17 -1 -3 8 5 16 2 4
FLT4 (VEGFR3) Km app 44 12 15 6 12 26 14 12
FRAP 1 (mTOR) Km app -2 -14 -16 -12 -2 -10 -3
FRK (PTK5) Km app 16 25 46 33 36 50 58 55
FYN Km app -13 31 29 27 27 44 42 46
GSK3B (GSK3 beta) Km app -8 -1 -13 8 6 5 -4 -8
HCK Km app 18 25 30 29 30 46 38 43
1KB KB (IKK beta) Km app -5 -3 -8 5 3 2 -1 -4
JAK2 Km app 25 -4 -10 34 1 1 1 1 1 1 2
KDR (VEGFR2) Km app 70 22 31 28 15 38 34 35
KIT Km app 17 56 18 20 17 8 18 15
LCK Km app 49 13 1 13 17 36 37 36
LYN A Km app 68 68 78 47 52 74 73 66
LYN B Km app 68 75 81 60 64 70 77 69
MAP2K1 (MEK1 ) 100 μΜ 7 35 41 -8 -4 1 8 8
MAPK1 (ERK2) Km app 7 -1 1 -12 1 1 2 8 6
MAPK14 (p38 alpha) Km app 13 3 6 19 24 24 30 25
MAPK8 (JNK1 ) 100 μΜ 32 -4 -19 0 7 2 26 25
MAPKAPK2 Km app -1 -4 -4 12 8 4 3 0
MET (cMet) Km app 1 1 -14 -15 -9 -2 14 0 1
NEK1 Km app -8 3 -1 0 1 -2 -1 -6
PDGFRA (PDGFR alpha) Km app 5 29 51 6 6 8 -3 -4
PDGFRB (PDGFR beta) Km app -5 8 21 -1 0 0 -1 1 -10
PIM2 Km app -16 1 1 9 5 7 -6 -1 1
PLK1 Km app -9 0 -6 1 -20 -12 2 1
PRKACA (PKA) Km app -10 1 7 1 -1 1 -19 -9
PRKCA (PKC alpha) Km app 2 -1 -4 17 15 5 2 5
PRKCB 1 (PKC beta I) Km app -2 16 10 1 8 10 8 8
PRKCQ (PKC theta) Km app -4 -14 -1 1 12 5 9 6 7
PTK2 (FAK) Km app -3 12 14 -12 -2 -5 -4 -10
RET Km app 64 17 7 14 15 26 19 32
SRC Km app 42 66 75 59 59 74 66 63
STK22B (TSSK2) Km app -13 -7 -8 -3 -2 -2 -1 0
SYK Km app 14 21 28 29 14 42 36 23
TEK (Tie2) Km app 5 0 9 1 0 5 0 -1
YES1 Km app 29 67 73 65 68 81 72 68
Not determined
These studies revealed that in addition to EGFR, the new pyrrolopyrimidine structures also have significant ABL, CSFIR, HER2, HER4 and SRC kinase family selectivity (FGR, LYN A, LYN B, SRC, YES). This profile might be beneficial in a clinical setting.
Some of the new pyrrolopyrimidine based EGFR-TK inhibitors were evaluated in genetically modified model cells Ba/F3-EGFRL858R cells, and four human cancer cell lines including A-431 (epidermoid carcinoma), AU-565 (breast adenocarcinoma), K-562 (leukemia) and C-33A (cervix carcinoma). The results are shown in Table 7. Table 7. Summary of cell proliferation data (IC50) of pyrrolopyrimidines towards
Ba/F3-EGFRL858R, the cancer cell lines A-431, AU-565, C-33A, K-562 and the normal lung fibroblast cell line MRC-5.
Ba/F3- A-431 AU- C-33A K-562 MRC-5 μΜ EG F RL858R μ Μ ( n 565 μ Μ ( n μ Μ ( n ( n = 3) nM (n = 3) = 3) μΜ (n = 2) = 3)
Figure imgf000047_0001
si rV Not
127±29 0.3±0.1 2.5±0.4 0.7±0.0 15±2 determined rmined 3 rmined
Figure imgf000047_0002
Erlotinib 142±65 0.4±0.1 3.3±0.6 0.9±0.0 55±9 6 ± 1 Several of the new compounds compare favourably with Erlotinib in these studies. The synthesised compounds showed a reduced toxicity towards the normal cell line MRC-5 as compared to Erlotinib.
Synthesis - The following protocols can be used to synthesize the compounds mentioned above.
Thienopyrimidine
Methyl 2-aminothiophene-3-carboxylate (1).
l,4-Dithiane-2,5-diol (312.8 g, 2.05 mol) was mixed with MeOH (1000 mL), and methyl cyanoacetate (304.05 g, 3.06 mol) was added while stirring at 0 °C. Then NEt3 (72.55 g, 0.72 mol) was added drop wise over 2.5 h at 0 °C. The reaction temperature was slowly increased to 40 °C over 1 h. and kept stirring. After cooling to rt, the mixture was filtered and the liquid fraction was concentrated. The concentrated oil was extracted with boiling pet. ether (bp. 60-80 °C) (10χ750 mL). The combined organic fraction was concentrated which gave a solid crystalline product. After drying this gave 408.2 g (86%) of 1, mp. 78 - 80 °C Rf (CHC13) = 0.33; 1H NMR (400 MHz, DMSO-d6) δ: 7.24 (s br, 2H, NH2), 6.81 (d, J= 5.8, 1H), 6.27 (d, J= 5.8, 1H), 3.69 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 164.8, 164.0, 124.9, 106.5, 103.7, 50.5. IR (neat, cm"1): 3421, 3319, 1655, 1316, 1273, 676;
HRMS (EI, 70 eV, m/z): 157.0201 (calcd C6H7N02S, 157.0198, M+). Thieno[2,3-d]pyrimidin-4(3H)-one (2).
Methyl 2-aminothiophene-3-carboxylate (1) (26.0 g, 165 mmol) was mixed with formamide (285 mL) and heated at 215 °C for 5 h. The reaction mixture was then cooled to rt. and water (500 mL) was added, followed by extraction with EtOAc (4 x 500 mL). The combined organic fractions were dried over Na2S04, and
concentrated. Upon storage at -18 °C a solid material precipitated which was washed with cold EtOAc (4 x 100 mL). This gave 12.7 g (83.0 mmol, 50%) of 2 as a yellowish solid, mp. 245 °C (dec); 1H NMR (400 MHz, DMSO-d6) δ: 12.45 (s br, 1H), 8.12 (s, 1H), 7.57 (d, J= 5.8, 1H), 7.39 (d, J= 5.8, 1H); 13C NMR (100 MHz, DMSO-de) δ: 164.2, 157.5, 145.6, 124.6, 123.8, 121.6. IR (neat, cm"1): 2879, 1647, 1577, 800, 701, 635; HRMS (EI, 70 eV, m/z): 152.0045 (calcd C6H4N2OS,
152.0044, M+).
6-Bromothieno[2,3-d]pyrimidin-4(3H)-one (3).
Thieno[2,3-d]pyrimidin-4(3H)-one (2) (50.03 g, 329 mmol) was mixed with cone, acetic acid (600 mL), and bromine (30.0 mL, 93.0 g, 582 mmol) was added slowly before the mixture was heated at 80 °C for 1 h. The reaction mixture was then cooled to rt. and filtered. The filtrate was washed with water and saturated aq NaHC03 solution. Drying gave 72.24 g (313 mmol, 95%) of 3 as a light brown solid, mp. 299 - 301 °C (dec); 1H NMR (400 MHz, DMSO-d6) δ: 12.63 (s br, 1H), 8.14 (s, 1H), 7.55 (s, 1H); 13C NMR (100 MHz, DMSO-d6) δ: 165.0, 156.0, 146.5, 125.6, 124.6, 110.3; IR (neat, cm 1): 3074, 1663, 1649, 1504, 755; HRMS (EI, 70 eV, m/z): 229.9146 (calcd C6H3Br79N2OS, 229.9144, M+).
6-Bromo-4-Chlorothieno[2,3-d]pyrimidine (4).
Compound 3 (50.00 g, 216 mmol) was mixed with POCl3 (110 mL, 1180 mmol) and heated at 120 °C for 2.5 h. Then the mixture was quenched into 5 M aq NaOH (900 mL) and ice. The formed precipitate was isolated by filtration and washed with water (3 x 200 mL). Drying gave 50.57 g (203 mmol, 94%) of 4 as a brown solid, mp. 113 - 114 °C. A fraction of this material was further purified: the dry material was applied to the top of a packed silica gel plug and eluted with CH2C12, mp. 1 15 - 116 °C; R/(CH2Cl2/MeOH, 98/2) = 0.67; 1H NMR (400 MHz, DMSO-d6) δ: 8.95 (s, 1H), 7.89 (s, 1H); 13C NMR (100 MHz, DMSC /6) δ: 168.9, 153.2, 152.5, 130.2, 122.9, 118.5; IR (neat, cm 1): 3082, 1508, 14011 , 959, 832, 816, 760; HRMS (EI, 70 eV, m z): 247.8813 (calcd C6H2Br79Cl35N2S, 247.881 1, M+). (R)-6-Bromo-N-(l-phenylethyl)thieno[2,3-d]pyrimidin-4-amine hydrochloride ((R)-ll-HCl) Compound 4 (5.00 mg, 20.04 mmol) was mixed with (R)-l-phenylethanamine (5) (7.27g, 60.0 mmol) and 1-butanol (17 mL), and agitated at 145 °C for 18 h. Then the mixture was cooled to rt, mixed with water (300 mL) and extracted with diethyl ether (3* 150 mL). The combined organic fraction was washed with water (300 mL) and saturated aq NaCl solution (150 mL). After drying over Na2S04 and
concentration in vacuum the crude oil was dried under reduced pressure for 48 h. The crude product was purified by precipitation from diethyl ether (150 mL)/HCl in diethyl ether (8 mL) at 20 °C for 24 h. The product was isolated by filtration and washed with diethyl ether (3 x 60 mL). Drying gave 6.89 g (18.6 mmol, 93%) of (R)-10 HC1 as a white solid, mp. 210 - 214 °C; [a ]∞= -184.3 (c 0.95, DMSO);
HPLC purity (method A): 99%, tR = 27.3 min; 1H NMR (400 MHz, DMSO-d6) δ: 8.27 (s, 1H), 8.23 (d, J = 7.8, 1H), 7.99 (s, 1H), 7.42 - 7.37 (m, 2H), 7.34 - 7.28 (m, 2H), 7.24 - 7.19 (m, 1H), 5.51 - 5.42 (m, 1H), 1.53 (d, J = 7.0, 3H); 13C NMR (100 MHz, DMSO-de) δ: 166.5, 154.8, 154.1, 144.4, 128.3 (2C), 126.7, 126.0 (2C), 122.8, 116.8, 109.6, 49.1, 22.4; IR (neat, cm 1): 1602, 1583, 1474, 1364, 765, 698; HRMS (EI, 70 eV, m/z): 332.9929 (calcd Ci4Hi2Br79N3S, 332.9930, M+).
Potential EGFR inhibitors General procedure Suzuki coupling
Compound (i?)-ll*HCl (201 mg, 0.54 mmol) was mixed with the selected boronic acid (0.81 mmol), fine powdered K2C03 (298 mg, 2.16 mmol), Pd(PPh3)4 (6 mg,
0.005 mmol), 1,4-dioxane (2 mL) and water (2 mL). The reaction was then stirred at 80 °C for 2 - 3 h under N2 atmosphere. The solvent was removed and the product was diluted with water (20 mL) and diethyl ether (25 mL) or EtOAc (25 mL), the wather phase was extracted with diethyl ether (3 x 10 mL) or EtOAc (3 x 10 mL). The combined organic phases were washed with saturated aq NaCl solution (10 mL), dried over andyrous Na2S04, filtered and concentrated in vacuo.
(R)-6-(4-Fluoro-2-methoxyphenyl)-N-(l-phenylethyl)thieno[2,3-d]pyrim amine ((R)-17p) Compound 11 ΉΟ (302 mg, 0.815 mmol), (4-fluor-2-metoxyphenyl)boronic acid (185 mg, 1.09 mmol), K2C03 (422 mg, 3.05 mmol) and Pd(PPh3)4 (11 mg, 9.5 μτηοΐ) were mixed with water (3 mL) and 1,4-dioksan (3 mL) under a nitrogen atmosphere. The mixture was stirred at 60 °C for 4 h. Upon completion the mixture evaporated to dryness, and mixed with diethyl ether (75 mL) and water (30 mL). The water phase was extracted with additional diethyl ether (2x45 mL). The combined organic fraction was dried over anhydrous Na2S04, concentrated and absorbed onto silica-gel. The product was purified by silica-gel column
chromatography (n-pentane/EtOAc, 11/7). After drying this gave 239 mg (0.630 mmol, 77%) of (R)-17p as an off-white solid, mp. 122 - 148 °C; HPLC purity
(Method A): 95%, tR = 29.6 min.; [a ]2° = -292.3 (c 0.65, DMSO); 1H NMR (400
MHz, DMSO-dg) δ: 8.27 (s, 1H). 8.20 (d, J= 8.0, 1H), 8.17 (s, 1H), 7.74 (dd, J = 15.4, 2.0, 1H), 7.44 - 7.42 (m, 2H), 7.34 - 7.30 (m, 2H), 7.24 - 7.20 (m, 1H), 7.14 (dd, J= 11.2, 2.5, 1H), 6.99 - 6.94 (m, 1H), 5.57 - 5.50 (m, 1H), 3.96 (s, 3H), 1.57 (d, J= 7.0, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 165.4, 162.9 (d, J = 246.1), 156.9 (d, J = 10.2), 155.6, 153.2, 144.7, 133.2, 129.4 (d, J = 10.4), 128.3 (2 C), 126.7, 126.1 (2 C), 118.5 (d, J =3.5), 117.0, 116.1, 107.8 (d, J = 21.9), 100.7 (d, J = 26.1), 56.4, 48.9, 22.5; 19F NMR (564 MHz, DMSO-d6): -112.56 (s); IR (neat, cm"1): 3236, 2966, 1577, 1491, 1282, 1108, 1029, 962, 832, 776, 698, 554. HRMS
(ASAP+, m/z): found 380.1229, (calcd. for C2iHi9N3OSF, 380.1233, [M+H]+).
(R)-6-(2,4-Dimethoxyphenyl)-N-(l-phenylethyl)thieno[2,3-d]pyrimidin-4-am ((R)-17u)
Compound 11 ΉΟ (200 mg, 0.540 mmol), (2,4-dimetoksyphenyl)boronic acid (118 mg, 0.647 mmol), K3P04 (389 mg, 1.83 mmol) and Pd(PPh3)4 (7 mg, 6 πιοΐ) were mixed with water (2 mL) and 1,4-dioksan (2 mL) under a nitrogen atmosphere. The mixture was stirred at 60 °C for 9 h. The mixture evaporated to dryness, and mixed with diethyl ether (50 mL) and water (20 mL). After phase separation, the water phase was extracted with additional diethyl ether (2x30 mL). The combined organic fraction was dried over Na2S04, concentrated and absorbed onto silica-gel. The product was purified by silica-gel column chromatography (CH2Cl2/EtOAc, 4/1), TLC (CH2Cl2/EtOAc, 4/1): R/= 0.24. After drying this gave 147 mg (0.375 mmol, 70%) of (i?)-17u as an off-white solid; crystallisation from CH2Cl2/pentane gave a white solid; mp. 130 - 137 °C; HPLC purity (Method A): 98%, tR = 28.9 min.; [a ]2°
= -334.8 (c 0.55, DMSO); 1H NMR (400 MHz, DMSO-d6) δ: 8.24 (s, 1H), 8.14 (d, J = 8.0, 1H), 8.09 (s, 1H), 7.66 - 7.64 (m, 1H), 7.44 - 7.42 (m, 2H), 7.34 - 7.30 (m, 2H), 7.23 - 7.20 (m, 1H), 6.74 - 6.73 (m, 1H), 6.71 - 6.69 (m, 1H), 5.46 - 5.49 (m, 1H), 3.94 (s, 3H), 3.84 (s, 3H), 1.57 (d, J= 7.0, 3H); 13C NMR (100 MHz, DMSO- de) δ: 165.0, 160.8, 156.7, 155.4, 153.2, 144.8, 134.4, 128.9, 128.3 (2C), 126.6, 126.1 (2C), 116.2, 115.3, 114.7, 106.3, 99.1, 55.7, 55.3, 48.9, 22.5; IR (neat, cm"1): 3224, 2934, 1588, 1493, 1308, 1204, 1026, 822, 777, 697, 554. HRMS (ASAP+, m/z): found: 392.1429, (calcd. for C22H22N302S+, 392.1433, [M+H]+).
(R)-3-Methoxy-4-(4-((l-phenylethyl)amino)thieno[2,3-i |pyrimidin-6- yl)benzaldehyde ((R)-17q)
The reaction and extractive work-up were performed as described in Section 0 starting with (4-formyl-2-methoxyphenyl)boronic acid (363 mg, 2.02 mmol). The reaction time was 2.5 h. The crude product was absorbed onto silica and purified by silica gel column chromatography (n-pentane/EtOAc, 1/1). TLC (n-pentane/EtOAc, 1/1): Rf = 0.30. Drying gave 355 mg (0.911 mmol, 68%) of (R)-17q as a bright yellow solid, mp. 155 - 158 °C; HPLC purity (method A): 99%, tR = 30.5 min.;
[a fD° = -486.3 (c 0.98, DMSO); 1H NMR (400 MHz, DMSO-d6) δ: 10.03 (s, 1H),
8.47 (s, 1H), 8.34 (d, J= 7.9, 1H), 8.31 (s, 1H), 8.01 - 7.97 (m, 1H), 7.70 -7.66 (m, 2H), 7.46 - 7.42 (m, 2H), 7.36 - 7.30 (m, 2H), 7.25 - 7.20 (m, 1H), 5.59 - 5.50 (m, 1H), 4.06 (s, 3H), 1.58 (d, J= 7.1, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 192.2, 166.4, 155.8, 155.7, 154.2, 144.6, 136.6, 132.3, 128.3 (2C), 128.2, 127.6, 126.7, 126.1 (2C), 122.8, 119.4, 116.1, 112.2, 56.2, 49.0, 22.4; IR (neat, cm"1): 3277, 3100, 2935, 2810, 1687, 1579, 1499, 1254, 1028, 735, 696, 559; HRMS (APCI/ASAP, m/z): 390.1274 (calcd. C22H2oN302S, 390.1276, [M+H]+).
(R)-2-Methoxy-3-(4-((l-phenylethyl)amino)thieno[2,3-d]pyrimidin-6- yl)benzaldehyde ( (R)-l 7r) Compound ll -HCl (400 mg, 7.08 mmol), (3-formyl-2-metoksyphenyl)boronic acid (236 mg, 1.31 mmol), K3P04 (778 mg, 3.67 mmol) and Pd(PPh3)4 (13 mg, 11 πιοΐ) were mixed with water (4 mL) and 1 ,4-dioksan (4 mL) under a nitrogen
atmosphere. The mixture was stirred at 80 °C for 5 h. The mixture evaporated to dryness, and mixed with diethyl ether (100 mL) and water (40 mL). After phase separation, the water phase was extracted with additional diethyl ether (2x60 mL). The combined organic fraction was dried over anhydrous Na2S04, concentrated and absorbed onto silica-gel. The product was purified by silica-gel column
chromatography (CH2Cl2/EtOAc, 4/1), TLC (CH2Cl2/EtOAc, 4/1): Rf = 0.27, followed by crystallisation from diethyl ether using pentane as anti- solvent. After drying this gave 328 mg (0.842 mmol, 78%) of (i?)-17r as an slight yellowish solid; mp. 71 - 90 °C; HPLC purity (Method A): 98%, tR = 28.0 min.; [a ]2° = -309.6 (c
0.60, DMSO). 1H NMR (400 MHz, DMSC /6) δ: 10.37 (s, 1H), 8.35 (s, 1H), 8.32 (d, J= 7.9, 1H), 8.32 (s, 1H), 8.09 - 8.07 (m, 1H), 7.83 - 7.81 (m, 1H), 7.50 - 7.46 (m, 1H), 7.45 - 7.43 (m, 2H), 7.35 - 7.31 (m, 2H), 7.24 - 7.21 (m, 1H), 5.58 - 5.51 (m, 1H), 3.85 (s, 3H), 1.58 (d, J= 7.0, 3H); 13C NMR (100 MHz, DMSC /6) δ: 189.8, 166.4, 159.0, 155.8, 154.2, 144.6, 131.7, 134.5, 129.9, 128.9, 128.3, 128.1 (2C), 126.7, 126.1 (2C), 125.3, 118.8, 116.2, 64.1, 49.0, 22.4; IR (neat, cm"1): 2967, 1688, 1577, 1508, 1351, 1305, 1245, 1105, 989, 777, 698. HRMS: (ASAP+, m/z): found 390.1272, (calcd. for C22H20N3O2S+, 390.1276, [M+H]+).
(R)-4-Methoxy-3-(4-((l-phenylethyl)amino)thieno[2,3-d]pyrimidin-6- yl)benzaldehyde ((R)-17s)
Compound ll -HCl (1.027 g, 2.770 mmol), (5-formyl-2-metoksyphenyl)boronic acid (584 mg, 3.24 mmol), K3P04 (1.945 g, 9.160 mmol) and Pd(PPh3)4 (32 mg, 32 μτηοΐ) were mixed with water (10 mL) and 1,4-dioksan (10 mL) under a nitrogen atmosphere. The mixture was stirred at 60 °C for 29 h. The mixture evaporated to dryness, and mixed with diethyl ether (150 mL) and water (60 mL). After phase separation, the water phase was extracted with additional diethyl ether (2x70 mL). The combined organic fraction was dried over anhydrous Na2S04, concentrated and the residue crystallisation from MeOH giving 771 mg (1.98 mmol, 72%) of (i?)-17s as a slight yellowish solid; mp. 173-174 °C; HPLC purity (Method A): 97%, t* = 27.5 min.; [a ]™ = -376.4 (c 0.96, DMSO). 1H NMR (400 MHz, DMSO-d6) δ: 9.99
(s, 1H), 8.39 (s, 1H), 8.35 (d, J= 8.0, 1H), 8.31 (d, J= 2.0, 1H), 8.29 (s, 1H), 7.98 (dd, J= 8.5, 2.0, 1H), 7.45 - 7.43 (m, 2H), 7.45 - 7.42 (m, 1H), 7.35 - 7.31 (m, 2H), 7.24 - 7.20 (m, 1H), 5.58 - 5.50 (m, 1H), 4.08 (s, 3H), 1.58 (d, J= 7.0, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 191.4, 166.0, 159.9, 155.7, 154.0, 144.7, 132.4,
132.4, 129.8, 128.3 (2C), 128.0, 126.7, 126,1 (2C), 122.6, 117.9, 115.9, 112.8, 56.5, 49.0, 22.5; IR (neat, cm"1): 3125, 1688, 1592, 1266, 1111, 1016, 820, 767, 703, 640, 553. HRMS (ASAP+, m/z) found 390.1272, (calcd. for C22H2oN302S+, 390.1276, [M+H]+).
(R)-(3-Methoxy-4-(4-((l^henylethyl)amino)thienof2,3-dJpyrimidin-6- yl)phenyl) methanol ((R)-17v)
Compound (i?)-17v (151 mg, 0.388 mmol) was dissolved in methanol (15 mL) and added NaBH4 (15 mg, 0.388 mmol). The mixture was stirred for 10 minutes on an ice bath, before another portion of NaBH4 (15 mg, 0.388 mmol) was added. The stirring was continued at rt for 1 h. The reaction mixture was concentrated in vacuo, diluted with water (25 mL) and EtOAc (25 mL) and the water phase was extracted with EtOAc (3 x 25 mL). The combined organic phases were washed with saturated aq NaCl solution (15 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The crude product was absorbed onto silica and purified by silica gel column chromatography (n-pentane/EtOAc, 3/7). TLC (n-pentane/EtOAc, 3/7): R = 0.21. Drying gave 85 mg (0.217 mmol, 56%) of (i?)-17v as a white solid, mp. 177 - 179 °C; HPLC purity (method A): 98%, tR = 26.6 min.; [a ]£> = -377.3 (c 1.04,
DMSO); 1H NMR (400 MHz, DMSO-d6) δ: 8.26 (s, 1H), 8.22 (s, 1H), 8.19 (d, J = 8.0, 1H), 7.72 - 7.69 (m, 1H), 7.46 - 7.41 (m, 2H), 7.35 - 7.29 (m, 2H), 7.24 - 7.19 (m, 1H), 7.16 - 7.14 (m, 1H), 7.08 - 7.04 (m, 1H), 5.58 - 5.49 (m, 1H), 5.31 (t, J = 5.8, 1H), 4.56 (d, J= 5.7, 2H), 3.95 (s, 3H), 1.57 (d, J= 7.1, 3H); 13C NMR (100 MHz, DMSC /g) δ: 165.5, 155.5, 155.4, 153.5, 144.8, 144.7, 134.1, 128.3 (2C), 127.6, 126.7, 126.1 (2C), 120.1, 118.9, 116.5, 116.2, 110.2, 62.6, 55.8, 48.9, 22.5; IR (neat, cm"1): 3281, 2971, 2933, 2361, 1588, 1499, 1019, 767, 698; HRMS (APCI/ASAP, m/z): 392.1426 (calcd. C22H22N302S, 392.1433, [M+H]+). (R)-(2-Methoxy-3-(4-((l-phenylethyl)amino)thieno[2,3-d]pyrimidin-6- yl)phenyl)methanol ((R)-17w)
Compound (i?)-17w (151 mg, 0.388 mmol) was reduced as described for ( ?)-17v. The product was first purified by silica-gel column chromatography
(EtOAc/pentane, 6/4), TLC (EtOAc/pentane, 6/4): R/ = 0.24. Then the resulting oil was dissolved in Et20/EtOAc and pentane was added drop wise until crystallisation started. Isolation and drying gave 110 mg (0.281 mmol, 73%) of (i?)-17w as a white powder; mp. 113-124 °C; HPLC purity (Method A): 99%, tR = 24.2 min.; [a ]2° = - 274.9 (c 0.59, DMSO); 1H NMR (400 MHz, DMSO-d6) δ: 8.29 (s, 1H), 8.26 (d, J = 7.9, 1H), 8.26 (s, 1H), 7.66 (dd, J= 7.8, 1.4, 1H), 7.50 - 7.48 (m, 1H), 7.45 - 7.43 (m, 2H), 7.35 - 7.31 (m, 2H), 7.31 - 7.29 (m, 1H), 7.24 - 7.20 (m, 1H), 5.56 - 5.49 (m, 1H), 5.25 (t, J= 5.6, 1H), 4.64 (d, J= 5.4, 2H), 3.66 (s, 3H), 1.57 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 166.1, 155.7, 153.8, 153.7, 144.7, 136.7, 133.5, 128.8, 128.3 (2C), 127.0, 126.7, 126.3 (2C), 126.1, 124.6, 117.6, 116.2, 61.2, 57.7, 49.0, 22.5; IR (neat, cm"1): 3849, 3740, 3242, 2353, 2156, 2026, 1574, 1501, 997, 774, 691, 613, 577. HRMS (ASAP+, m/z): found 392.1425, (calcd for
C22H22N302S+, 392.1433, [M+H]+). (R)-(4-Methoxy-3-(4-((l^henylethyl) mino)thienof2,3-dJpyrimidin-6- yl)phenyl)methanol ((R)-17x)
Compound (i?)-17x (400 mg, 1,027 mmol) was reduced as described for (i?)-17v, but reacted at 50 °C. The product was first purified by silica-gel column
chromatography (EtOAc/pentane, 7/3), TLC (EtOAc/pentane, 7/3): R = 0.24. Then the resulting oil was dissolved in Et20/EtOAc and pentane was added drop wise until crystallisation started. Isolation and drying gave 76 mg (0.19 mmol, 19%) of as a white powder, mp. 196 - 197 °C; HPLC purity (Method A): 98%, tR = 23.2 min.; [a ]2° = -350.3 (c 0.54, DMSO); 1H NMR (400 MHz, DMSO-d6) δ: 8.29
(d, J= 8.0, 1H), 8.26 (s, 2H), 7.76 (d, J= 1.9, 1H), 7.44 - 7.43 (m, 2H), 7.34 - 7.31 (m, 2H), 7.30 - 7.29 (m, 1H), 7.24 - 7.20 (m, 1H), 7.17 - 7.14 (m, 1H), 5.57 - 5.50 (m, 1H), 5.25 (t, J= 5.5, 1H), 4,52 (d, J= 5,4, 2H), 3.94 (s, 3H), 1.57 (d, J= 7.0, 3H); 13C NMR (100 MHz, DMSO-dg) δ: 165.6, 155.6, 154.3, 153.6, 144.8, 135.2, 134.1, 128.3 (2C), 127.9, 126.6, 126.1 (2C), 126.0, 121.4, 116.6, 116.0, 112.3, 62.4, 56.0, 48.9, 22.5; HRMS (ASAP+, m/z): found 392.1430, (calcd for C22H22N302S+, 392.1433, [M+H]+).
(S)-2-(4-((2-Hydroxy-l-phenylethyl)amino)thieno[2,3-d]pyrimidin-6-yl)phenol ((S)-19c)
Compound (5)-13·ΗΟ (150 mg, 0.39 mmol) was mixed with (2- hydroxyphenyl)boronic acid (80 mg, 0.58 mmol), K2C03 (214 mg, 1.55 mmol), Pd(PPh3)4 (4.48 mg, 0.039 mmol), 1 ,4-dioxane (2 mL) and water (2 mL). The reaction was then stirred at 80 °C for 3 h under N2 atmosphere. Dioxane was removed under reduced pressure and the product was dissolved in water (20 ml) and extracted with EtOAc (3 x 25 mL) and washed with and sat. NaCl solution (15 mL). The organic phase was dried over anhydrous Na2S04, filtered, absorbed onto silica and purified with silica gel column chromatography (EtOAc). TLC (EtOAc): R/ =
0.30. The purified product was dissolved in diethyl ether (2 mL) and slowly added to n-pentane (30 mL) to give precipitation. Drying gave 116.4 mg (0.32 mmol, 82%) of (S)-19c as a white solid, mp. 137 - 139 °C; HPLC purity (method A): 97%, tR = 23.5 min.; [a]D 25 = -293 (c 0.84, DMSO); 1H NMR (400 MHz, DMSO- 6) δ: 10.4 (s, 1H), 8.28 (s, 1H), 8.24 (s, 1H), 8.18 - 8.16 (m, 1H), 7.70 - 7.68 (m, 1H), 7.45 - 7.43 (m, 2H), 7.34 - 7.30 (m, 2H), 7.24 - 7.19 (m, 2H), 7.02 - 7.00 (m, 1H), 6.96 - 6.93 (m, 1H), 5.49 - 5.42 (m, 1H), 5.04 - 5.01 (m, 1H), 3.83 - 3.72 (m, 2H); 13C NMR (100 MHz, DMSO- e) δ: 165.8, 156.6, 154.4, 153.8, 141.7, 135.3, 129.8, 128.6 (2C), 128.3, 127.5 (2C), 127.3, 120.6, 120.1, 117.0, 116.9, 116.8, 65.2, 60.2; IR (neat, cm" l): 3057, 1578, 1450, 747, 698; HRMS (APCI/ASAP, m/z): 364.118 (calcd.
C20Hi7N3O2S, 364.1120, [M+H]+). (S)-2-((6-(2-Methoxyphenyl)thieno[2,3-d]pyrimidin-4-yl)amino)-2-phenylethan l-ol ((S)-19d)
Compound (5)-13ΉΟ (150 mg, 0.39 mmol) was mixed with (2- methoxyphenyl)boronic acid (88 mg, 0.58 mmol), K2C03 (214 mg, 1.55 mmol), Pd(PPh3)4 (4.48 mg, 0.039 mmol), 1 ,4-dioxane (2 mL) and water (2 mL). The reaction was then stirred at 80 °C for 4 h under N2 atmosphere. Dioxane was removed under reduced pressure and the product was dissolved in water (20 ml) and extracted with EtOAc (3 x 25 mL) and washed with and sat. NaCl solution (15 mL). The organic phase was dried over anhydrous Na2S04, filtered, absorbed onto silica and purified with silica gel column chromatography (n-pentane/EtOAc, 2/8). TLC (n-pentane/EtOAc, 2/9): R/ = 0.29. The purified product was dissolved in diethyl ether (2 mL) and slowly added to n-pentane (30 mL) to give precipitation. Drying gave 129.9 mg (0.32 mmol, 82%) of (S)-19d as a white solid, mp. 186-189 °C; HPLC purity (method A): 98%, tR = 23 min.; [a]D 25 = -307.8 (c 0.70, DMSO); 1H NMPv (400 MHz, DMSC /6) δ: 8.28-8.26 (m, 2H), 8.18-8.16 (m, 1H), 7.78-7.76 (m, lH), 7.45-7.37 (m, 3H), 7.34-7.30 (m, 2H), 7.25-7.19 (m, 2H), 7.13-7.09 (m, 1H), 5.50-5.45 (m , 1H), 5.06-5.03 (m, 1H), 3.96 (s, 3H), 3.83-3.73 (m, 2H); 13C NMPv (100 MHz, DMSC /6) 6: 166.1 , 156.7, 155.9, 154.0, 141.6, 134.3, 130.2, 128.6 (2C), 128.4, 127.5 (2C), 127.3, 122.3, 121.6, 1 17.5, 1 16.8, 1 13.0, 65.1 , 56.8, 56.3; IR (neat, cm"1) 3274, 3085, 2836, 1582, 1509, 1024, 747, 697; HRMS
(APCI/ASAP, m/z): 378.1274 (calcd. C2iHi9N302S, 378.1276, [M+H]+).
(S)-2-((6-(4-(Hydroxymethyl)phenyl)thieno[2,3-d]pyrimidin-4-yl)amino)-2- phenylethan-l-ol ((S)-19m)
Compound (5)-13·ΗΟ (150 mg, 0.39 mmol) was mixed with (4- (hydroxymethyl)phenyl)boronic acid (88 mg, 0.58 mmol), K2C03 (214 mg, 1.55 mmol), Pd(PPh3)4 (4.48 mg, 0.039 mmol), 1 ,4-dioxane (2 mL) and water (2 mL). The reaction was then stirred at 80 °C for 4 h under N2 atmosphere. Dioxane was removed under reduced pressure and the product was dissolved in water (20 ml) and extracted with EtOAc (3 x 25 mL) and washed with and sat. NaCl solution (15 mL). The organic phase was dried over anhydrous Na2S04, filtered, absorbed onto silica and purified with silica gel column chromatography EtOAc. TLC EtOAc: R/ = 0.18. The purified product was dissolved in diethyl ether (2 mL) and slowly added to n- pentane (30 mL) to give precipitation. Drying gave 123 mg (0.33 mmol, 84%) of (5)-19m as a white solid, mp. 206-209 °C; HPLC purity (method A): >96%, tR = 19.3 min.; [a]D 25 = -309.3 (c 0.63, DMSO); 1H NMR (400 MHz, DMSO-d6) δ: 8.27 (s,lH), 8.24 (s,lH), 8.19-8.17 (m, 1H), 7.69-7.67 (m,2H), 7.46-7.44 (m, 4H), 7.34- 7.31 (m, 2H), 7.26-7.21 (m, 1H), 5.48-5.43 (m, 1H), 5.30-5.28 (m, 1H), 5.06-5.03 (m, 1H), 4.56-4.55 (m, 2H), 3.82-3.72 (m, 2H); 13C NMR (100 MHz, DMSO-d6) δ: 165.4, 156.9, 154.2, 143.6, 141.5, 138.6,134.1, 128.6 (2C),127.8(2C), 127.5(2C), 127.4 125.8 (2C), 118.1, 115.7, 65.2, 62.9, 56.8; IR (neat, cm"1): 3326, 3261, 1595, 1316, 1041, 699; HRMS (APCI/ASAP, m/z): 378.1276 (calcd. C21H19N3O2S, 378.1276, [M+H]+).
(S)-4-(4-((2-Hydroxy-l-phenylethyl)amino)thieno[2,3-d]pyrimidin-6-yl)-3- methoxybenzaldehyde ((S)-19q)
The reaction and extractive work-up were performed as described in for 17q starting with (4-formyl-2-methoxyphenyl)boronic acid (168 mg, 0.931 mmol) and compound (S)-17 (301 mg, 0.778 mmol). The reaction time was 3 h. The crude product was absorbed onto silica and purified by silica gel column chromatography (rc-pentane/EtOAc, 1/9). TLC (rc-pentane/EtOAc, 1/9): R/= 0.27. Drying gave 225 mg (0.554 mmol, 71%) of (5)-19q as a bright yellow solid, mp. 203 - 206 °C; HPLC purity (method A): 96%, tR = 22.6 min.; [a ¾° = -249.8 (c 0.92, DMSO); 1H NMR
(400 MHz, DMSO-de) δ: 10.04 (s, 1H), 8.52 (s, 1H), 8.33 - 8.27 (m, 2H), 8.04 - 7.99 (m, 1H), 7.71 - 7.66 (m, 2H), 7.48 - 7.42 (m, 2H), 7.37 - 7.29 (m, 2H), 7.27 - 7.20 (m, 1H), 5.53 - 5.43 (m, 1H), 5.06 (t, J= 5.6, 1H), 4.07 (s, 3H), 3.83 - 3.73 (m, 2H); 13C NMR (100 MHz, DMSC /6) δ: 192.2, 166.3, 156.5, 155.7, 154.2, 141.0, 136.6, 132.3, 128.20 (2C), 128.18, 127.6, 127.0 (2C), 126.9, 122.8, 119.5, 116.2, 112.2, 64.6, 56.4, 56.2; IR (neat, cm"1): 3270, 3064, 2837, 1693, 1592, 1509, 1266, 1154, 1030, 773, 696, 562; HRMS (APCI/ASAP, m/z): 406.1222 (calcd. C22H20N3O3S, 406.1225, [M+H]+). (S)-2-((6-(4-(Hydroxymethyl)-2-methoxyphenyl)thieno[2,3-d]pyrimidin-4- yl)amino)-2-phenylethanol ((S)- 19v)
Compound ((S)-17p) (71 mg, 0.174 mmol) was dissolved in methanol (20 mL) and added NaBH4 (15 mg, 0.396 mmol). The mixture was stirred for 20 min at rt, before another portion of NaBH4 (15 mg, 0.396 mmol) was added. The stirring was continued at rt for 1 h. The reaction mixture was concentrated in vacuo, diluted with water (25 mL) and EtOAc (25 mL) and the water phase was extracted with EtOAc
(3 x 25 mL). The combined organic phases were washed with saturated aq NaCl solution (15 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The crude product was absorbed onto silica and purified by silica gel column chromatography (EtOAc). TLC (EtOAc): R/ = 0.20. Drying gave 81 mg (0.081 mmol, 47%) of (S)-19v as a pale yellow solid, mp. 183 - 185 °C; HPLC purity
(method A): 96%, tR = 18.8 min.; [a ¾» = -332.0 (c 1.00, DMSO); 1H NMR (400
MHz, DMSO- g) δ: 8.27 - 8.24 (m, 2H), 8.15 (d, J= 8.0, 1H), 7.75 - 7.71 (m, 1H), 7.46 - 7.42 (m, 2H), 7.35 - 7.29 (m, 2H), 7.26 - 7.20 (m, 1H), 7.17 - 7.15 (m, 1H), 7.09 - 7.04 (m, 1H), 5.50 - 5.43 (m, 1H), 5.32 (t, J= 5.7, 1H), 5.03 (t, J= 5.7, 1H), 4.56 (d, J= 5.7, 2H), 3.95 (s, 3H), 3.83 - 3.72 (m, 2H); 13C NMR (100 MHz, DMSO-de) δ: 165.45, 156.19, 155.37, 153.46, 144.80, 141.19, 134.02, 128.17 (2C), 127.59, 127.01 (2C), 126.88, 120.16, 118.91, 116.53, 116.31, 110.22, 64.68, 62.56, 56.33, 55.81; IR (neat, cm"1): 3267, 3027, 2834, 1595, 1509, 1049, 772, 692, 559; HRMS (APCI/ASAP, m/z): 408.1380 (calcd. C22H22N303S, 408.1382, [M+H]+).
Pyrrolopyrimidines route 1 The following is representative: Ethyl 2-amino-5-phenyl-lH-pyrrole-3-carboxylate (25)
Ethyl 3-amino-3-iminopropanoate hydrochloride (23) (8.3 g, 50.0 mmol) and NaOEt (5.1 g, 75.0 mmol) were dissolved in abs. EtOH at 0 °C and stirred for 20 min. under argon. The mixture was heated to 60 °C, and 2-bromo-l-phenylethanone (24) (5.0 g, 25.0 mmol) was added portion wise over 5 min. After 1.5 h the mixture was cooled to 20 °C and the solvent was evaporated under reduced pressure. The residue was diluted with distilled water (20 mL) and extracted with EtOAc (3 X80 mL). The organic layer was washed with water (3 X20 mL) and brine (3 X20 mL). The combined water fractions were back extracted with EtOAc (2X20 mL). The organic phases were dried over MgSC^, and the solvent was evaporated under reduced pressure. Purification was by silica-gel column chromatography (EtOAc/n-pentane, 7/3). This gave 3.7 g (16.1 mmol, 64%) of a beige solid, mp. 101-104 °C; 1H NMR (400 MHz, DMSO-<¾) δ: 10.72 (s, 1H, NH), 7.50-7.44 (m, 2H), 7.32-25 (m, 2H), 7.1 1-7.05 (m, 1H), 6.46 (d, J=2.4, 1H), 5.65 (s, 2H), 4.15 (q, J=6.8, 2H), 1.25 (t, J=6.8, 3H). 13C NMR (100 MHz, DMSO-<¾), δ: 165.4, 148.6, 132.6, 129.1 (2C), 125.4, 123.6, 122.8 (2C), 104.0, 93.9, 58.6, 15.2. HRMS (ESI): 231.1 124 (calcd C13H14N2O2, 231.1 128, M+H+). IR (neat, cm"1): 3417, 3328, 1662, 1589, 1281 , 1 120, 757, 691.
6-Phenyl-7H-pyrrolo [2,3-d\ pyrimidin-4-ol (26)
To a solution of anhydrous DMF (28 mL), formic acid (1 1.3 mL), ethyl 2-amino-5- phenyl- lH-pyrrole-3-carboxylate (25) (3.50 g, 16.57 mmol) and an excess formamide (75 mL) were added. The mixture was heated at 150 °C for 20 h. Then 2- propanol (12 mL) was added, and the mixture was cooled to 20 °C. The formed precipitate was isolated by filtration, washed with 2-propanol (10 mL) and n-hexane (2X 15 mL), and dried under reduced pressure to yield 2.1 g (9.9 mmol, 65%) of a beige solid, mp. > 300 °C, lit.28 >300 °C. 1H NMR (400 MHz, DMSO-<¾) δ: 12.33 (s, 1H, NH), 1 1.84 (s, 1H), 7.87 (s, 1H), 7.82 (m, 2H), 7.41 (m, 2H), 7.27 (m, 1H), 6.93 (s, 1H). 13C NMR (100 MHz, OMSO-d6) δ: 158.2, 149.4, 143.7, 133.1 , 131.4, 128.8 (2C), 127.2, 124.6 (2C), 109.1 , 99.2. HRMS (ESI): 212.0821 (calcd
C12H9N3O, 212.0818, M+H+). IR (neat, cm"1): 2786, 1650, 1241 , 900, 749, 693, 620. Preparation of the 6-aryl-4-chloro-7H-pyrrolo[2,3-i ]pyrimidines (27)
6-Phenyl-7H-pyrrolo[2,3-d]pyrimidin-4-ol (26) (1.57 g, 7.43 mmol) and neat POCI3 (13.5 mL) were mixed and reacted at 90 °C for 3 h. The solution was cooled with an ice-salt bath, and water (60 mL) was added. NaOH (8 M, 80 mL) was used to adjust the pH to 12. The formed precipitate was isolated by filtration, washed with water and n-pentane and dried to give 1.49 g (6.49 mmol, 87%>) of a yellowish solid, mp. 255-257 °C. 1H NMR (400 MHz, OMSO-d6) δ: 13.03 (s, 1H, NH), 8.59 (s, 1H), 8.02 (m, 2H), 7.51 (m, 2H), 7.42 (m, 1H), 7.1 1 (d, J=2.0). 13C NMR (100 MHz, OMSO-d6) δ: 153.4, 150.9, 150.3, 140.7, 130.6, 129.5 (3C), 126.4 (2C), 1 18.3, 95.9. HRMS (ESI): 230.0488 (calcd Ci2H8N3Cl, 230.0480, M+H+). IR (neat, cm 1): 3093, 2955, 2826, 1556, 1234, 983, 866, 751 , 698.
(S)-2-Phenyl-2-((6-phenyl-7H-pyrrolo [2,3-d] pyrimidin-4-yl)amino)ethanol ((S)- 31a)
4-Chloro-6-phenyl-7H-pyrrolo[2,3-d]pyrimidine (150 mg, 0.653 mmol) and (S)-2- amino-2-phenylethan-l-ol (269 mg, 1.959 mmol) were added to 1-butanol (3 mL) under N2 atmosphere and heated at 145 °C for 21 h. After cooling to rt, 1-butanol was evaporated in vacuo, and water (30 mL) was added. The mixture was extracted with EtOAc (3x30 mL). The combined organic phases were washed with sat. NaCl solution (10 mL) and dried over anhydrous Na2S04, filtrated and evaporated. The obtained material was purified by silica gel chromatography with EtOAc. TLC EtOAc,: R/ = 0.1. Isolation and drying gave 161.7 mg, (0.489 mmol, 75%) of S)- 31a as a white solid, mp. 219-22 °C; HPLC purity (method A): 96%, tR = 20.0 min[a = -273.0 (c 0.67, DMSO); 1H NMR (400 MHz, DMSO- 6) δ: 12.04 (s, 1H),
8.06 (s, 1H), 7.81 - 7.79 (m, 2H), 7.73 - 7.71 (m, 1H), 7.47 - 7.43 (m, 4H), 7.33 - 7.28 (m, 3H), 7.23 - 7.20 (m, 1H), 7.14 (s, 1H), 5.46 - 5.41 (m, 1H), 4.97 (t, J = 5.6, 1H), 3.80 - 3.70 (m, 2H); 13C NMR (100 MHz, DMSO-d6) δ: 165.1 , 152.2,152.0,
142.4, 133.9, 132.3, 129.4 (2C), 128.5 (2C), 127.7, 127.5 (2C), 127.1 , 125.1(2C),
104.5, 96.6, 65.5, 56.5; HRMS (APCI/ASAP, m/z): 331.1558 (calcd. C20Hi8N4O, 331.1559, [M+H]+). (S)-N-(2-Methoxy-l^henylethyl)-6^henyl-7H^yrrolof2,3-dJpyrimidin-4- mine ((S)-32a)
4-Chloro-6-phenyl-7H-pyrrolo[2,3-d]pyrimidine (27) (150 mg, 0.653mmol) and (S)- 2-methoxy-l-phenylethan-l -amine (296.2 mg, 1.959 mmol) were added to 1-butanol (3 mL) under N2 atmosphere and heated at 145 °C for 21 h. After cooling to rt, 1- butanol was evaporated in vacuo, and water (30 mL) was added. The mixture was extracted with EtOAc (3x30 mL). The combined organic phases were washed with sat. NaCl solution (10 mL) and dried over anhydrous Na2S04, filtrated and evaporated. The crude product was absorbed onto silica and purified by silica gel column chromatography (n-pentane/EtOAc, 2/8). TLC (n-pentane/EtOAc, 2/8): R/ = 0.20. Isolation and drying gave 149.6 mg, (0.434 mmol, 67%) of (5)-32a as a white solid. 1H NMR (400 MHz, DMSO-d6) δ: 12.05 (s, 1H), 7.86-7.83 (m, 1H), 7.81-7.79 (m, 2H), 7.48-7.43 (m, 4H), 7.34-7.28 (m, 3H), 7.25-7.21(m, 1H), 7.14 (s, 1H), 5.67-5.62 (m, 1H), 3.76-3.72 (m, 1H), 3.65-3.61 (m, 1H), 3.31 (s, 3H); 13C NMR (100 MHz, DMSO-de) δ; 155.8, 152.2, 152.0, 141.8, 133.9, 132.2, 129.4 (2C), 128.6 (2C), 127.7, 127.5 (2C), 127.4, 124.9 (2C), 104.4, 96.6, 75.6, 60.2, 58.5; HRMS (APCI/ASAP, m/z): 345.1714 (calcd. C2iH20N4O, 345.1715, [M+H]+).
Pyrrolopyrimidines route 1
The following is representative 4-Chloro-7H-pyrrolo[2,3-i ]pyrimidine (34)
Compound 33 (70.0 g, 518 mmol) was mixed with POCl3 (300 ml, 3.2 mol) and heated to 90 °C for 6 hours. The reaction mixture was poured
Figure imgf000062_0001
into water and ice to a total volume of 2.3 1, and the pH was adjusted to
12 using sodium hydroxide (pellets, 700 g). The resulting mixture was cooled to 22
°C, and left for precipitation. The solution was filtrated, and the resulting solid was extracted with EtOAc (a total volume of 7 1 of organic phase was collected). The combined organic phases were washed with brine, dried over Na2S04 and concentrated. The crude product was crystallised from acetonitrile (59 g in 1.5 1). This gave 51.4 g of 34 (335 mmol, 65 %) as a brown solid, mp 190 - 191 °C
(acetonitrile), (lit.[7] 189 - 190 °C) ; purity: 99% (HPLC), tR= 12.5 min; 1H NMR
(400 MHz, DMSO-dg) δ: 12.57 (s, 1H), 8.59 (s, 1H), 7.70 (d, J= 3.5, 1H), 6.60 (d, J = 3.5, 1H); 13C NMR (100 MHz, DMSO-d6) δ: 151.8, 150.4, 150.3, 128.4, 116.5, 98.8; 13C NMR matched that of Seala et al. except C-2 which resided at 150.3 ppm instead of 150.0[8]; IR (neat, cm 1): 3118, 2964, 2819, 1553, 1352, 1259, 1205, 962, 844, 737; HRMS (EI, 70 eV, m/z): 153.0091 (calcd. C6H4N3 35C1, 153.0088, [M]+).
4-Chloro-7-(phenylsulfonyl)-7H-pyrrolo[2,3-i ]pyrimidine (35) Compound 34 (19.8 g, 129 mmol) was mixed with sodium tert- butoxide (13.1 g, 136 mmol) in dry THF (600 mL) and cooled to 10 °C. Benzenesulfonyl chloride (18.0 ml, 141 mmol) was added and the mixture was stirred at 22 °C for 2 hours before water (50 mL) was
Figure imgf000063_0001
added. The mixture was stirred for another 10 minutes and concentrated. The residue was mixed with water (600 ml) and extracted with EtOAc (4 x 600 ml). The combined organic phases were washed with brine (200 ml), dried over Na2S04, filtered and concentrated in vacuo. The crude product was crystallised from acetonitrile (40 g in 400 ml). Drying gave 34.8 g (119 mmol, 92%) of 52 as a brown solid, mp 168 - 169 °C (lit.[9] 169 - 170 °C; purity > 99% (HPLC), tR = 24.1 min; 1H NMR (400 MHz, DMSO-d6) δ: 8.82 (s, 1H), 8.18 - 8.14 (m, 3H), 7.81 - 7.77 (m, 1H), 7.69 - 7.66 (m, 2H), 6.98 (d, J= 4.0, 1H). 13C NMR (100 MHz, DMSO-de) δ: 152.6, 152.1, 150.7, 136.6, 135.5, 129.9 (2C), 128.6, 127.8 (2C), 119.3, 103.4; IR (neat, cm 1): 1542, 1373, 1142, 1016, 726; HRMS (EI, 70 eV, m/z): 293.0021 (calcd. Ci2H802N3S35Cl, 293.0020, [M]+).
4-Chloro-6-iodo-7-(phenylsulfonyl)-7H-pyrrolo[2,3-i ]pyrimidine (36)
Compound 35 (10.0 g, 34.0 mmol) was mixed with dry THF (300 ml) under nitrogen atmosphere, and cooled to -78 °C. Lithium diisopropylamide (2M in THF/n-heptane/ethylbenzene, 26 mL, 51.1 mmol) was added drop wise over 30 minutes using a syringe pump.
Figure imgf000063_0002
After 1 hour, iodine (11.2 g, 44.3 mmol) dissolved in dry THF (60 ml) was added drop wise over 30 minutes using a syringe pump. The reaction mixture was stirred for 1 hour before HC1 (1M, 180 ml) was added. The mixture was stirred until the temperature reached 22 °C, and concentrated. To this residue CH2C12 (250 ml) and water (200 ml) were added. The layers were separated and the aqueous phase was extracted with CH2C12 (2 x 90 ml). The combined organic phases were washed with brine (100 ml), dried over Na2S04, filtered and concentrated. The crude, yellow product was crystallised from acetonitrile (150 ml). This gave 10.5 g (25.1 mmol, 74 %) of compound 53 as a pale yellow solid, mp 190 °C (dec); purity: 99% (HPLC), tR= 25.9 min; 1H NMR (400 MHz, DMSO-d6) δ: 8.77 (s, 1H), 8.12 - 8.09 (m, 2H), 7.82 - 7.78 (m, 1H), 7.71 -7.67 (m, 2H), 7.34 (s, 1H); I3C NMR (100 MHz, DMSO-de) δ: 153.3, 152.1, 150.0, 137.2, 135.5, 130.0 (2C), 127.6 (2C), 120.6, 116.6, 84.6; IR (neat, cm 1): 1394, 1087, 724; HRMS (EI, m/z): 418.8988 (calcd. Ci2H702N3S35ClI, 418.8987, [M]+).
4-Chloro-6-iodo-7H-pyrrolo [2,3-i/] yrimidine (37)
Compound 53 (14.4 g, 34.2 mmol) was mixed with THF (300 ml) and a NaOH/MeOH-solution (5M, 49 ml, 244 mmol) and stirred for 1 hour
Figure imgf000064_0001
before a saturated aq. NH4C1 solution (300 ml) was added. The mixture was concentrated in vacuo to remove most of the solvent before it was filtered and washed with water (2 x 30 ml). The crude product was precipitated from boiling acetonitrile (9.2 g in 100 ml). This gave 8.99 g (32.2 mmol, 94 %) of 54 as a white solid, mp 220 °C (dec); purity: 98% (HPLC), tR = 17.5 min; 1H NMR (400 MHz, DMSO-dg) δ: 13.14 (s, 1H), 8.53 (s, 1H), 6.89 (d, J= 1.8, 1H); 13C NMR (100 MHz, DMSO-dg) δ: 153.8, 150.3, 148.3, 118.6, 108.2, 84.8; IR (neat, cm"1): 2790, 1549, 1327, 983, 752; HRMS (EI, 70 eV, m/z): 278.9053 (calcd. C6H3N3 35C1I, 278.9050, [M]+).
General procedure thermal amination of 37
Compound 37 (1 mmol) was mixed with the selected amine (3 mmol, 3 eq) and n- butanol (5 ml) and agitated at 145 °C for 14-24 hours. Then the mixture was cooled to 22 °C, diluted with water (15 ml) and ethyl acetate (40 ml). After phase separation, the water phase was back extracted with more EtOAc (2 x 10 ml). The combined organic phases were washed with brine (10 ml), dried over anhydrous Na2S04, filtered and concentrated in vacuo, before the crude mixture was purified as specified for each individual compound. V-Benzyl-6-iodo-7H-pyrrolo [2,3-i ] pyrimidin-4-amine (38)
The synthesis was performed as described above starting with compound 37 (250 mg, 0.895 mmol) and benzylamine (0.293 ml, 2.68 mmol). The reaction time was 22 hours. The crude product was
Figure imgf000064_0002
absorbed onto Celite 545 and purified by silica-gel column chromatography (diethyl ether/THF, 85/15, R/ = 0.26). Drying gave 266 mg (0.759 mmol, 85%) of 38 as a pale yellow solid, mp 210 - 212 °C (dec); HPLC purity: 96%, tR = 19.8 min; 1H NMR (400 MHz, DMSO-d6) δ: 12.08 (s, 1H), 8.05 (s, 1H), 7.95 (t, J= 6.0, 1H), 7.33-7.28 (m, 4H), 7.25-7.20 (m, 1H), 6.81 (s, 1H), 4.70 (d, J= 6.0, 2H). 13C NMR (100 MHz, DMSO-de): δ: 154.2, 152.6, 151.6, 140.1, 128.2 (2C), 127.1, 126.7 (2C), 108.2, 105.0, 72.9, 43.0. IR (neat, cm"1): 3415, 3091, 3012, 2914, 2795, 2701, 1578, 1413, 1343, 886, 721, 695; HRMS (APCI/ASAP, m/z): 351.0109 (calcd. Ci3Hi2N4I, 351.0107 [M+H]+).
o-N-(l-phenylethyl)-7H-pyrrolo[2,3-i ]pyrimidin-4-amine is was performed as described above starting with
Figure imgf000065_0001
compound 37 (2.01 g, 7.20 mmol) and (i?)-l-phenylethaneamine (2.73 mL, 21.5 mmol). The reaction time was 24 hours. The crude product was purified by silica-gel column chromatography (EtOAc/n-pentane, 4/1 , Rf = 0.28). Drying gave 2.24 g (6.20 mmol, 86%) of (R)-39 as a pale yellow solid, mp 214 - 216 °C; HPLC purity: 96%, tR = 20.9 min; [a]D 2° = -227.4 (c 0.34, DMSO); 1H NMR (400 MHz, DMSO-dg) δ: 12.06 (s, 1H), 7.97 (s, 1H), 7.74 - 7.72 (m, 1H), 7.40 - 7.38 (m, 2H), 7.31 - 7.27 (m, 2H), 7.20 - 7.16 (m, 1H), 6.93 (s, 1H), 5.50 - 5.42 (m, 1H), 1.50 (d, J = 6.9, 3H); 13C NMR (100 MHz, DMSO-de) δ: 153.5, 152.6 , 151.5, 145.4, 128.2 (2C), 126.4, 126.0 (2C), 108.5, 105.0, 72.7, 48.6, 22.8; IR (neat, cm"1): 3088, 2710, 1585, 1476, 1304, 888, 695; HRMS (APCI/ASAP, m/z): 365.0263 (calcd.
C14H14N4I, 365.0263 [M+H]+).
(S)-2-((6-Iodo-7H-pyrrolo [2,3- \ pyrimidin-4-yl)amino)-2-phenylethan- l-ol ((S)-40)
The synthesis was performed as described above starting with compound 37 (730 mg, 5.30 mmol) and (5)-2-amino-2-phenylethanol (2.73 ml, 21.5 mmol). The reaction time was 24 hours. The crude
Figure imgf000065_0002
product was purified by silica-gel column chromatography (EtOAc/n-pentane, 4/1 , Rf = 0.12). Drying gave 560 mg (6.20 mmol, 55%) of (S)-40 as a pale yellow solid, mp 207 - 208 °C; purity: 98% (HPLC), tR = 17.1 min; [a]D 2° = -199.4 (c 0.80, DMSO); 1H NMR (400 MHz, DMSO-<¾) δ: 12.04 (s, 1H), 7.95 (s, 1H), 7.66 - 7.64 (m, 1H), 7.40 - 7.38 (m, 2H), 7.31 - 7.27 (m, 2H), 7.22 - 7.18 (m, 1H), 6.95 (s, 1H), 5.40 - 5.35 (m, 1H), 4.93 (t, J = 5.7, 1H), 3.75 - 3.66 (m, 2H); 13C NMR (100 MHz, OMSO-d6) δ: 154.1 , 152.6, 151.4, 141.8, 128.0 (2C), 127.0 (2C), 126.7, 108.6, 105.1 , 72.7, 64.9, 56.0; IR (neat, cm"1): 3253, 3083, 2777, 1581 , 1304, 1061 , 753, 700; HRMS (APCI/ASAP, m/z): 381.0215 (calcd. C14H14N4OI, 381.0212 [M+H]+).
(S)-2-(4-Fluorophenyl)-2-((6-iodo-7H-pyrrolo[2,3-d]pyrimidin-4- yl)amino)ethan-l-ol ((S)-41)
The synthesis was performed as described above starting with compound 37 (302 mg, 1.08 mmol) and (5)-2-amino-2-(4- fluorophenyl)ethan-l-ol (503 mg, 3.243 mmol). The reaction time was
18 hours. The crude product was purified by silica-gel column chromatography (EtOAc/n-pentane, 4/1 , Rf = 0.24). Drying gave 334
Figure imgf000066_0001
mg (0.840 mmol, 78%) of (S)-4l as a pale yellow solid, mp 203 - 206 °C; purity: 97% (HPLC), tR = 17.3 min; [a]D 2° = -267.4 (c 0.76, DMSO); 1H NMR (400 MHz, DMSO- d6) δ: 12.06 (s, 1H), 7.95 (s, 1H), 7.66 - 7.64 (m, 1H), 7.44 -7.41 (m, 2H), 7.14 - 7.09 (m, 2H), 6.93 (s, 1H), 5.39 - 5.34 (m, 1H), 4.95 (t, J = 5.6, 1H), 3.74 - 3.64 (m, 2H); 13C NMR (100 MHz, DMSO-d6) δ: 161.1 (d, J= 242.1), 153.9, 152.6, 151.4, 137.9 (d, J = 2.9), 128.9 (d, J = 8.0, 2C), 1 14.7 (d, J= 21.1 , 2C), 108.5, 105.1 , 72.9, 64.8, 55.2; 19F NMR (564 MHz, DMSO-d6) δ: -1 18.8 (s); IR (neat, cm" l): 331 1 , 3097, 2872, 1562, 1507, 1222, 830, 775; HRMS (APCI/ASAP, m/z):
399.01 17 (calcd. d4Hi3N4OFI, 399.0118 [M+H]+).
General procedure Suzuki coupling on 6-iodopyrrolo[2,3-i ]pyrimidines:
The following is representative: Compound (R)-39 (300 mg, 0.825 mmol) was mixed with (2-methoxyphenyl)boronic acid (150 mg, 0.989 mmol, 1.2 eq), fine powdered K2C03 (398 mg, 2.88 mmol, 3.5 eq), XPhos (20 mg, 4.00 μιηοΐ, 5 mol %>), 2nd generation XPhos precatalyst (34 mg, 4.00 μιηοΐ, 5 mol %>), 1 ,4-dioxane (2 ml) and water (2 ml). The reaction was then stirred at 100 °C for 1.5 - 18 hours under nitrogen atmosphere. The solvent was removed and the product was diluted with water (20 ml) and extracted with EtOAc (30 ml), the water phase was extracted with more EtOAc (3 x 30 ml). The combined organic phases were washed with brine (20 ml), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The crude mixture was purified as specified for each individual compound.
4-(4-(Benzylamino)-7H-pyrrolo[2,3-i ]pyrimidin-6-yl)-3-methoxybenzaldehyde (28q)
The synthesis was performed as described above starting with compound 38 (219 mg, 0.627 mmol) and (4-formyl-2- methoxyphenyl)boronic acid (135 mg, 0.752 mmol). The reaction time was 1 hour. The crude product was absorbed onto
Figure imgf000067_0001
Celite and purified by silica-gel column chromatography (diethyl ether/THF, 7/3, R/ = 0.23). Drying gave 195 mg (0.544 mmol, 87%) of product as a yellow solid, mp 230 - 232 °C (dec); purity: 98% (HPLC), tR = 27.5 min; 1H NMR (400 MHz, OMSO-de) δ: 1 1.94 (s, 1H), 9.99 (s, 1H), 8.19 (t, J= 6.0, 1H), 8.16 (s, 1H), 8.02 - 8.00 (m, 1H), 7.61 - 7.59 (m, 2H), 7.44 (s, 1H), 7.38 - 7.30 (m, 4H), 7.25 - 7.22 (m, 1H), 4.75 (d, J = 6.0, 2H), 4.03 (s, 3H); 13C NMR (100 MHz, DMSO-de) δ: 192.2, 156.3, 156.1 , 152.6, 151.2, 140.2, 135.4, 128.8, 128.3 (2C), 127.3 (2C), 127.1 , 126.7, 126.1 , 123.2, 1 10.9, 103.8, 102.9, 55.8, 43.1 ; IR (neat, cm"1): 3397, 31 17, 2920, 2842, 1676, 1588, 1262, 1 144, 733; HRMS
(APCI/ASAP, m/z): 359.1504 (calcd. C2iHi9N402, 359.1508, [M+H]+).
(4-(4-(Benzylamino)-7H-pyrrolo [2,3-d] pyrimidin-6-yl)-3- methoxyphenyl)methanol (28v)
Compound 28q (154 mg, 0.413 mmol) was dissolved in a mixture of THF (20 mL) and MeOH (10 ml), sodium borohydride (35.0 mg, 0.920 mmol) was added in 3 portions.
The mixture was stirred at 0 °C for 10 min and continued at 22
Figure imgf000067_0002
°C for 5 hours before being concentrated in vacuo. The reaction mixture was diluted with water (30 ml) and extracted with EtOAc (4 x 40 ml). The combined organic phases were washed with brine (30 ml), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The crude product was absorbed onto Celite and purified by silica gel column chromatography (THF/diehyl ether, 7/3, R/ = 0.22). Drying gave 117 mg (0.311 mmol, 75%) of product as a white solid, mp
236 - 238 °C; purity: 99% (HPLC), tR = 19.1 min; 1H NMR (400 MHz, DMSO-d6) δ: 11.65 (s, 1H), 8.10 (s, 1H), 7.99 (t, J= 6.0, 1H), 7.71 - 7.69 (m, 1H), 7.37 - 7.29 (m, 4H), 7.24 - 7.21 (m, 1H), 7.12 - 7.09 (m, 2H), 6.97 - 6.95 (m, 1H), 5.23 (t, J = 5.7, 1H), 4.73 (d, J= 6.0, 2H), 4.53 (d, J= 5.7, 2H), 3.91 (s, 3H); 13C NMR (100 MHz, DMSO-de) δ: 156.0, 155.6, 151.5, 150.6, 143.2, 140.5, 130.3, 128.2 (2C), 127.2 (2C), 126.9, 126.6, 118.7, 118.5, 109.8, 103.6, 99.4, 62.7, 55.4, 43.1; IR (neat, cm"1): 3413, 3226, 3138, 2925, 1597, 1350, 1072, 1031, 780, 729; HRMS
(APCI/ASAP, m/z): 361.1659 (calcd. C2iH2iN402, 361.1665, [M+H]+).
(R)-3-Methoxy-4-(4-((l-phenylethyl)amino)-7H-pyrrolo[2,3-i ]pyrimidin-6- yl)benzaldehyde ((R)-29q)
The synthesis was performed as described above starting with compound (i?)-39 (201 mg, 0.551 mmol) and (4-formyl-2- methoxyphenyl)boronic acid (121 mg, 0.672 mmol). The reaction time was 2 hours. The crude product was absorbed onto
Figure imgf000068_0001
Celite and purified by silica-gel column chromatography
(diethyl ether/MeOH, 19/1, R/= 0.21). Drying gave 150 mg (0.402 mmol, 73%) of product as a yellow solid, mp 224 - 226 °C; purity: 97% (HPLC), tR = 22.9 min; [a]D 20 = -408.6 (c 0.96, DMSO); 1H NMR (400 MHz, DMSC /6) δ: 11.91 (s, 1H), 9.99 (s, 1H), 8.10 (s, 1H), 8.02 - 7.97 (m, 2H), 7.61-7.60 (m, 2H), 7.50 (s, 1H), 7.44 - 7.43 (m, 2H), 7.33 - 7.60 (m, 2H), 7.22 - 7.18 (m, 1H), 5.57 - 5.50 (m, 1H), 4.05 (s, 3H), 1.55 (d, J= 7.0, 3H); 13C NMR (100 MHz, DMSC /6) δ: 192.2, 156.3, 155.3, 152.5, 151.3, 145.4, 135.4, 128.7, 128.2 (2C), 127.2, 126.5, 126.11, 126.07 (2C), 123.2, 110.9, 103.8, 103.1, 55.9, 48.6, 22.76; IR (neat, cm"1): 3382, 3216, 2966, 2920, 2852, 1683, 1590, 1247, 1148, 1032, 781, 698; HRMS (APCI/ASAP, m/z): 373.1658 (calcd. C22H2iN402, 373.1665, [M+H]+). (R)-(3-Methoxy-4-(4-((l-phenylethyl)amino)-7H-pyrrolo[2,3-d]pyrimidin-6- yl)phenyl)methanol ((R)-29v)
Compound (i?)-29q (126 mg, 0.338 mmol) was dissolved in a mixture of THF (20 mL) and MeOH (10 ml), sodium borohydride (25.8 mg, 0.682 mmol) was added in 3 portions.
The mixture was stirred at 0 °C for 10 minutes and continued
Figure imgf000069_0001
at 22 °C for 2 hours before being concentrated in vacuo. The reaction mixture was diluted with water (20 ml), and extracted with EtOAc (4 x 20 ml). The combined organic phases were washed with brine (30 ml), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The crude product was absorbed onto Celite and purified by silica-gel column chromatography (EtOAc, R = 0.10). Drying gave 100 mg (0.267 mmol, 80 %) of product as a white solid, mp
202 - 203 °C; purity: 98% (HPLC), tR = 20.0 min; [a]D = "339.2 (c 0.57, DMSO); 1H NMR (400 MHz, DMSC /6) δ: 11.61 (s, 1H), 8.03 (s, 1H), 7.79 - 7.77 (m, 1H), 7.70 - 7.68 (m, 1H), 7.44 - 7.42 (m, 2H), 7.32 - 7.29 (m, 2H), 7.21 - 7.18 (m, 2H), 7.10 (s, 1H), 6.98 - 6.96 (m, 1H), 5.55 - 5.48 (m, 1H), 5.24 (t, J= 5.7, 1H), 4.54 (d, J = 5.7, 2H), 3.93 (s, 3H), 1.54 (d, J= 7.0, 3H); 13C NMR (100 MHz, DMSC /6) δ: 156.0, 154.9, 151.5, 150.7, 145.6, 143.2, 130.2, 128.1 (2C), 126.9, 126.4, 126.1 (2C), 118.7, 118.5, 109.8, 103.5, 99.5, 62.7, 55.5, 48.7, 22.9; IR (neat, cm 1): 3418, 3262, 3107, 2971, 1604, 1316, 1159, 1034, 777; HRMS (APCI/ASAP, m/z):
375.1815 (calcd. C22H23N402, 375.1821, [M+H]+).
(5)-2-((6-(2-Methoxyphenyl)-7H-pyrrolo[2,3-i ]pyrimidin-4-yl)amino)-2- phenylethan-l-ol ((S)-31d)
Figure imgf000069_0002
gave 115 mg (0.319 mmol, 81%) of product as a white solid, mp 189 - 192 °C; purity: 97% (HPLC), tR = 19.9 min; [a]D 2° = -286.4 (c 0.73, DMSO); 1H NMR (400 MHz, DMSO-dg) δ: 1 1.65 (s, 1H), 8.05 (s, 1H), 7.77 - 7.75 (m, 1H), 7.71 - 7.69 (m, 1H), 7.45 - 7.43 (m, 2H), 7.32 - 7.27 (m, 3H), 7.24 - 7.19 (m, 2H), 7.15 - 7.13 (m, 1H), 7.05 - 7.01 (m, 1H), 5.47 - 5.41 (m, 1H), 4.95 (t, J = 5.7, 1H), 3.94 (s, 3H), 3.81 -3 .71 (m, 2H); 13C NMR (100 MHz, DMSO-d6) δ: 156.1 , 155.5, 151.5, 150.7, 142.0, 130.0, 128.3, 128.0 (2C), 127.2, 127.0 (2C), 126.6, 120.6, 120.3, 1 1 1.9, 103.7, 100.0, 65.0, 59.7, 55.5; IR (neat, cm 1): 3144, 1590, 746, 698; HRMS
(APCI/ASAP, m/z): 361.1665 (calcd. C21H21N4O2, 361.1665 [M+H]+).
(S)-2-((6-(4-(Hydroxymethyl)phenyl)-7H-pyrrolo[2,3-i |pyrimidin-4-yl)amino)- 2-phenylethan-l-ol ((5)-31m)
The synthesis was performed as described above starting with compound (S)-40 (150 mg, 0.395 mmol) and (4-
(hydroxymethyl)phenyl)boronic acid (72 mg, 1.474 mmol). The
Figure imgf000070_0001
reaction time was 3 hours. The crude product was purified by silica-gel column chromatography (EtOAc/MeOH, 19/1 , R = 0.21). Drying gave
129 mg (0.359 mmol, 91%) of product as a white solid, mp 267 °C (dec); purity: 97% (HPLC), t* = 15.9 min; [a]D 2° = -278.9 (c 0.50 DMSO); 1H NMR (400 MHz, DMSO-dg) δ: 1 1.99 (s, 1H), 8.05 (s, 1H), 7.76 - 7.74 (m, 2H), 7.71 - 7.68 (m, 1H), 7.45 - 7.42 (m, 2H), 7.38 - 7.37 (m, 2H), 7.32 - 7.29 (m, 2H), 7.23 - 7.19 (m, 1H), 7.1 1 (s, 1H), 5.45 - 5.39 (m, 1H), 5.20 (t, J= 5.7, 1H), 4.96 (t, J = 5.7, 1H), 4.52 (d, J = 5.7, 2H), 3.79 - 3.71 (m, 2H); 13C NMR (100 MHz, DMSO-d6) δ: 155.5, 151.6, 151.5, 142.0, 141.7, 133.5, 130.3, 128.0 (2C), 127.0 (4C), 126.6, 124.3 (2C), 104.0, 95.8, 65.0, 62.6, 56.0; IR (neat, cm"1): 3568, 3336, 31 15, 2932, 1595, 1051 , 767, 706; HRMS (APCI/ASAP, m/z): 361.1664 (calcd. C21H21N4O2, 361.1665 [M+H]+). (5)-4-(4-((2-Hydroxy-l-phenylethyl)amino)-7H-pyrrolo[2,3-i ]pyrimidin-6-yl)-
Figure imgf000070_0002
Drying gave 210 mg (0.542 mmol, 83%) of product as a yellow solid, mp 210 - 213 °C; purity: 97% (HPLC), tR = 18.9 min; [a]D 2° = -443.6 (c 0.48, DMSO); 1H NMR (400 MHz, DMSO-de) δ: 11.89 (s, 1H), 9.99 (s, 1H), 8.09 (s, 1H), 8.02 - 8.00 (m, 1H), 7.90 - 7.88 (m, 1H), 7.61 - 7.59 (m, 2H), 7.52 (s, 1H), 7.45 - 7.43 (m, 2H), 7.33 - 7.29 (m, 2H), 7.23 - 7.19 (m, 1H), 5.48 - 5.43 (m, 1H), 4.97 (t, J= 5.7, 1H), 4.06
(s, 3H), 3.82 - 3.71 (m, 2H); 13C NMR (100 MHz, DMSO-d6) δ: 192.1, 156.3, 155.9, 152.4, 151.3, 141.8, 135.4, 128.6, 128.0 (2C), 127.1, 127.0 (2C), 126.6, 126.1, 123.1, 110.9, 103.9, 103.1, 64.9, 56.0, 55.9; IR (neat, cm 1): 3399, 3147, 1677, 1667, 1592, 1567, 786, 697; HRMS (APCI/ASAP, m/z): 389.1614 (calcd. C22H21N4O3, 389.1614 [M+H]+).
(S)-2-((6-(4-(Hydroxymethyl)-2-methoxyphenyl)-7H-pyrrolo[2,3-i ]pyrimidin-4- yl)amino)-2-phenylethan-l-ol ((5)- 31v)
Compound (5)-31q (123.5 mg, 0.318 mmol) was dissolved in a mixture of THF (10 ml) and MeOH (5 ml), NaBH4 (36.1 mg,
0.96 mmol) was added. The mixture was stirred at 0 °C for 10 minutes and continued at 22 °C for 5 hours before being
Figure imgf000071_0001
concentrated in vacuo. The reaction mixture was diluted with water (30 ml) and the water phase was extracted with EtOAc (4 x 30 ml). The combined organic phases were washed with brine (30 ml), dried over anhydrous
Na2S04, filtered and concentrated in vacuo. The crude product was purified by silica-gel column chromatography (THF/diethyl ether, 4/1, R = 0.22). Drying gave 109 mg (0.28 mmol, 88%) of product as a white solid, mp 189 - 191 °C; purity: 99% (HPLC), t* = 16.42 min; [a]D 2° = -282.2 (c 0.99, DMSO); 1H NMR (400 MHz, DMSO-dg) δ: 11.60 (s, 1H), 8.03 (s, 1H), 7.71 - 7.66 (m, 2H), 7.45 - 7.43 (m, 2H), 7.32 - 7.28 (m, 2H), 7.22 - 7.19 (m, 2H), 7.10 (s, 1H), 6.98 - 6.96 (m, 1H), 5.46 - 5.41 (m, 1H), 5.23 (t, J= 5.7, 1H), 4.94 (t, J= 5.7, 1H), 4.54 (d, J= 5.8, 2H), 3.94 (s, 3H), 3.81 - 3.70 (m, 2H); 13C NMR (100 MHz, DMSC /6) δ: 156.0, 155.4, 151.4, 151.0, 143.2, 142.1, 130.2, 128.1 (2C), 127.0 (2C), 127.0, 126.9, 118.7, 118.5, 109.8, 103.7, 99.5, 65.1, 62.7, 56.0, 55.5; IR (neat, cm"1): 3555, 3366, 3157,3023, 1607, 1489, 1035, 813, 698; HRMS (APCI/ASAP, m/z): 391.1767 (calcd.
C22H23N403, 391.1700 [M+H]+). (S)-4-(4-((l-(4-Fluorophenyl)-2-hydroxyethyl)amino)-7H-pyrrolo[2,3- d]pyrimidin-6-yl)-3-methoxybenzaldehyde ((S)- 42q)
The synthesis was performed as described in above starting with compound ( )-41 (201.6 mg, 0.506 mmol) and (4-formyl- 2-methoxyphenyl)boronic acid (109 mg, 0.608 mmol). The reaction time was 3.5 hours. The crude product was purified by silica-gel column chromatography (THF/diethyl ether, 1/1, Rf
Figure imgf000072_0001
= 0.22). Drying gave 170 mg (0.419 mmol, 88%) of product as a white solid, mp 201 - 203 °C; purity: 98% (HPLC), tR = 19.39 min; [a]D 2° = -291.4 (c 0.49, DMSO); 1H NMR (400 MHz, DMSO-d6) δ: 11.90 (s, 1H), 9.99 (s, 1H), 8.09 (s, 1H), 8.02 - 8.00 (m, 1H), 7.90 - 7.88 (m, 1H), 7.61 - 7.59 (m, 2H), 7.50 - 7.46 (m, 3H), 7.16 - 7.11 (m, 2H), 5.47 -5 .42 (m, 1H), 4.98 (t, J= 5.6, 1H), 4.06 (s, 3H), 3.80 - 3.69 (m, 2H); 13C NMR (100 MHz, DMSO-^) S: 192.2, 161.1 (d, J= 242), 156.3, 155.8, 152.4, 139.2, 138.0 (d, J= 2.8), 135.4, 128.9 (d, J= 8.0, 2C), 128.7, 127.2, 126.1, 124.9, 123.2, 114.7 (d, J= 21.1, 2C), 110.9, 103.1, 64.7, 55.9, 55.3; 19F NMR (564 MHz, DMSO-dg) δ: -118.7 (s); IR (neat, cm"1); 3333, 3126, 2947, 1686, 1592, 1568, 1032, 782; HRMS (APCI/ASAP, m/z): 407.1518 (calcd. C22H20N4O3F, 407.1519 [M+H]+).
(S 2-(4-Fluorophenyl)-2-((6-(4-(hydroxymethyl)-2-methoxyphenyl)-7H- pyrrolo[2,3-i ]pyrimidin-4-yl)amino)ethan-l-ol ((S)-42v)
Compound (5)-42q (129 mg, 0.318 mmol) was dissolved in a mixture of THF (10 ml) and MeOH (5 ml), NaBH4 (36.0 mg, 0.96 mmol) was added. The mixture was stirred at 0 °C for 10 min and continued at 22 °C for 4 hours before being concentrated in vacuo. The reaction mixture was diluted
Figure imgf000072_0002
with water (30 ml) and EtOAc (30 ml) and the water phase was extracted with EtOAc (3 x 30 ml). The combined organic phases were washed with brine (30 ml), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The crude product was purified by silica-gel column chromatography (THF/diethyl ether, 3/1 , Rf = 0.24). Drying gave 113 mg (0.28 mmol, 88%) of product as a white solid, mp 189 - 192 °C; purity: 98% (HPLC), tR = 17.02 min; [a]D 2° = -275.4 (c 0.59, DMSO); 1H NMR (400 MHz, DMSO-d6) δ: 11.60 (s, 1H), 8.04 (s, 1H), 7.71 - 7.69 (m, 2H), 7.49 - 7.45 (m, 2H), 7.19 (s, 1H), 7.15 - 7.11 (m, 3H), 6.98 - 6.96 (m, 1H), 5.45 - 5.40 (m, 1H), 5.24 (t, J= 5.8, 1H), 4.97 (t, J= 5.7, 1H), 4.54 (d, J= 5.7, 2H), 3.94 (s, 3H), 3.80 - 3.68 (m, 2H); 13C NMR (100 MHz, DMSO-d6) δ: 161.1 (d, J= 242.2), 156.1, 156.0, 151.4, 150.6, 143.2, 138.3 (d, J= 2.9), 130.2, 128.9 (d, J= 7.9, 2C), 126.9, 118.7, 118.5, 114.7 (d, J= 21.1, 2C), 109.8, 103.7, 99.5, 64.8, 62.7, 59.7, 55.5; 19F NMR (564 MHz, DMSO-d6) δ: -118.6 (s) IR (neat, cm"1): 3301, 3153, 2916, 1596, 1034, 782; HRMS (APCI/ASAP, m/z): 409.1669 (calcd. C22H22N4O3F, 409.1676 [M+H]+).
Data for (R)-3-Methoxy-4-(4-((l-phenylethyl)amino)-7H-pyrrolo[2,3- d]pyrimidin-6-yl)benzamide (63b)
Figure imgf000073_0001
Ή NMR (400 MHz, DMSO- d6) δ: 11.76 (s, 1H), 8.07 (s, 1H), 8.03 (s, br, 1H), 7.89 (m, 1H), 7.87-7.82 (m, 1H), 7.61-7.60 (m, 1H), 7.57-7.54 (m, 1H), 7.44-7.36 (m, 4H), 7.33-7.29 (m, 2H), 7.22-7.18 (m, 1H), 5.54-5.49 (m, 1H), 4.00 (s, 3H), 1.55 (d, J=7.0, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 167.27, 155.78, 155.16, 152.03, 150.97, 145.53, 133.55, 129.19, 128.17 (2C), 126.55, 126.43, 126.08 (2C), 122.90, 119.83, 110.87, 103.65, 101.46, 55.74, 48.63, 22.83.
Data for (R)-6-(2,6-Dimethoxyphenyl)-N-(l-phenylethyl)-7H-pyrrolo[2,3- d]pyrimidin-4-amine (63c)
Figure imgf000074_0001
1H NMR (400 MHz, DMSO- d6) δ: U .25(s, 1H), 8.03 (s, 1H), 7 '.69-7.67 (m, 1H), 7.45-7.43 (m, 2H), 7.34-7.29 (m, 3H), 7.21-7.18 (m, 1H), 6.80-6.75 (m, 3H), 5.57- 5.50 (m, 1H), 3.77 (s, 6H), 1.54 (d, J=7.0, 3H); 13C NMR (100 MHz, DMSO-de) δ: 157.9 (2C), 154.7, 150.9, 149.8, 145.7, 129.4, 128.1 (2C), 126.4, 126.1 (2C), 125.9, 110.0, 104.3 (2C), 102.9, 100.6, 55.8 (2C), 48.5, 22.8.
Furopyrimidines
4-Methoxyphenyl-2-aminofurane-3-carbonitrile (441)
A solution of 2-bromo-(4-methoxyphenyl)ethanone (20.0 g, 87.31 mmol, 1.0 eq.) and malonitrile (7.50 g, 113.5 mmol, 1.3 eq.) was stirred at 0 °C in DMF (120 mL) for 10 min. Diethylamine (14.05 g, 19.86 mL, 192.1 mmol, 2.2 eq.) was added at 0 °C over 30 min and the mixture was stirred at rt for 4.5 h. The solution was poured onto water and left standing for 24 h at 4 °C. The precipitate formed was filtered off, washed with water (2 x 50 mL) and petroleum ether (50 mL) and dried in vacuo to give 7.0 g (32.69 mmol, 37%) of a brown solid. 1H NMR (400 MHz, DMSO-d6) <S:7.49 (s, 2 H), 7.42 (d, J= 8.8 Hz, 2 H), 6.95 (d, J= 8.8 Hz, 2H), 6.79 (s, 1 H), 3.76 (s, 3 H); 13C NMR (100 MHz, CDC13) δ: 164.2, 158.6, 142.5, 124.2 (2C), 122.8, 116.7, 114.8 (2C), 104.9, 66.3, 55.6 (OMe). IR (neat, cm ): 3413, 3327, 2205, 1646, 1508, 1245, 1026, 828.
6-(4-Methoxyphenyl)furano[2,3-i ]-pyrimidine-4-one (451)
Compound 32 (6.0 g, 28.01 mmol, 1.0 eq.) and formamide (63.08 g, 55.67 mL, 1.40 mol) were stirred in a solution of dimethylformamide (60 mL) and formic acid (24 mL) at 150 °C for 24 h. The reaction was cooled to rt and poured onto 2-propanol (200 mL). The mixture was left standing for 72 h at 4 °C. The precipitate was filtered, washed with 2-propanol (3 x70 mL) and n-pentane (50 mL) and dried in vacuo to give the compound 33 (2.01 g, 8.31 mmol, 30%)) as a brown solid. 1H NMR (400 MHz, DMSO-d6) δ: 12.61 (s. br, 1 H), 8.11 (s, 1 H), 7.79 (d, J= 8.7 Hz, 2 H), 7.31 (s, 1 H), 7.05 (d, J = 8.7 Hz, 2 H), 3.82 (s, 3 H); 13C NMR (100 MHz, CDC13) δ : 164.6, 160.2, 158.7, 152.0, 146.6, 126.3 (2C), 122.1, 115.0 (2C), 110.0, 99.7, 55.8; IR (neat, cm 1): 3100, 2838, 1674, 1253, 817, 780, 625.
4-Chloro-6-(4-methoxyphenyl)furano[2,3-i ]-pyrimidine (461)
Phosphoryl chloride (31.26 g, 19.0 mL, 203.8 mmol, 28.9 eq.) was slowly added to 6- (4- methoxyphenyl)furano[2,3-JJ-pyrimidine-4-one (451) (1.71 g, 7.05 mmol). The stirred solution was heated to 120 °C for 3.5 h. The reaction mixture was cooled down to rt and poured onto ice (800 mL). Under stirring a sodium hydroxide solution (250 mL, 5 M) was added to gain a pH value of 12. The mixture was filtered and washed with water (200 mL) to a pH of 7. Drying under reduced pressure gave the crude product as a brown solid. Silica-gel column chromatography (EtOAc) gave 1.65 g (6.34 mmol, 90%) of a yellowish solid. Mp. 173 - 174 °C; Rf = 0.69 (EtOAc); 1H NMR (400 MHz, DMSO-dg) δ: 8.78 (s, 1 H), 8.01 - 7.98 (m, 2 H), 7.59 (s, 1 H), 7.15 - 7.12 (m, 2 H), 3.86 (s, 3 H); 13C NMR (100 MHz, CDC13) δ: 166.6, 162.6, 156.9, 152.7, 151.2, 127.8 (2C), 120.7, 120.1, 115.3 (2C), 97.0, 55.9; IR (neat, cm"1): 2978, 2842, 1502, 1245, 835, 768.
(S)-2-((6-(4-methoxyphenyl)furo[2,3-d]pyrimidin-4-yl)amino)-2-phenyleth^ ((S)-50l)
4-Chloro-6-(4-methoxyphenyl)furo[2,3-d]pyrimidine (461) (300 mg, 1.151 mmol) and (S)-2-amino-2-phenylethan-l-ol (473.6 mg, 3.453 mmol) were added to 1- butanol (3 mL) under N2 atmosphere and heated at 145 °C for 12 h. After cooling to rt, 1-butanol was evaporated in vacuo, and water (30 mL) was added. The mixture was extracted with EtOAc (3x30 mL). The combined organic phases were washed with sat. NaCl solution (10 mL) and dried over anhydrous Na2S04, filtrated and evaporated. The crude product was absorbed onto silica and purified by silica-gel column chromatography (n-pentane/EtO Ac, 1/9). TLC (n-pentane/EtOAc, 1/9): R = 0.44. Isolation and drying gave 352.3 mg, (0.975 mmol, 83%) of (5)-35 as a white solid. HPLC purity (method A): 98%, tR = 22.8 min; 1H NMR (400 MHz, DMSO- d6) δ: 8.22-8.20 (m, 1H), 8.17 (s, 1H), 7.75-7.71 (m, 2H), 7.45-7.43 (m, 2H), 7.39 (m, 1H), 7.34-7.30 (m, 2H), 7.24-7.21 (m, 1H), 7.09-7.07 (m,2H), 5.44-5.38 (m, 1H), 5.03 (t, J= 5.6, 1H), 3.82 (s, 3H), 3.77-3.72 (m, 2H); 13C NMR (100 MHz, DMSO-de) δ; 166.1, 160.2, 157.1, 153.6, 151.1, 141.7, 128.6 (2C), 127.5 (2C), 127.3, 126.2 (2C), 122.4, 115.2 (2C), 103.3, 97.8, 65.3, 56.9, 55.8; HRMS
(APCI/ASAP, m/z): 362.1503 (calcd. C21H19N3O3, 362.1505, [M+H]+).
(S)-4-(4-( (2-Hydroxy-l-phenylethyl)amino)furo[2,3-d]pyrimidin-6-yl)phenol ( (S)- 50k)
(5)-2-((6-(4-Methoxyphenyl)furo[2,3-d]pyrimidin-4-yl)amino)-2-phenylethanol ((S)-501) (150 mg, 0.415 mmol) was dissolved in dry CH2C12 (6 mL) under N2 atmosphere. BBr3 (0.39 mL, 4.15 mmol) was added drop wise over 1 h at -78 °C. Then the mixture was allowed to react at 20 °C for 24 h. The reaction was quenched by addition of saturated NaHC03 solution (10 mL), and the mixture was extracted with EtOAc (3*25 mL). The combined organic phase was washed with brine (30 mL), dried over Na2S04 and concentrated. The resulting residue was purified by silica gel column chromatography (MeOH/Et20, 1/9). TLC (MeOH/Et20, 1/9),: R/ = 0.60. Drying gave 136 mg, (0.392 mmol, 94%) of (S)-37 as a yellowish solid; HPLC purity (method A): 96%, tR = 19.1 min.; 1H NMR (400 MHz, DMSO-d6) δ: 9.87 (s, 1H), 8.15 (s, 1H), 8.15 - 8.13 (m, 1H), 7.63 - 7.61 (m, 2H), 7.44 - 7.42 (m, 2H), 7.34 - 7.30 (m, 2H), 7.30 (s, 1H), 7.24 - 7.21 (m, 1H), 6.91 - 6.89 (m, 2H), 5.41 - 5.39 (m, 1H), 5.00 (s, 1H), 3.74 (m, 2H); 13C NMR (100 MHz, DMSC /6) δ: 165.5, 158.2, 156.6, 152.9, 151.0, 141.3, 128.1 (2C), 127.0 (2C), 126.8, 125.8 (2C), 120.3, 116.0 (3C), 96.4, 64.9, 56.5.
Other compounds of the invention can be prepared following protocols analogous to above. Analyses
1H and 13C NMR spectra were recorded with Bruker Avance 400 spectrometer, operating at 400 MHz and 100 MHz, respectively. HPLC (Agilent 110-Series) with a G1379A degasser, G1311A Quatpump, G1313A ALS autosampler and a G1315D Agilent detector (230 nm) was used to determine the purity of the synthesised compounds. The software used with the HPLC was Agilent ChemStation. Accurate mass determination (ESI) was performed on an Agilent G1969 TOF MS instrument equipped with a dual electrospray ion source, or EI (70eV) using a Finnigan MAT 95 XL. Accurate mass determination in positive and negative mode was performed on a "Synapt G2-S" Q-TOF instrument from Waters. FTIR spectra were recorded on a Thermo Nicolet Avatar 330 infrared spectrophotometer. All melting points are uncorrected and measured by a Buchi melting point instrument. In vitro EGFR (ErbBl) activity
The activity measurements of compounds towards ErbB 1 was performed by Invitrogen using their Z'-LYTE® assay technology.38 The test compounds were diluted in 1% DMSO solution, and subjected to a duplicate 10 points titration, using an ATP concentration equal to Km. The IC50 values were calculated from activity data with a four parameter logistic model using SigmaPlot (Windows Version 11.0 from Systat Software, Inc.)
Ba/F3 cell reporter cell analysis
Transfected Ba/F3 cells containing expression vectors for the L858R and T790M EGFR mutants were a kind gift from Dr. Nikolas von Bubnoff at the Technical University of Munich, Munich, Germany. The cells were cultured in RPMI 1640 (Gibco, Invitrogen) supplemented with 10% FCS (Gibco, Invitrogen), 1% L- glutamine (Gibco, Invitrogen) and 0.1% Gentamycin (Sanofi Aventis). Erlotinib was purchased from LC Laboratories (Woburn, MA). All inhibitors were reconstituted in DMSO, and appropriate stock solutions were prepared using cell culture medium. The final percentage concentrations of DMSO were < 0.2%. Proliferation analysis: Ba/F3 cells (1 x 104 per well) were plated into 96-well plates. Inhibitors were added in different concentrations as indicated. Cell growth was measured at 48 hours using TACS® XTT Cell Proliferation Assay (Trevigen) according to the manufacturer's instructions.
Human cancer cell lines A-431 and K-562
Cell proliferation assays were performed by ReactionBiology Corp. A-431 human epidermal carcinoma and K-562 chronic myelogenous leukemia cell lines were obtained from American Type Culture Collection (Manassas, VA). Staurosporine was obtained from Sigma-Aldrich (Saint Louis, MI). Cell Titer-Glo® Luminescent cell viability assay reagent was obtained from Promega (Madison, WI). A-431 and K-562 cells were grown in Dulbecco's Modified Eagle Medium (DMEM). Both cell culture mediums were supplemented with 10% heat-inactivated fetal bovine serum (FBS), 100 μg/ml penicillin, and 100 μg/ml of Streptomycin. Cultures were maintained at 37 °C in a humidified atmosphere of 5% C02 and 95% air. Compound, Erlotinib and Staurosporine (positive control) were all dissolved in DMSO in 10 mM stock. Culture medium (10 μΐ) was added to each well of 384 well cell culture plates. The compounds were diluted in a source plate in DMSO at 3 fold serial dilutions starting at 10 mM, total 10 doses. The compounds (0.25 μΐ) were delivered from source plate to each well of the cell culture plates by Echo 550. Then, 15 μΐ of culture medium containing 5000 cells of A-431 or K-562 were added to the wells of the cell culture plates. The cells were incubated with the compounds at 37 °C, 5% C02 for 72 hours. 25 μΐ of Cell Titer Glo reagent (25 μΐ) was added to each well according to the instruction of the kit. The contents were mixed on an orbital shaker for 2 minutes and incubated at room temperature for 10 minutes to stabilize luminescent signal. Luminescence was recorded by Envision 2104 Multilabel Reader (PerkinElmer, Santa Clara, CA). The maximum luminescence for each cell line in the absence of test compound, but in the presence of 0.4% DMSO, was similarly recorded after incubation for 72 hours. The number of viable cells in the culture was determined based on quantitation of the ATP present in each culture well. The percentage growth after 72 hours (%-growth) was calculated as follows: 100% x (luminescence t = 72 hours / luminescence untreated, t = 72 hours).
Human cancer cell line testing AU-565 and C-33A
Cell proliferation assays were performed by Netherlands Translational Research Center B.V (NTRC). All cell lines were licensed from the American Type Culture Collection (ATCC) Manassas, Virginia (US). Master and working cell banks (MCB and WCB) were prepared by subculturing in ATCC-recommended media and freezing according to ATCC recommended protocols (www.atcc.org). Cell line stocks for the assays were prepared from the WCB. The MCB, WCBs and assay stocks were prepared within respectively 3, 6 and 9 passages of the ATCC vial. Proliferation analysis: Compounds were weighed on a calibrated balance and dissolved in 100% DMSO to a concentration of 10 mM. The samples were stored at room temperature. The compound stock was diluted in 3.16 fold steps in 100% DMSO to obtain a 10-point dilution series. This was further diluted 31.6 times in 20 mM sterile Hepes buffer pH 7.4. A volume of 5 ml was transferred to the cells to generate a test concentration range from 3.16x 10"5 M to 3.16χ 10"9 M in duplicate. The final DMSO concentration during incubation was 0.4% in all wells. An assay stock was thawed and diluted in its ATCC recommended medium and dispensed in a 384-well plate, depending on the cell line used, at a concentration of 400 - 1600 cells per well in 45 μΐ medium. For each used cell line the optimal cell density was used. The margins of the plate were filled with phosphate-buffered saline. Plated cells were incubated in a humidified atmosphere of 5% C02 at 37 °C. After 24 hours, 5 μΐ of compound dilution was added and plates were further incubated for another 72 hours. After 72 hours, 25 μΐ of ATPlite IStep™ (PerkinElmer) solution was added to each well, and subsequently shaken for 2 minutes. After 10 minutes of incubation in the dark, the luminescence was recorded on an Envision multimode reader (PerkinElmer). The maximum luminescence for each cell line in the absence of test compound, but in the presence of 0.4% DMSO, was similarly recorded after incubation for 72 hours {luminescence untreated, t = 72 hours). The percentage growth after 72 hours (%-growth) was calculated as follows: 100% x (luminescence t = 72 hours / luminescence untreated, t = 72 hours). IC50 values were calculated from % growth and compound concentration with a four parameter logistic model using SigmaPlot (Windows Version 12.0 from Systat Software, Inc.)
Controls: t = 0 hours signal: On a parallel plate, 45 μΐ cells were dispensed and incubated in a humidified atmosphere of 5% C02 at 37 °C. After 24 hours 5 μΐ DMSO-containing Hepes buffer and 25 μΐ ATPlite IStep™ solution were mixed, and luminescence measured after 10 minutes incubation (= luminescence, t = 0 hours). Positive control: The IC50 of the reference compound doxorubicin was measured on a separate plate. The IC50 was trended. If the IC50 is out of specification (0.32 - 3.16 times deviating from historic average) the assay is invalidated. Cell growth control: The cellular doubling times of all cell lines were calculated from the t = 0 hour and t = 72 hours growth signals of the untreated cells. If the doubling time is out of specification (0.5 - 2.0 times deviating from historic average) the assay is invalidated.
MRC-5 Cell Titer Glo Lumiscent Cell Viability Assay Protocol
Materials:
The compounds and the reference staurosporine (Sigma- Aldrich , Saint Louis, MI) were all dissolved in DMSO in 10 mM stock. Cell Titer-Glo® Luminescent cell viability assay reagent was obtained from Promega (Madison, WI). MRC-5 human normal lung fibroblast cell line was obtained from American Type Culture
Collection (Manassas, VA). MRC-5 cells were grown in DMEM medium
supplemented with 10%> heat-inactivated fetal bovine serum (FBS), 100 μg/ml penicillin, and 100 μg/ml streptomycin. Cultures were maintained at 37°C in a humidified atmosphere of 5% C02 and 95% air. Procedure:
Culture medium (10 μΐ) were added to each well of 384 well cell culture plates. The compounds were diluted in a source plate in DMSO at 3 fold serial dilutions starting at 10 mM, total 10 doses. Then, 250 nl of the test compounds or 25 nl of
staurosporine were delivered from the source plate to each well of the cell culture plates by Echo 550. Culture medium (15 μΐ) containing 5000 cells of MRC-5 cells were added to the wells of the cell culture plates. The cells were incubated with the compounds at 37°C, 5% C02 for 72 h. Cell Titer Glo reagent (25 μΐ) were added to each well according to the instruction of the kit. The contents were mixed on an orbital shaker for 2 min and incubated at room temperature for 10 min to stabilize luminescent signal. Luminescence was recorded by Envision 2104 Multilabel Reader (PerkinElmer, Santa Clara, CA). The number of viable cells in culture were determined based on quantitation of the ATP present in each culture well. The IC50 values were calculated using the GraphPad Prism 4 program based on a sigmoidal dose-response equation. In most cases, the bottoms of the curves were constrained to 0.

Claims

Claims
1. A compound of formula (Γ) or (I)
Figure imgf000082_0001
wherein
X is NH, S or O,;
Ri is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR*, CHO, NR*2, COR*, C02R, CONR2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-CH=CH- CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR, CH2NR*2, SO3R or CH(OH)-
R*;
each R is independently H or Ci_6 alkyl; R2 is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR*, CHO, NR* 2, COR*, C02R, CONR2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-CH=CH- CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR, CH2NR* 2, SO3R or CH(OH)-
R*;
R3 is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR*, CHO, NR*2, COR*, C02R,
CONR2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-CH=CH- CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR, CH2NR*2, SO3R or CH(OH)-
R*;
R4 is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR*, CHO, NR*2, COR*, C02R, CONR2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-CH=CH- CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR, CH2NR*2, SO3R or CH(OH)-
R;
each n is independently 1 to 3;
R5 is H, Hal, Ci_6-alkyl, N02, NH2, NHCO-CH=CH-CH2-NR'2, or NHCO- CH=CH-R';
Re is H, Ci_6 alkyl, C6_io aryl, C7-12 arylalkyl, Hal, OR*, SR*, N02, NR"2;
NHCO-CH=CH-CH2-NR'2 NHCO-CH=CH-R', or C02R';
R7 is H, linear Ci_6alkyl, (CH2)nOR*, C02R, CONR2, CH2NR2, CH2Hal, CH2SR, (CH2)nC02R or (CH2)nCONR2;
R8 is H, Hal, Ci_6 alkyl, OCi_6 alkyl, OH, (CH2)nOR, or CF3;
R9 is H, Hal, Ci_6 alkyl, OCi_6 alkyl, OH, (CH2)nOR; or CF3
Rio is H, Hal, Ci_6 alkyl, or (CH2)nOR;
R11 is H, Hal, Ci_6 alkyl, OCi_6 alkyl, OH, (CH2)nOR, or CF3;
Ri2 is H, Hal, Ci_6 alkyl, OCi_6 alkyl, OH, (CH2)nOR, or CF3;
Ri3 is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR, CHO, NR*2, COR*,
C02R, CONR2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO- CH=CH-CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR*, CH2NR2, SO3R or CH(OH)-R';or a salt, ester, solvate, N-oxide, or prodrug thereof;
with the proviso that at least two of Ri- R4 and R13 are not H, preferably at least two of Ri- R4 are not H.
2. A compound of formula (II)
Figure imgf000084_0001
preferably of formula (ΙΓ)
Figure imgf000084_0002
wherein
X is NH, S or O;
Y is (CH2)nOR";
Ri is H, OH, OCi_6 alkyl, Hal, CN, d_6 alkyl, SR', CHO, NR'2, COR', C02R, CONR*2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-CH=CH- CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR*, CH2NR* 2, SO3R or CH(OH)-
R*;
each R is independently H or Ci_6 alkyl;
R2 is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR*, CHO, NR* 2, COR*, C02R, CONR2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-CH=CH- CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR, CH2NR* 2, SO3R or CH(OH)-
R;
R3 is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR*, CHO, NR* 2, COR*, C02R, CONR2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-CH=CH- CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR, CH2NR* 2, SO3R or CH(OH)-
R;
R4 is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR*, CHO, NR* 2, COR*, C02R, CONR2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO-CH=CH- CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR, CH2NR* 2, SO3R or CH(OH)- R;
each n is independently 1 to 3;
R5 is H, Hal, Ci_6-alkyl, N02, NH2, NHCO-CH=CH-CH2-NR'2, or NHCO- CH=CH-R';
Re is H, Ci_6 alkyl, C6_io aryl, C7-12 arylalkyl, Hal, OR*, SR*, N02, NR"2;
NHCO-CH=CH-CH2-NR'2, NHCO-CH=CH-R' , or C02R' ;
R8 is H, Hal, Ci_6 alkyl, OCi_6 alkyl, OH, (CH2)nOR, or CF3;
R9 is H, Hal, Ci_6 alkyl, OCi_6 alkyl, OH, (CH2)nOR; or CF3
Rio is H, Hal, Ci_6 alkyl, or (CH2)nOR;
R11 is H, Hal, Ci_6 alkyl, OCi_6 alkyl, OH, (CH2)nOR, or CF3;
Ri2 is H, Hal, Ci_6 alkyl, OCi_6 alkyl, OH, (CH2)nOR, or CF3;
Ri3 is H, OH, OCi_6 alkyl, Hal, CN, Ci_6 alkyl, SR, CHO, NR* 2, COR*, C02R, CONR2, NR'CONR*2, S02R, S02NR2, NHCO-CH=CH-R' or NHCO- CH=CH-CH2-NR'2, OCF3, OCF2H, CHF2, CH2F, (CH2)nOR*, CH2NR2, SO3R or CH(OH)-R; and
R" is H, or Ci_6 alkyl, preferably Me or ethyl;
or a salt thereof or a salt, ester, solvate, N-oxide, or prodrug thereof.
3. A compound as claimed in any preceding claim in which X is S or NH, especially NH.
4. A compound as claimed in any preceding claim at least two of R1-R4 are not H.
5. A compound as claimed in any preceding claim wherein Ri is H, OH, - (CH2)„OH, CHO, CH2OCi_6 alkyl or OCi_6 alkyl, more preferably Ri is H, OH or OCi_6 alkyl, especially OMe.
6. A compound as claimed in any preceding claim wherein R2 is H, OH, - (CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl.
7. A compound as claimed in any preceding claim wherein R3 is H, OH, F, CHO, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl.
8. A compound as claimed in any preceding claim wherein R4 is H, OH, CHO, Hal -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl. 9. A compound as claimed in any preceding claim wherein Ri3 is H or OMe.
10. A compound as claimed in any preceding claim wherein Rs_i2 are independently H, F or CI, most preferably H. 11. A compound as claimed in any preceding claim wherein R5 and R6 are H.
12. A compound as claimed in any preceding claim wherein R7 is linear Ci_6 alkyl, (CH2)nOCi_2 alkyl or (CH2)nOH, especially H, Me or CH2OH. 13. A compound as claimed in any preceding claim wherein the group Y is
(CH2)nOR", n is 1-3 and R" is H or Me, most preferably (CH2)OH or (CH2)OMe.
14. A compound as claimed in any preceding claim wherein n is 1 or 2, preferably 1.
15. A compound as claimed in any preceding claim wherein X=NH or S, at least one of R1-R4 is not H, Rs=H and Y=CH20R" where R" is as defined in claim 2, e.g. H or Me.
16. A compound as claimed in any preceding claim wherein X=NH, Rs=H and Y=CH20R" where R" is as defined in claim 2, e.g. H or Me.
A compound as claimed in any preceding claim of formula
Figure imgf000088_0001
wherein R1-R13, X, Y, n and R" are as defined in claim 2.
18. A compound as claimed in any preceding claim of formula (IV)

Figure imgf000089_0001
wherein Ri-R^ X and n are as defined in claim 1 ; with the proviso that in compounds of formula (IV), (IV) or (IV") at least two of Ri- R4 and R13 are not H; or a salt, ester, solvate, N-oxide, or prodrug thereof, in particular a salt thereof.
19. A compound as claimed in any preceding claim of formula (V)
Figure imgf000090_0001
wherein R1-R10 and n are as defined in claim 1,
with the proviso that at least two of Ri- R4 are not H; or of formula (VI)
Figure imgf000091_0001
wherein R1-R10, n and R" are as defined in claim 2, or a salt, ester, solvate, N-oxide, or prodrug thereof, in particular a salt thereof.
20. A compound as claimed in any preceding claim of formula (VIF) or
IIF)
Figure imgf000091_0002
(VIF) (VIIF)
wherein R1-R4 and n are as defined in claims 1 or 2, or a salt, ester, solvate, N-oxide, or prodrug thereof, in particular a salt thereof; with the proviso that in compounds of formula (VIF) at least two of Ri- R4 are not H.
21. A compound as claimed in any preceding claim of formula (IX') and (Χ')
Figure imgf000092_0001
(ΙΧ') (Χ')
wherein R1-R4 and n are as defined in claims 1 and 2; or a salt, ester, solvate, N- oxide, or prodrug thereof, in particular a salt thereof;
with the proviso that in compounds of formula (IX') at least two of Ri- R4 are not H.
22. A compound as claimed in any preceding claim wherein the phenyl ring 1 is represented by the group:
Figure imgf000092_0002
where Ri is H, OH, -(CH2)„OH, CHO, CH2OCi_6 alkyl or OCi_6 alkyl; and
R3 is H, OH, F, CHO, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl; and n is preferably 1 or 2. A compound as claimed in any preceding claim of formula (XI) or (XII)
Figure imgf000093_0001
Figure imgf000093_0002
(ΧΓ) (ΧΙΓ) wherein Ri is H, OH, -(CH2)„OH, CHO, CH2OCi_6 alkyl or OCi_6 alkyl; and R3 is H, OH, F, CHO, -(CH2)nOH, CH2OCi_6 alkyl or OCi_6 alkyl where for compound (XII) and (ΧΙΓ) Ri and R3 are not H; or a salt, ester, solvate, N-oxide, or prodrug thereof, in particular a salt thereof.
24. A compound of formula
Figure imgf000094_0001
or a salt, ester, solvate, N-oxide, or prodrug thereof, in particular a salt thereof.
25. A pharmaceutical composition comprising a compound as defined in any one of claims 1 to 24 and at least pharmaceutically acceptable carrier.
26. A compound as defined in any one of claims 1 to 24 for use in medicine.
27. A compound as defined in any one of claims 1 to 24 for use in the treatment of cancer or for the treatment of neuropathic pain.
28. Use of a compound as defined in any one of claims 1 to 24 for the manufacture of a medicament for the treatment of cancer or for the treatment of neuropathic pain.
29. A method of treating cancer or neuropathic pain in a patient comprising administering to a patient in need thereof, an effective amount of a compound as defined in claims 1 to 24.
PCT/EP2014/064067 2013-07-03 2014-07-02 4-amino-6-aryl[2,3-d]pyrimidines for the inhibition of egfr tyrosine kinase WO2015000959A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GB1311943.3 2013-07-03
GB201311943A GB201311943D0 (en) 2013-07-03 2013-07-03 Compounds
GB1403809.5 2014-03-04
GB201403809A GB201403809D0 (en) 2014-03-04 2014-03-04 Compounds

Publications (1)

Publication Number Publication Date
WO2015000959A1 true WO2015000959A1 (en) 2015-01-08

Family

ID=51178888

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2014/064067 WO2015000959A1 (en) 2013-07-03 2014-07-02 4-amino-6-aryl[2,3-d]pyrimidines for the inhibition of egfr tyrosine kinase

Country Status (1)

Country Link
WO (1) WO2015000959A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107226814A (en) * 2016-03-23 2017-10-03 罗欣生物科技(上海)有限公司 A kind of Ba Ruike replaces the preparation method of Buddhist nun's intermediate
WO2020142559A1 (en) * 2018-12-31 2020-07-09 Biomea Fusion, Llc Inhibitors of menin-mll interaction
US11084825B2 (en) 2018-12-31 2021-08-10 Biomea Fusion, Llc Substituted pyridines as irreversible inhibitors of menin-MLL interaction
CN113767100A (en) * 2019-03-01 2021-12-07 锐新医药公司 Bicyclic heteroaryl compounds and uses thereof
RU2803136C2 (en) * 2017-07-17 2023-09-07 Сейнт Луис Юниверсити THIENO[2,3-d]PYRIMIDINES AS ANTIMICROBIAL AGENTS
WO2024079342A1 (en) 2022-10-13 2024-04-18 Norwegian University Of Science And Technology (Ntnu) Compound active as inhibitor of colony stimulation factor-1 receptor (csf-1r)
WO2024079345A1 (en) 2022-10-13 2024-04-18 Norwegian University Of Science And Technology (Ntnu) Compound active as inhibitor of colony stimulation factor-1 receptor (csf-1 r)

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BUGGE STEFFEN ET AL: "Structure-activity study leading to identification of a highly active thienopyrimidine based EGFR inhib", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, vol. 75, 31 January 2014 (2014-01-31), pages 354 - 374, XP028625372, ISSN: 0223-5234, DOI: 10.1016/J.EJMECH.2014.01.042 *
STEFFEN BUGGE ET AL: "Route selection in the synthesis of C-4 and C-6 substituted thienopyrimidines", TETRAHEDRON, vol. 68, no. 45, 1 September 2012 (2012-09-01), pages 9226 - 9233, XP055141908, ISSN: 0040-4020, DOI: 10.1016/j.tet.2012.08.090 *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107226814A (en) * 2016-03-23 2017-10-03 罗欣生物科技(上海)有限公司 A kind of Ba Ruike replaces the preparation method of Buddhist nun's intermediate
RU2803136C2 (en) * 2017-07-17 2023-09-07 Сейнт Луис Юниверсити THIENO[2,3-d]PYRIMIDINES AS ANTIMICROBIAL AGENTS
WO2020142559A1 (en) * 2018-12-31 2020-07-09 Biomea Fusion, Llc Inhibitors of menin-mll interaction
US11084825B2 (en) 2018-12-31 2021-08-10 Biomea Fusion, Llc Substituted pyridines as irreversible inhibitors of menin-MLL interaction
US11174263B2 (en) 2018-12-31 2021-11-16 Biomea Fusion, Inc. Inhibitors of menin-MLL interaction
US11702421B2 (en) 2018-12-31 2023-07-18 Biomea Fusion, Llc Substituted pyridines as irreversible inhibitors of menin-MLL interaction
US11845753B2 (en) 2018-12-31 2023-12-19 Biomea Fusion, Inc. Inhibitors of menin-mll interaction
CN113767100A (en) * 2019-03-01 2021-12-07 锐新医药公司 Bicyclic heteroaryl compounds and uses thereof
WO2024079342A1 (en) 2022-10-13 2024-04-18 Norwegian University Of Science And Technology (Ntnu) Compound active as inhibitor of colony stimulation factor-1 receptor (csf-1r)
WO2024079345A1 (en) 2022-10-13 2024-04-18 Norwegian University Of Science And Technology (Ntnu) Compound active as inhibitor of colony stimulation factor-1 receptor (csf-1 r)

Similar Documents

Publication Publication Date Title
JP6698799B2 (en) Bicyclic heterocycle as FGFR inhibitor
JP6860267B2 (en) Azetidine derivatives, their preparation methods, and their use
CA2990089C (en) New aminoacid derivatives, a process for their preparation and pharmaceutical compositions containing them
RU2597609C2 (en) Substituted 4-(selenophen-2(or 3)-ylamino) pyrimidine compounds and methods of use thereof
CA2988896C (en) Fused-ring or tricyclic aryl pyrimidine compound used as kinase inhibitor
WO2015000959A1 (en) 4-amino-6-aryl[2,3-d]pyrimidines for the inhibition of egfr tyrosine kinase
TWI385174B (en) Fused bicyclic and tricyclic pyrimidine compounds as tyrosine kinase inhibitors
CN111635408B (en) Triazolo-pyrimidine compounds and uses thereof
JP2020519589A (en) Heteroaryl compounds that inhibit G12C mutant RAS protein
WO2018166493A1 (en) Heteroaryl[4,3-c]pyrimidine-5-amine derivative, preparation method therefor, and medical uses thereof
US20120309739A1 (en) Akt / pkb inhibitors
KR100963644B1 (en) Novel pyrazolopyrimidinone derivatives and preparation thereof
TW200407116A (en) Compounds
AU2015320142B2 (en) Novel imidazopyridazine compounds and their use
CN102227422A (en) Heterocyclic jak kinase inhibitors
AU2010267815B2 (en) Novel 1,2,3,4-tetrahydro-pyrimido(1,2-a)pyrimidin-6-one derivatives, preparation thereof, and pharmaceutical use thereof
EP3173413B1 (en) Pyrazolopyrimidinyl inhibitors of ubiquitin-activating enzyme
WO2013013614A1 (en) 4-(3-heteroarylarylamino)quinazoline and 1-(3-heteroarylarylamino)isoquinoline as hedgehog pathway inhibitor and use thereof
CN101679426A (en) 9-(pyrazol-3-yl)-9h-purine-2-amine and 3-(pyraz0l-3-yl)-3h-imidaz0[4,5-b]pyridin-5-amine derivatives and their use for the treatment of cancer
BRPI0609437A2 (en) compounds
CA2799926A1 (en) Heterocyclic compounds as janus kinase inhibitors
WO2019120276A1 (en) Pyrimidone compound and application thereof
WO2021239133A1 (en) Pyrimidine compound as axl inhibitor
TW202204351A (en) Compounds having a macrocyclic structure and uses thereof
CN108191837A (en) PI3K α/mTOR bidifly enzyme inhibitors and its pharmaceutical composition and application

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: 14739091

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: 14739091

Country of ref document: EP

Kind code of ref document: A1