WO2017182495A1 - Indazole derivatives that down-regulate the estrogen receptor and possess anti-cancer activity - Google Patents

Indazole derivatives that down-regulate the estrogen receptor and possess anti-cancer activity Download PDF

Info

Publication number
WO2017182495A1
WO2017182495A1 PCT/EP2017/059240 EP2017059240W WO2017182495A1 WO 2017182495 A1 WO2017182495 A1 WO 2017182495A1 EP 2017059240 W EP2017059240 W EP 2017059240W WO 2017182495 A1 WO2017182495 A1 WO 2017182495A1
Authority
WO
WIPO (PCT)
Prior art keywords
fluoro
methyl
compound
formula
pharmaceutically acceptable
Prior art date
Application number
PCT/EP2017/059240
Other languages
French (fr)
Inventor
Sebastien Louis Degorce
Thomas Andrew MOSS
James Stewart Scott
Bin Yang
Scott Gibson LAMONT
Original Assignee
Astrazeneca Ab
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Astrazeneca Ab filed Critical Astrazeneca Ab
Publication of WO2017182495A1 publication Critical patent/WO2017182495A1/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

Definitions

  • the specification relates to certain indazole compounds and pharmaceutically acceptable salts thereof that selectively down-regulate the estrogen receptor and possess anti-cancer activity.
  • the specification also relates to use of said indazole compounds and pharmaceutically acceptable salts thereof in methods of treatment of the human or animal body, for example in prevention or treatment of cancer.
  • the specification also relates to processes and intermediate compounds involved in the preparation of said indazole compounds and to pharmaceutical compositions containing them.
  • Estrogen receptor alpha (ERcc, ESR1, NR3A) and estrogen receptor beta (ER , ESR2, NR3b) are steroid hormone receptors which are members of the large nuclear receptor family. Structured similarly to all nuclear receptors, ERcc is composed of six functional domains (named A-F) (Dahlman- Wright, et al., Pharmacol.
  • Estrogen Receptor Elements The ERcc gene is located on 6q25.1 and encodes a 595AA protein and multiple isoforms can be produced due to alternative splicing and translational start sites.
  • the receptor In addition to the DNA binding domain (Domain C) and the ligand binding domain (Domain E) the receptor contains a N- terminal (A/B) domain, a hinge (D) domain that links the C and E domains and a C- terminal extension (F domain). While the C and E domains of ERcc and ER are quite conserved (96% and 55% amino acid identity respectively) conservation of the A/B, D and F domains is poor (below 30% amino acid identity). Both receptors are involved in the regulation and development of the female reproductive tract and in addition play roles in the central nervous system, cardiovascular system and in bone metabolism.
  • ERs The genomic action of ERs occurs in the nucleus of the cell when the receptor binds EREs directly (direct activation or classical pathway) or indirectly (indirect activation or non-classical pathway).
  • ERs are associated with heat shock proteins, Hsp90 and Hsp70, and the associated chaperone machinery stabilizes the ligand binding domain (LBD) making it accessible to ligand.
  • LBD ligand binding domain
  • Liganded ER dissociates from the heat shock proteins leading to a conformational change in the receptor that allows dimerisation, DNA binding, interaction with co-activators or co-repressors and modulation of target gene expression.
  • AP-1 and Sp-1 are alternative regulatory DNA sequences used by both isoforms of the receptor to modulate gene expression.
  • ER does not interact directly with DNA but through associations with other DNA bound transcription factors e.g. c-Jun or c-Fos (Kushner et al., Pure Applied Chemistry 2003, 75: 1757-1769).
  • c-Jun or c-Fos DNA bound transcription factors
  • the precise mechanism whereby ER affects gene transcription is poorly understood but appears to be mediated by numerous nuclear factors that are recruited by the DNA bound receptor.
  • the recruitment of co-regulators is primarily mediated by two protein surfaces, AF2 and AF1 which are located in E-domain and the A/B domain respectively.
  • AF1 is regulated by growth factors and its activity depends on the cellular and promoter environment whereas AF2 is entirely dependent on ligand binding for activity.
  • the two domains can act independently, maximal ER transcriptional activity is achieved through synergistic interactions via the two domains (Tzukerman, et al., Mol. Endocrinology, 1994, 8:21-30).
  • ERs are considered transcription factors they can also act through non-genomic mechanisms as evidenced by rapid ER effects in tissues following E2 administration in a timescale that is considered too fast for a genomic action.
  • phosphorylation of Serl 18 and an example of cross-talk between ER and growth factor signalling is the phosphorylation of Ser 118 by MAPK in response to growth factors such as IGF-1 and EGF (Kato, et al., Science, 1995, 270: 1491-1494).
  • SERMs selective estrogen receptor modulators
  • tamoxifen have the ability to act as both receptor agonists and antagonists depending on the cellular and promoter context as well as the ER isoform targeted.
  • tamoxifen acts as an antagonist in breast but acts as a partial agonist in bone, the cardiovascular system and uterus. All SERMs appear to act as AF2 antagonists and derive their partial agonist characteristics through AFl.
  • a second group fulvestrant being an example, are classified as full antagonists and are capable of blocking estrogen activity via the complete inhibition of AFl and AF2 domains through induction of a unique conformation change in the ligand binding domain (LBD) on compound binding which results in complete abrogation of the interaction between helix 12 and the remainder of the LBD, blocking co-factor recruitment (Wakeling, et al., Cancer Res., 1991, 51:3867-3873; Pike, et al., Structure, 2001, 9: 145-153).
  • LBD ligand binding domain
  • Intracellular levels of ERa are down-regulated in the presence of E2 through the ubiquitin/proteosome (Ub/26S) pathway.
  • Polyubiquitinylation of liganded ERa is catalysed by at least three enzymes; the ubiquitin- activating enzyme El activated ubiquitin is conjugated by E2 with lysine residues through an isopeptide bond by E3 ubiquitin ligase and polyubiquitinated ERa is then directed to the proteosome for degradation.
  • ER-dependent transcription regulation and proteosome-mediated degradation of ER are linked (Lonard, et al., Mol.
  • ER and/or progesterone receptors implying the hormone dependence of these tumour cells for growth.
  • Other cancers such as ovarian and endometrial are also thought to be dependent on ERa signalling for growth.
  • Therapies for such patients can inhibit ER signalling either by antagonising ligand binding to ER e.g. tamoxifen which is used to treat early and advanced ER positive breast cancer in both pre and post menopausal setting; antagonising and down-regulating ERa e.g. fulvestrant which is used to treat breast cancer in women which have progressed despite therapy with tamoxifen or aromatase inhibitors; or blocking estrogen synthesis e.g.
  • aromatase inhibitors which are used to treat early and advanced ER positive breast cancer. Although these therapies have had an enormously positive impact on breast cancer treatment, a considerable number of patients whose tumours express ER display de novo resistance to existing ER therapies or develop resistance to these therapies over time.
  • Several distinct mechanisms have been described to explain resistance to first-time tamoxifen therapy which mainly involve the switch from tamoxifen acting as an antagonist to an agonist, either through the lower affinity of certain co-factors binding to the tamoxifen-ERa complex being off-set by over-expression of these co-factors, or through the formation of secondary sites that facilitate the interaction of the tamoxifen-ERa complex with co-factors that normally do not bind to the complex.
  • Resistance could therefore arise as a result of the outgrowth of cells expressing specific co-factors that drive the tamoxifen-ERa activity.
  • growth factor signalling pathways directly activate the ER receptor or co-activators to drive cell proliferation independently of ligand signalling.
  • ESR1 mutations in ESR1 have been identified as a possible resistance mechanism in metastatic ER-positive patient derived tumour samples and patient-derived xenograft models (PDX) at frequencies varying from 17-25%. These mutations are predominantly, but not exclusively, in the ligand-binding domain leading to mutated functional proteins; examples of the amino acid changes include Ser463Pro, Val543Glu, Leu536Arg, Tyr537Ser, Tyr537Asn and Asp538Gly, with changes at amino acid 537 and 538 constituting the majority of the changes currently described. These mutations have been undetected previously in the genomes from primary breast samples characterised in the Cancer Genome Atlas database.
  • Fulvestrant is currently the only SERD approved for clinical use, yet despite its
  • the pharmacological properties of the drug have limited its efficacy due to the current limitation of a 500mg monthly dose which results in less than 50% turnover of the receptor in patient samples compared to the complete down-regulation of the receptor seen in in vitro breast cell line experiments (Wardell, et al., Biochem. Pharm., 2011, 82: 122-130).
  • ER targeting agents that have the required pharmaceutical properties and SERD mechanism to provide enhanced benefit in the early, metastatic and acquired resistance setting.
  • the compounds of the specification have been found to possess potent anti-tumour activity, being useful in inhibiting the uncontrolled cellular proliferation which arises from malignant disease.
  • the compounds of the specification provide an anti-tumour effect by, as a minimum, acting as SERDs.
  • R 1 is fluoro or methoxy
  • R 2 , R 3 and R 4 are each independently hydrogen or fluoro
  • R 5 is hydrogen or methyl
  • R 6 is hydrogen or methyl
  • R 7 is methyl, CHF 2 or cyclopropyl
  • R 8 is hydrogen or methyl
  • R 9 is hydrogen, fluoro, CH 2 OH, CH 2 OMe, CH 2 F or CHF 2 ;
  • R 10 is hydrogen, methyl or fluoro
  • R 11 is hydrogen, methyl, fluoro or CH 2 F;
  • R 10 and R 11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring.
  • the compounds of Formula (I) have two or more chiral centres and it will be recognised that the compound of Formula (I) may be prepared, isolated and/or supplied with or without the presence, in addition, of one or more of the other possible enantiomeric and/or diastereomeric isomers of the compound of Formula (I) in any relative proportions.
  • enantioenriched/ enantiopure and/or diastereoenriched/ diastereopure compounds may be carried out by standard techniques of organic chemistry that are well known in the art, for example by synthesis from enantioenriched or enantiopure starting materials, use of an appropriate enantioenriched or enantiopure catalyst during synthesis, and/or by resolution of a racemic or partially enriched mixture of stereoisomers, for example via chiral chromatography.
  • composition comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof, optionally together with one or more of the other stereoisomeric forms of the compound of Formula (I) or pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) or pharmaceutically acceptable salt thereof is present within the composition with a diastereomeric excess (%de) of > 90%.
  • the %de in the above-mentioned composition is > 95%.
  • the %de in the above-mentioned composition is > 98%.
  • the %de in the above-mentioned composition is > 99%.
  • composition comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof, optionally together with one or more of the other stereoisomeric forms of the compound of Formula (I) or
  • the %ee in the above-mentioned composition is > 95%.
  • the %ee in the above-mentioned composition is > 98%.
  • the %ee in the above-mentioned composition is > 99%.
  • composition comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof, optionally together with one or more of the other stereoisomeric forms of the compound of Formula (I) or
  • %ee enantiomeric excess
  • %de diastereomeric excess
  • a pharmaceutical composition which comprises a compound of the Formula (I) or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable diluent or carrier.
  • a pharmaceutical composition which comprises a compound of the Formula (I) or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable diluent or carrier, optionally further comprising one or more of the other stereoisomeric forms of the compound of Formula (I) or pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) or pharmaceutically acceptable salt thereof is present within the composition with an enantiomeric excess (%ee) of > 90%.
  • the %ee in the above-mentioned composition is > 95%.
  • the %ee in the above-mentioned composition is > 98%.
  • the %ee in the above-mentioned composition is > 99%.
  • a pharmaceutical composition which comprises a compound of the Formula (I) or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable diluent or carrier, optionally further comprising one or more of the other stereoisomeric forms of the compound of Formula (I) or pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) or pharmaceutically acceptable salt thereof is present within the composition with a diastereomeric excess (%de) of > 90%.
  • the %de in the above-mentioned composition is > 95%.
  • the %de in the above-mentioned composition is > 98%. In a further embodiment the %de in the above-mentioned composition is > 99%.
  • a pharmaceutical composition which comprises a compound of the Formula (I) or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable diluent or carrier, optionally further comprising one or more of the other stereoisomeric forms of the compound of Formula (I) or pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) or pharmaceutically acceptable salt thereof is present within the composition with an enantiomeric excess (%ee) of > 90% and a diastereomeric excess (%de) of > 90%.
  • %ee and %de may take any combination of values as listed below:
  • the compounds of Formula (I) and pharmaceutically acceptable salts thereof may be prepared, used or supplied in amorphous form, crystalline form, or semicrystalline form and any given compound of Formula (I) or pharmaceutically acceptable salt thereof may be capable of being formed into more than one crystalline / polymorphic form, including hydrated (e.g. hemi-hydrate, a mono-hydrate, a di-hydrate, a tri-hydrate or other stoichiometry of hydrate) and/or solvated forms. It is to be understood that the present specification encompasses any and all such solid forms of the compound of Formula (I) and pharmaceutically acceptable salts thereof.
  • R 1 is fluoro or methoxy
  • R 2 , R 3 and R 4 are each independently hydrogen or fluoro
  • R 5 is hydrogen or methyl
  • R 6 is hydrogen or methyl
  • R 7 is methyl, CHF 2 or cyclopropyl
  • R 8 is hydrogen or methyl
  • R 9 is hydrogen, fluoro, CH 2 OH, CH 2 OMe, CH 2 F or CHF 2 ;
  • R 10 is hydrogen, methyl or fluoro
  • R 11 is hydrogen, methyl, fluoro or CH 2 F;
  • R 10 and R 11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring;
  • R 1 is methoxy
  • R 1 is fluoro
  • R 4 is fluoro
  • R 4 is hydrogen
  • R 1 is methoxy and R 4 is fluoro.
  • R 1 is methoxy and R 4 is hydrogen. In one embodiment R 1 is fluoro and R 4 is fluoro.
  • R 2 is hydrogen
  • R 2 is fluoro
  • R 3 is hydrogen
  • R 3 is fluoro
  • R 2 is hydrogen and R 3 is hydrogen.
  • R 1 is methoxy and R 2 is fluoro.
  • R 1 is methoxy and R 3 is fluoro.
  • R 1 is methoxy
  • R 2 is hydrogen
  • R 3 is hydrogen
  • R 4 is fluoro
  • R 1 is methoxy and R 2 , R 3 and R 4 are hydrogen.
  • R 1 is fluoro
  • R 2 is hydrogen
  • R 3 is hydrogen
  • R 4 is fluoro
  • R 5 is hydrogen
  • R 5 is methyl
  • R 6 is hydrogen
  • R 6 is methyl
  • R 7 is methyl or CHF 2 .
  • R 7 is methyl
  • R 6 is hydrogen and R 7 is methyl.
  • R 6 is hydrogen and R 7 is CHF 2 .
  • R 6 is methyl and R 7 is methyl.
  • R 8 is hydrogen
  • R 9 is fluoro
  • R 9 is CH 2 OH, CH 2 OMe, CH 2 F or CHF 2 and R 10 is methyl.
  • R 9 is CH 2 OH, CH 2 OMe, CH 2 F or CHF 2 and R 10 is fluoro.
  • R 9 is CH 2 OH, CH 2 OMe, CH 2 F or CHF 2 and R 10 and R 11 taken together with the carbon atom to which they are attached form a cyclopropyl ring.
  • the group -CH(R 8 )-C(R 9 )(R 10 )(R n ) in the compound of Formula (I) is selected from the group consisting of:
  • the group -CH(R 8 )-C(R 9 )(R 10 )(R n ) in the compound of Formula (I) is selected from the group consisting of:
  • the group -CH(R 8 )-C(R 9 )(R 10 )(R n ) in the compound of Formula (I) is selected from the group consisting of:
  • the group -CH(R 8 )-C(R 9 )(R 10 )(R n ) in the compound of Formula (I) is selected from the group consisting of:
  • R 1 is methoxy
  • R 2 and R 3 are hydrogen
  • R 4 is hydrogen or fluoro
  • R 5 is hydrogen or methyl
  • R 6 is hydrogen or methyl
  • R 7 is methyl, CHF 2 or cyclopropyl
  • R 8 is hydrogen or methyl
  • R 9 is hydrogen, fluoro, CH 2 OH, CH 2 OMe, CH 2 F or CHF 2 ;
  • R 10 is hydrogen, methyl or fluoro
  • R 11 is hydrogen, methyl, fluoro or CH 2 F; or R 10 and R 11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring.
  • R 1 is methoxy
  • R 2 and R 3 are hydrogen
  • R 4 is hydrogen or fluoro
  • R 5 is hydrogen or methyl
  • R 6 is hydrogen or methyl
  • R 7 is methyl, CHF 2 or cyclopropyl
  • R 8 is hydrogen or methyl
  • R 9 is hydrogen, fluoro, CH 2 OH, CH 2 OMe, CH 2 F or CHF 2 ;
  • R 10 is hydrogen, methyl or fluoro
  • R 11 is hydrogen, methyl, fluoro or CH 2 F;
  • R 10 and R 11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring;
  • R 1 is methoxy
  • R 2 , R 3 and R 4 are hydrogen
  • R 5 is hydrogen or methyl
  • R 6 is hydrogen or methyl
  • R 7 is methyl, CHF 2 or cyclopropyl
  • R 8 is hydrogen or methyl
  • R 9 is hydrogen, fluoro, CH 2 OH, CH 2 OMe, CH 2 F or CHF 2 ;
  • R 10 is hydrogen, methyl or fluoro
  • R 11 is hydrogen, methyl, fluoro or CH 2 F;
  • R 10 and R 11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring.
  • R 1 is methoxy
  • R 2 , R 3 and R 4 are hydrogen
  • R 5 is hydrogen or methyl
  • R 6 is hydrogen or methyl
  • R 7 is methyl, CHF 2 or cyclopropyl
  • R 8 is hydrogen or methyl
  • R 9 is hydrogen, fluoro, CH 2 OH, CH 2 OMe, CH 2 F or CHF 2 ;
  • R 10 is hydrogen, methyl or fluoro
  • R 11 is hydrogen, methyl, fluoro or CH 2 F;
  • R 10 and R 11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring;
  • R 1 is methoxy
  • R 2 and R 3 are hydrogen
  • R 4 is hydrogen or fluoro
  • R 5 is methyl
  • R 6 is hydrogen or methyl
  • R 7 is methyl, CHF 2 or cyclopropyl
  • R 8 is hydrogen or methyl
  • R 9 is hydrogen, fluoro, CH 2 OH, CH 2 OMe, CH 2 F or CHF 2 ;
  • R 10 is hydrogen, methyl or fluoro
  • R 11 is hydrogen, methyl, fluoro or CH 2 F;
  • R 10 and R 11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring.
  • R 1 is fluoro
  • R 2 and R 3 are hydrogen
  • R 4 is fluoro
  • R 5 is hydrogen or methyl
  • R 6 is hydrogen or methyl
  • R 7 is methyl, CHF 2 or cyclopropyl
  • R 8 is hydrogen or methyl
  • R 9 is hydrogen, fluoro, CH 2 OH, CH 2 OMe, CH 2 F or CHF 2 ;
  • R 10 is hydrogen, methyl or fluoro
  • R 11 is hydrogen, methyl, fluoro or CH 2 F;
  • R 10 and R 11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring;
  • R 5 is hydrogen or methyl
  • R 6 is hydrogen or methyl
  • R 7 is methyl, CHF 2 or cyclopropyl
  • R 8 is hydrogen or methyl
  • R 9 is hydrogen, fluoro, CH 2 OH, CH 2 OMe, CH 2 F or CHF 2 ;
  • R 10 is hydrogen, methyl or fluoro
  • R 11 is hydrogen, methyl, fluoro or CH 2 F;
  • R 10 and R 11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring;
  • Ring Y is selected from:
  • R 5 is hydrogen or methyl
  • R 6 is hydrogen or methyl
  • R 7 is methyl, CHF 2 or cyclopropyl
  • R 8 is hydrogen or methyl
  • R 9 is hydrogen, fluoro, CH 2 OH, CH 2 OMe, CH 2 F or CHF 2 ;
  • R 10 is hydrogen, methyl or fluoro
  • R 11 is hydrogen, methyl, fluoro or CH 2 F;
  • R 10 and R 11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring.
  • Ring Y is selected from: ⁇
  • R 5 is hydrogen or methyl
  • R 6 is hydrogen or methyl
  • R 7 is methyl, CHF 2 or cyclopropyl; R is hydrogen or methyl;
  • R 9 is hydrogen, f uoro, CH 2 OH, CH 2 OMe, CH 2 F or CHF 2 ;
  • R 10 is hydrogen, methyl or fluoro
  • R 11 is hydrogen, methyl, fluoro or CH 2 F;
  • R 10 and R 11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring;
  • g Y in the compound of Formula (IA) is selected from:
  • R 5 is hydrogen or methyl
  • R 6 is hydrogen or methyl
  • R 7 is methyl, CHF 2 or cyclopropyl
  • R 12 is selected from the group consisting of
  • R 5 is hydrogen or methyl
  • R 6 is hydrogen and R 7 is CHF 2 or cyclopropyl
  • R 6 is methyl and R 7 is methyl
  • R 12 is selected from the group consisting of:
  • R 5 is hydrogen or methyl
  • R 6 is hydrogen or methyl
  • R 7 is methyl, CHF 2 or cyclopropyl
  • R 4 is hydrogen or fluoro
  • R 5 is hydrogen or methyl
  • R 6 is hydrogen or methyl
  • R 7 is methyl, CHF 2 or cyclopropyl
  • R 12 is selected from the group consisting of:
  • R 12 is not:
  • R 4 is hydrogen or fluoro
  • R 5 is hydrogen or methyl
  • R 6 is hydrogen or methyl
  • R 7 is methyl, CHF 2 or cyclopropyl
  • R 12 is selected from the group consisting of:
  • R 4 is hydrogen or fluoro
  • R 5 is hydrogen or methyl
  • R 6 is hydrogen
  • R 7 is methyl
  • R 12 is selected from the group consisting of:
  • R 4 is hydrogen or fluoro
  • R 6 is hydrogen or methyl
  • R 7 is methyl, CHF 2 or cyclopropyl
  • R 12 is selected from the group consisting of:
  • isotopes of hydrogen include tritium and deuterium.
  • isotopes of carbon include 13 C and 14 C.
  • R 5 is deuterium.
  • a suitable pharmaceutically acceptable salt of a compound of the Formula (I) is, for example, an acid addition salt or a base addition salt.
  • a base addition salt may be formed using a metal salt, for example an alkali or alkaline earth metal salt such as a sodium, potassium, lithium, calcium or magnesium salt, or an ammonium salt, or a salt with an organic base such as methylamine, dimethylamine, trimethylamine, piperidine, morpholine or tris-(2-hydroxyethyl)amine.
  • a metal salt for example an alkali or alkaline earth metal salt such as a sodium, potassium, lithium, calcium or magnesium salt, or an ammonium salt, or a salt with an organic base such as methylamine, dimethylamine, trimethylamine, piperidine, morpholine or tris-(2-hydroxyethyl)amine.
  • suitable pharmaceutically acceptable salts of a compound of the Formula (I) may be other metal salts, such as zinc, or other such metal cations known in the art.
  • Suitable pharmaceutically acceptable salts of a compound of the Formula (I) may be salts formed with a base such as one of the following: calcium acetate, diethylamine, ethanolamine, ethylenediamine, hydroxyethylpyrrolidine, magnesium acetate, meglumine, piperazine, potassium hydroxide, sodium hydroxide, t- butylamine, triethanolamine, tris(hydroxymethyl)-aminomethane (Tris) or N,N- diethylethanolamine .
  • a base such as one of the following: calcium acetate, diethylamine, ethanolamine, ethylenediamine, hydroxyethylpyrrolidine, magnesium acetate, meglumine, piperazine, potassium hydroxide, sodium hydroxide, t- butylamine, triethanolamine, tris(hydroxymethyl)-aminomethane (Tris) or N,N- diethylethanolamine .
  • a pharmaceutically acceptable salt of a compound of Formula (I) which is a salt comprising a metal cation, an ammonium cation or a salt formed with an organic base.
  • a further suitable pharmaceutically acceptable salt of a compound of the Formula (I) is, for example, a salt formed within the human or animal body after administration of a compound of the Formula (I) to said human or animal body.
  • a suitable pharmaceutically acceptable salt of a compound of the Formula (I) may also be, for example, an acid-addition salt of a compound of the Formula (I), for example an acid-addition salt with an inorganic or organic acid such as hydrochloric acid, hydrobromic acid, sulphuric acid or trifluoroacetic acid.
  • an acid-addition salt of a compound of the Formula (I) for example an acid-addition salt with an inorganic or organic acid such as hydrochloric acid, hydrobromic acid, sulphuric acid or trifluoroacetic acid.
  • Pharmaceutically acceptable salts of a compound of the Formula (I) may also be an acid-addition salt with an acid such as one of the following: acetic acid, adipic acid, benzene sulfonic acid, benzoic acid, cinnamic acid, citric acid, D,L-lactic acid, ethane disulfonic acid, ethane sulfonic acid, fumaric acid, L-tartaric acid, maleic acid, malic acid, malonic acid, methane sulfonic acid, napadisylic acid, phosphoric acid, saccharin, succinic acid, /7-toluenesulfonic acid or toluene sulfonic acid.
  • an acid such as one of the following: acetic acid, adipic acid, benzene sulfonic acid, benzoic acid, cinnamic acid, citric acid, D,L-lactic acid, ethane disulfonic acid, ethan
  • the compound of Formula (I) or pharmaceutically acceptable salt thereof may be prepared as a co-crystal solid form. It is to be understood that a pharmaceutically acceptable co-crystal of a compound of the Formula (I) or pharmaceutically acceptable salts thereof, form an aspect of the present specification.
  • the compounds of the specification may be administered in the form of a prodrug, which is a compound that is broken down in the human or animal body to release a compound of the specification.
  • a pro-drug may be used to alter the physical properties and/or the pharmacokinetic properties of a compound of the specification.
  • a pro-drug can be formed when the compound of the specification contains a suitable group or substituent to which a property-modifying group can be attached. Examples of pro-drugs include in- vivo cleavable ester or amide derivatives that may be formed at the carboxy group in a compound of the Formula (I).
  • one aspect of the present specification includes those compounds of Formula (I) as defined hereinbefore when made available by organic synthesis and when made available within the human or animal body by way of cleavage of a pro-drug thereof. Accordingly, the present specification includes those compounds of the Formula (I) that are produced by organic synthetic means and also such compounds that are produced in the human or animal body by way of metabolism of a precursor compound, that is a compound of the Formula (I) may be a synthetically-produced compound or a metabolically- produced compound.
  • a suitable pharmaceutically acceptable pro-drug of a compound of the Formula (I) is one that is based on reasonable medical judgement as being suitable for administration to the human or animal body without undesirable pharmacological activities and without undue toxicity.
  • pro-drug Various forms of pro-drug have been described, for example in the following documents :- a) Methods in Enzymology, Vol. 42, p. 309-396, edited by K. Widder, et al.
  • a suitable pharmaceutically acceptable pro-drug of a compound of the Formula (I) that possesses a carboxy group is, for example, an in-vivo cleavable ester thereof.
  • An in- vivo cleavable ester of a compound of the Formula (I) containing a carboxy group is, for example, a pharmaceutically acceptable ester which is cleaved in the human or animal body to produce the parent acid.
  • Suitable pharmaceutically acceptable esters for a carboxy group include (l-6C)alkyl esters such as methyl, ethyl and ie/t-butyl, (l-6C)alkoxymethyl esters such as methoxymethyl esters, (l-6C)alkanoyloxymethyl esters such as
  • pivaloyloxymethyl esters 3-phthalidyl esters, (3-8C)cycloalkylcarbonyloxy-(l-6C)alkyl esters such as cyclopentylcarbonyloxymethyl and 1-cyclohexylcarbonyloxyethyl esters, 2- oxo-l,3-dioxolenylmethyl esters such as 5-methyl-2-oxo-l,3-dioxolen-4-ylmethyl esters and (l-6C)alkoxycarbonyloxy-(l-6C)alkyl esters such as methoxycarbonyloxymethyl and 1 -methoxycarbonyloxyethyl esters .
  • a suitable pharmaceutically acceptable pro-drug of a compound of the Formula (I) which have a carboxy group is for example an in-vivo cleavable amide such as a N-Ci-e alkyl and N,N-di-(Ci-6alkyl)amide such as N-methyl, N-ethyl, N-propyl, N,N-dimethyl, N- ethyl-N-methyl or N,N-diethylamide.
  • the in-vivo effects of a compound of the Formula (I) may be exerted in part by one or more metabolites that are formed within the human or animal body after administration of a compound of the Formula (I). As stated hereinbefore, the in-vivo effects of a compound of the Formula (I) may also be exerted by way of metabolism of a precursor compound (a pro-drug).
  • Another aspect of the present specification provides a process for preparing a compound of the Formula (I), or a pharmaceutically acceptable salt thereof.
  • Necessary starting materials may be obtained by standard procedures of organic chemistry. The preparation of such starting materials is described in conjunction with the following representative process variants and within the accompanying Examples. Alternatively, necessary starting materials are obtainable by analogous procedures to those illustrated which are within the ordinary skill of an organic chemist.
  • Compounds of Formula (I) may be prepared by hydrolysis of a compound of Formula (II), wherein R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 and R 11 are as defined in the compound of Formula (I) and R 12 is (1-6C) alkyl. Hydrolysis is conveniently carried out in the presence of a suitable base (such as aqueous sodium hydroxide) or acid (such as hydrochloric acid) in a suitable solvent (such as THF/methanol or DCM) and at a suitable temperature, for example at room temperature.
  • a suitable base such as aqueous sodium hydroxide
  • acid such as hydrochloric acid
  • a suitable solvent such as THF/methanol or DCM
  • (1-6C) alkyl groups containing the requisite number of carbon atoms can be branched or unbranched.
  • suitable (1-6C) alkyl groups include methyl, ethyl, n-propyl, i-propyl, n-butyl, sec -butyl, t-butyl, n-pentyl, i-pentyl, neo- pentyl, n-hexyl and i-hexyl, such as methyl and t-butyl.
  • a compound of Formula (II), or a salt thereof is a useful intermediate towards the preparation of the compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • Compounds of formula (II) may be made by, for example: a Heck type coupling reaction of a compound of formula (III), where L is for example a halogen (such as Br), a trifluoromethanesulfonyl (triflate) group, a boronic acid or a boronate ester and P is optionally an appropriate protecting group (for example THP or Boc that may be subsequently removed by treatment with acid), with an acrylate ester of formula (IV), where R 12 is (1-6C) alkyl, using a suitable metal catalyst (for example Pd- 118) in a suitable solvent (for example 1,4-dioxane) in the presence of a suitable base (for example diisopropylethylamine) and at a suitable temperature (such as 100- 130°C).
  • L is for example a halogen (such as Br), a trifluoromethanesulfonyl (triflate) group, a boronic acid or a boron
  • Compounds of formula (III) where R 5 is methyl may be made from, for example, compounds of formula (V) by oxidation with a suitable reagent (for example
  • Compounds of formula (V) may be prepared by, for example, reaction of an aniline of Formula (VI) with suitable reagents to effect the construction of an indazole such as inorganic nitrite (such as sodium nitrite) in organic acid (such as propionic acid) optionally in the presence of water, at low temperature (typically -20 to 0 °C); or alternatively an acid anhydride (such as acetic anhydride) in the presence of a suitable base (such as potassium acetate) together with organic nitrite (such as isopentyl nitrite) optionally in the presence of a crown ether (such as 18-crown-6) in a suitable solvent (such as chloroform) at a suitable temperature (such as 70 °C).
  • an indazole such as inorganic nitrite (such as sodium nitrite) in organic acid (such as propionic acid) optionally in the presence of water, at low temperature (typically -20 to 0 °C); or
  • L is for example a halogen (such as Br) or a trifluoromethanesulfonyl (triflate) group or a boronic acid or a boronate ester or alternatively an acrylate ester.
  • halogen such as Br
  • triflate trifluoromethanesulfonyl
  • Compounds of formula (VI) may be made by reaction of a compound of formula (VII) with a compound of formula (VIII) under conditions known in the art as suitable for Pictet-Spengler reactions, such as in the presence of acid (such as acetic acid) and in a suitable solvent (for example toluene or water) and a suitable temperature (such as 60- 100°C).
  • acid such as acetic acid
  • solvent for example toluene or water
  • a suitable temperature such as 60- 100°C
  • L is for example a halogen (such as Br) or a trifluoromethanesulfonyl (triflate) group or a boronic acid or a boronate ester or alternatively an acrylate ester.
  • Compounds of formula (VII) may be prepared by functional group interconversions known to those skilled in the art of organic synthesis, for example by the amination of aryl halides, such as the aryl bromide of formula (IX), using a protected amine (such as diphenylmethanimine) in the presence of a suitable catalyst and ligand (such as bis(dibenzylideneacetone)palladium(0) and rac-2,2'-bis(diphenylphosphino)- 1,1'- binaphthyl), in the presence of a suitable base (such as sodium iert-butoxide), in a suitable solvent (such as toluene) and at a suitable temperature (such as 80-100°C); followed by deprotection of the protected amine using, for example, acid (such as hydrochloric acid) in a suitable solvent (such as DCM) at a suitable temperature (such as ambient temperature).
  • a protected amine such as diphenylmethani
  • a suitable solvent for example THF
  • a suitable reducing agent such as sodium triacetoxyborohydride
  • LG is a suitable leaving group (for example a halogen atom (such as bromo or chloro) or trifluoromethanesulfone), in the presence of a suitable base (such as
  • diisopropylethylamine in a suitable solvent (for example DCM or dioxane) and at a suitable temperature (such as 20-85 °C).
  • a suitable solvent for example DCM or dioxane
  • a suitable temperature such as 20-85 °C
  • Phase transfer alkylation in the presence of a chiral catalyst for example using (15,25,45,5R)-2-((R)-(allyloxy)(quinolin-4-yl)methyl)- l-(anthracen-9-ylmethyl)-5- vinylquinuclidin-l-ium bromide in toluene/KOH at 0°C, followed by functional group manipulation:
  • compounds of formula (V) may be prepared by alkylation of compounds of formula (XV) with a compound of formula (XVI), wherein LG is a suitable leaving group (for example a halogen atom (such as bromo or chloro) or
  • diisopropylethylamine in a suitable solvent (for example DCM or 1,4-dioxane) and at a suitable temperature (such as 20-85 °C).
  • a suitable solvent for example DCM or 1,4-dioxane
  • a suitable temperature such as 20-85 °C
  • Compounds of formula (XV) may be made by reaction of a compound of formula (XVII) with a compound of formula (VIII) under conditions known in the art as suitable for Pictet-Spengler reactions, such as in the presence of acid (such as acetic acid or trifluoroacetic acid) at a suitable temperature (such as 60-150°C) optionally with microwave irradiation.
  • acid such as acetic acid or trifluoroacetic acid
  • suitable temperature such as 60-150°C
  • L is for example a halogen (such as Br) or a trifluoromethanesulfonyl (triflate) group or a boronic acid or a boronate ester or alternatively an acrylate ester.
  • a pharmaceutically acceptable salt of a compound of the Formula (I) When a pharmaceutically acceptable salt of a compound of the Formula (I) is required it may be obtained by, for example, reaction of said compound with a suitable acid or suitable base.
  • a pharmaceutically acceptable pro-drug of a compound of the Formula (I) When a pharmaceutically acceptable pro-drug of a compound of the Formula (I) is required, it may be obtained using a conventional procedure.
  • an in-vivo cleavable ester of compound of the Formula (I) may be obtained by, for example, reaction of a compound of the Formula (I) containing a carboxy group with a pharmaceutically acceptable alcohol. Further information on pro-drugs has been provided hereinbefore.
  • a suitable protecting group for an amino or alkylamino group is, for example, an acyl group, for example an alkanoyl group such as acetyl, an alkoxycarbonyl group, for example a methoxycarbonyl, ethoxycarbonyl or i-butoxycarbonyl group, an acyl group, for example an alkanoyl group such as acetyl, an alkoxycarbonyl group, for example a methoxycarbonyl, ethoxycarbonyl or i-butoxycarbonyl group, an alkanoyl group such as acetyl, an alkoxycarbonyl group, for example a methoxycarbonyl, ethoxycarbonyl or i-butoxycarbonyl group, an acyl group, for example an alkanoyl group such as acetyl, an alkoxycarbonyl group, for example a methoxycarbonyl, ethoxycarbonyl or i-butoxycarbonyl group
  • arylmethoxycarbonyl group for example benzyloxycarbonyl, or an aroyl group, for example benzoyl.
  • the deprotection conditions for the above protecting groups necessarily vary with the choice of protecting group.
  • an acyl group such as an alkanoyl or alkoxycarbonyl group or an aroyl group may be removed for example, by hydrolysis with a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide.
  • an acyl group such as a i-butoxycarbonyl group may be removed, for example, by treatment with a suitable acid as hydrochloric, sulphuric or phosphoric acid or trifluoroacetic acid and an arylmethoxycarbonyl group such as a benzyloxycarbonyl group may be removed, for example, by hydrogenation over a catalyst such as palladium-on-carbon, or by treatment with a Lewis acid for example boron tris(trifluoroacetate).
  • a suitable alternative protecting group for a primary amino group is, for example, a phthaloyl group which may be removed by treatment with an alkylamine, for example dimethylaminopropylamine, or with hydrazine.
  • a suitable protecting group for a hydroxy group is, for example, an acyl group, for example an alkanoyl group such as acetyl, an aroyl group, for example benzoyl, or an arylmethyl group, for example benzyl.
  • the deprotection conditions for the above protecting groups will necessarily vary with the choice of protecting group.
  • an acyl group such as an alkanoyl or an aroyl group may be removed, for example, by hydrolysis with a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide.
  • a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide.
  • an arylmethyl group such as a benzyl group may be removed, for example, by hydrogenation over a catalyst such as palladium-on- carbon.
  • a suitable protecting group for a carboxy group is, for example, an esterifying group, for example a methyl or an ethyl group which may be removed, for example, by hydrolysis with a base such as sodium hydroxide, or for example a i-butyl group which may be removed, for example, by treatment with an acid, for example an organic acid such as trifluoroacetic acid, or for example a benzyl group which may be removed, for example, by hydrogenation over a catalyst such as palladium-on-carbon.
  • a base such as sodium hydroxide
  • a i-butyl group which may be removed, for example, by treatment with an acid, for example an organic acid such as trifluoroacetic acid, or for example a benzyl group which may be removed, for example, by hydrogenation over a catalyst such as palladium-on-carbon.
  • the protecting groups may be removed at any convenient stage in the synthesis using conventional techniques well known in the chemical art.
  • Estrogen Receptor Alpha Ligand binding domain ER alpha - LBD (GST)
  • GST LanthaScreenTM Time-Resolved Fluorescence Resonance Energy Transfer
  • TR-FRET Time-Resolved Fluorescence Resonance Energy Transfer
  • a suitable fluorophore Fluormone ES2, ThermoFisher, Product code P2645
  • recombinant human Estrogen Receptor alpha ligand binding domain residues 307-554 (expressed and purified in-house) were used to measure compound binding.
  • the assay principle is that ER alpha -LBD (GST) is added to a fluorescent ligand to form a receptor/fluorophore complex.
  • a terbium- labelled anti-GST antibody (Product code PV3551) is used to indirectly label the receptor by binding to its GST tag, and competitive binding is detected by a test compounds' ability to displace the fluorescent ligand resulting in a loss of TR-FRET signal between the Tb- anti-GST antibody and the tracer.
  • the assay was performed as follows with all reagent additions carried out using the Beckman Coulter BioRAPTR FRD microfluidic
  • the Echo 550 is a liquid handler that uses acoustic technology to perform direct microplate-to-microplate transfers of DMSO compound solutions and the system can be programmed to transfer multiple small nL volumes of compound from the different source plate wells to give the desired serial dilution of compound in the assay which is then back-filled to normalise the DMSO concentration across the dilution range.
  • TR-FRET dose response data obtained with each compound was exported into a suitable software package (such as Origin or Genedata) to perform curve fitting analysis.
  • Competitive ER alpha binding was expressed as an IC50 value. This was determined by calculation of the concentration of compound that was required to give a 50% reduction in tracer compound binding to ER alpha-LBD.
  • Estrogen Receptor (ER) numbers was assessed in a cell based immuno-fluorescence assay using the MCF-7 human ductal carcinoma breast cell line.
  • MCF-7 cells were revived directly from a cryovial (approx 5 x 10 6 cells) in Assay Medium (phenol red free Dulbecco's Modified Eagle's medium
  • DMEM fetal calf serum
  • Assay Medium a density of 3.75 x 10 4 cells per mL and 40 ⁇ ⁇ per well added to transparent bottomed, black, tissue culture treated 384 well plates (Costar, No. 3712) using a Thermo Scientific Matrix WellMate or Thermo
  • Multidrop Following cell seeding, plates were incubated overnight at 37°C, 5% C0 2 (Liconic carousel incubator). Test data was generated using the LabCyte EchoTM model 555 compound reformatter which is part of an automated workcell (Integrated Echo 2 workcell). lOmM compound stock solutions of the test compounds were used to generate a 384 well compound dosing plate (Labcyte P-05525-CV1). 40 ⁇ of each of the lOmM compound stock solutions was dispensed into the first quadrant well and then 1: 100 stepwise serial dilutions in DMSO were performed using a Hydra II (MATRIX UK) liquid handling unit to give 40 ⁇ L ⁇ of diluted compound into quadrant wells 2 (O.
  • PBS/Proclin PBS with a Biocide preservative
  • BioTek platewasher 40 ⁇ L ⁇ of PBS/Proclin was then added to all wells and the plates stored at 4°C.
  • the fixing method described above was carried out on the Integrated Echo 2 workcell. Immunostaining was performed using an automated AutoElisa workcell.
  • the PBS/Proclin was aspirated from all wells and the cells permeabilised with 40 ⁇ ⁇ PBS containing 0.5% TweenTM 20 (v/v) for 1 hour at room temperature.
  • the plates were washed three times in 250 ⁇ of PBS/0.05% (v/v) Tween 20 with Proclin (PBST with a Biocide preservative) and then 20 ⁇ L ⁇ of ERa (SP1) Rabbit monoclonal antibody (Thermofisher) 1: 1000 in PBS/TweenTM/3% (w/v) Bovine Serum Albumin was added.
  • the plates were incubated overnight at 4°C (Liconic carousel incubator) and then washed three times in 250 ⁇ of PBS/0.05% (v/v) TweenTM 20 with Proclin (PBST).
  • the plates were then incubated with 20 ⁇ 11 of a goat anti-rabbit IgG AlexaFluor 594 or goat anti-rabbit AlexaFluor 488 antibody (Molecular Probes) with Hoechst at 1:5000 in PBS/TweenTM/3% (w/v) Bovine Serum Albumin for lhour at room temperature.
  • the plates were then washed three times in 250 ⁇ of PBS/0.05% (v/v) TweenTM 20 with Proclin (PBST with a Biocide preservative). 20 ⁇ ⁇ of PBS was added to each well and the plates covered with a black plate seal and stored at 4°C before being read.
  • Plates were read using a Cellomics Arrayscan reading the 594nm (24hr time point) or 488nm (5hr timepoint) fluorescence to measure the ERa receptor level in each well. The mean total intensity was normalized for cell number giving the total intensity per cell. The data was exported into a suitable software package (such as Origin) to perform curve fitting analysis. Down-regulation of the ERa receptor was expressed as an IC50 value and was determined by calculation of the concentration of compound that was required to give a 50% reduction of the average maximum Total Intensity signal.
  • V incubation volume (0.25 mL); number of hepatocytes per well (0.25x10 6 cells).
  • a pharmaceutical composition which comprises a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore in association with a pharmaceutically acceptable diluent or carrier.
  • Suitable pharmaceutically acceptable excipients for a tablet formulation include, for example, inert diluents, granulating and disintegrating agents, binding agents, lubricating agents, preservative agents and antioxidants.
  • a further suitable pharmaceutically acceptable excipient may be a chelating agent.
  • Tablet formulations may be uncoated or coated either to modify their disintegration and the subsequent absorption of the active ingredient within the gastrointestinal tract, or to improve their stability and/or appearance, in either case, using conventional coating agents and procedures well known in the art.
  • compositions for oral use may alternatively be in the form of hard gelatin capsules in which the active ingredient is mixed with an inert solid diluent, or as soft gelatin capsules in which the active ingredient is mixed with water or an oil.
  • Aqueous suspensions generally contain the active ingredient in finely powdered form together with one or more suspending agents, dispersing or wetting agents.
  • the aqueous suspensions may also contain one or more preservatives, anti-oxidants, colouring agents, flavouring agents, and/or sweetening agents.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil or in a mineral oil.
  • the oily suspensions may also contain a thickening agent. Sweetening agents such as those set out above, and flavouring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water generally contain the active ingredient together with a dispersing or wetting agent, suspending agent and one or more preservatives. Additional excipients such as sweetening, flavouring and colouring agents, may also be present.
  • compositions of the specification may also be in the form of oil-in- water emulsions.
  • the oily phase may be a vegetable oil or a mineral oil or a mixture of any of these.
  • the emulsions may also contain sweetening, flavouring and preservative agents.
  • Syrups and elixirs may be formulated with sweetening agents, and may also contain a demulcent, preservative, flavouring and/or colouring agent.
  • compositions may also be in the form of a sterile injectable aqueous or oily suspension, which may be formulated according to known procedures using one or more of the appropriate dispersing or wetting agents and suspending agents, which have been mentioned above.
  • a sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent system.
  • Compositions for administration by inhalation may be in the form of a conventional pressurised aerosol arranged to dispense the active ingredient either as an aerosol containing finely divided solid or liquid droplets.
  • Conventional aerosol propellants such as volatile fluorinated hydrocarbons or hydrocarbons may be used and the aerosol device is conveniently arranged to dispense a metered quantity of active ingredient.
  • Dry powder inhalers may also be suitable.
  • the amount of active ingredient that is combined with one or more excipients to produce a single dosage form will necessarily vary depending upon the host treated and the particular route of administration.
  • oral administration to humans will generally require, for example, from 1 mg to 2 g of active agent (more suitably from lOOmg to 2g, for example from 250 mg to 1.8g, such as from 500mg to 1.8g, particularly from 500mg to 1.5g, conveniently from 500mg to lg) to be administered compounded with an appropriate and convenient amount of excipients which may vary from about 3 to about 98 percent by weight of the total composition.
  • unit dosage forms will contain about 10 mg to 0.5 g of a compound of this specification, although a unit dosage form may contain up to lg. Conveniently, a single solid dosage form may contain between 1 and 300mg of active ingredient.
  • the size of the dose for therapeutic or prophylactic purposes of compounds of the present specification will naturally vary according to the nature and severity of the disease state, the age and sex of the animal or patient and the route of administration, according to well known principles of medicine.
  • a daily dose in the range for example, 1 mg/kg to 100 mg/kg body weight is received, given if required in divided doses.
  • lower doses will be administered when a parenteral route is employed.
  • a dose in the range for example, 1 mg/kg to 25 mg/kg body weight will generally be used.
  • a dose in the range for example, 1 mg/kg to 25 mg/kg body weight will be used.
  • Oral administration is however preferred, particularly in tablet form.
  • compounds of the present specification or pharmaceutically acceptable salts thereof are administered as tablets comprising lOmg to 300mg of the compound of Formula (I) (or a pharmaceutically acceptable salt thereof), wherein one or more tablets are administered as required to achieve the desired dose.
  • the compounds of the present specification may be of value as anti- tumour agents, in particular as selective inhibitors of the proliferation, survival, motility, dissemination and invasiveness of mammalian cancer cells leading to inhibition of tumour growth and survival and to inhibition of metastatic tumour growth.
  • the compounds of the present specification may be of value as anti-proliferative and anti- invasive agents in the containment and/or treatment of solid tumour disease.
  • the compounds of the present specification may be useful in the prevention or treatment of those tumours which are sensitive to inhibition of ERa and that are involved in the signal transduction steps which lead to the proliferation and survival of tumour cells and the migratory ability and invasiveness of metastasising tumour cells.
  • the compounds of the present specification may be useful in the prevention or treatment of those tumours which are mediated alone or in part by antagonism and down-regulation of ERa, i.e. the compounds may be used to produce an ERa inhibitory effect in a warm-blooded animal in need of such treatment.
  • a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore for use as a medicament in a warm-blooded animal such as man.
  • a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore for use in the production of an anti-proliferative effect in a warm-blooded animal such as man.
  • a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore for use in a warm-blooded animal such as man as an anti-invasive agent in the containment and/or treatment of solid tumour disease.
  • a method for producing an anti-proliferative effect in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
  • a method for producing an anti-invasive effect by the containment and/or treatment of solid tumour disease in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
  • a method for the prevention or treatment of cancer in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
  • a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore for use in the prevention or treatment of solid tumour disease in a warm-blooded animal such as man.
  • a method for the prevention or treatment of solid tumour disease in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
  • a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore for use in the prevention or treatment of those tumours which are sensitive to inhibition of ERa that are involved in the signal transduction steps which lead to the proliferation, survival, invasiveness and migratory ability of tumour cells.
  • a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore in the manufacture of a medicament for use in the prevention or treatment of those tumours which are sensitive to inhibition of ERa that are involved in the signal transduction steps which lead to the proliferation, survival, invasiveness and migratory ability of tumour cells.
  • tumours which are sensitive to inhibition of ERa that are involved in the signal transduction steps which lead to the proliferation, survival, invasiveness and migratory ability of tumour cells which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
  • a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore for use in providing an inhibitory effect on ERa According to a further aspect of the specification there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore for use in providing an inhibitory effect on ERa. According to a further aspect of the specification there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore in the manufacture of a medicament for use in providing an inhibitory effect on ERa.
  • a method for providing an inhibitory effect on ERa which comprises administering an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
  • a method for providing a selective inhibitory effect on ERa which comprises administering an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
  • Described herein are compounds that can bind to ERa ligand binding domain and are selective estrogen receptor degraders.
  • the compounds of the present specification are shown to be potent estrogen receptor binders and reduce cellular levels of ERa and may therefore be useful in the treatment of estrogen sensitive diseases or conditions (including diseases that have developed resistance to endocrine therapies), i.e. for use in the treatment of cancer of the breast and gynaecological cancers (including endometrial, ovarian and cervical) and cancers expressing ERa mutated proteins which may be de novo mutations or have arisen as a result of treatment with a prior endocrine therapy such as an aromatase inhibitor.
  • a method for treating breast or gynaecological cancers which comprises administering an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
  • a method for treating cancer of the breast, endometrium, ovary or cervix which comprises administering an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
  • a method for treating breast cancer which comprises administering an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
  • a method for treating breast cancer wherein the cancer has developed resistance to one or more other endocrine therapies, which comprises administering an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
  • the cancer to be treated is breast cancer.
  • the breast cancer is Estrogen Receptor +ve (ER+ve).
  • the compound of Formula (I) is dosed in combination with another anticancer agent, such as an anti-hormonal agent as defined herein.
  • a method for treating ER+ve breast cancer which comprises administering an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
  • the in-vivo effects of a compound of the Formula (I) may be exerted in part by one or more metabolites that are formed within the human or animal body after administration of a compound of the Formula (I).
  • the present specification therefore also contemplates a method for inhibiting ER-cc in a patient, comprising administering to a patient an amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, effective in inhibiting ER-cc in the patient.
  • the present specification therefore also contemplates a method for inhibiting ER-cc in a patient, comprising administering to a patient an amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, effective in inhibiting ER-cc in the patient.
  • the anti-cancer treatment defined herein may be applied as a sole therapy or may involve, in addition to the compounds of the specification, conventional surgery or radiotherapy or chemotherapy.
  • Such chemotherapy may include one or more of the following categories of anti-tumour agents :-
  • antiproliferative/antineoplastic drugs and combinations thereof as used in medical oncology, such as alkylating agents (for example cis-platin, oxaliplatin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan, temozolamide and nitrosoureas); antimetabolites (for example gemcitabine and antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, and hydroxyurea); antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin- C, dactinomycin and mithramycin); antimitotic agents (for example vinca alkaloids like vincristine, vinblast
  • antihormonal agents such as antioestrogens (for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene), progestogens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrozole, vorazole and exemestane);
  • antioestrogens for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene
  • progestogens for example megestrol acetate
  • aromatase inhibitors for example as anastrozole, letrozole, vorazole and exemestane
  • kinase inhibitors examples include the anti-erbB2 antibody trastuzumab [HerceptinTM], the anti-EGFR antibody panitumumab, the anti-EGFR antibody cetuximab [Erbitux, C225] and tyrosine kinase inhibitors including inhibitors of the erbB receptor family, such as epidermal growth factor family receptor (EGFR/erbBl) tyrosine kinase inhibitors such as gefitinib or erlotinib, erbB2 tyrosine kinase inhibitors such as lapatinib, and mixed erbl/2 inhibitors such as afatanib; similar strategies are available for other classes of growth factors and their receptors, for example inhibitors
  • iv) modulators of DNA damage signalling pathways for example PARP inhibitors (e.g. Olaparib), ATR inhibitors or ATM inhibitors;
  • v) modulators of apoptotic and cell death pathways such as Bel family modulators (e.g. ABT-263 / Navitoclax, ABT-199);
  • antiangiogenic agents such as those which inhibit the effects of vascular endothelial growth factor, [for example the anti-vascular endothelial cell growth factor antibody bevacizumab (AvastinTM) and for example, a VEGF receptor tyrosine kinase inhibitor such as sorafenib, axitinib, pazopanib, sunitinib and vandetanib (and compounds that work by other mechanisms (for example linomide, inhibitors of integrin ⁇ 3 function and angio statin)];
  • vascular endothelial growth factor for example the anti-vascular endothelial cell growth factor antibody bevacizumab (AvastinTM) and for example, a VEGF receptor tyrosine kinase inhibitor such as sorafenib, axitinib, pazopanib, sunitinib and vandetanib (and compounds that work by other mechanisms (for example linomide
  • vascular damaging agents such as Combretastatin A4
  • anti-invasion agents for example c-Src kinase family inhibitors like (dasatinib, L Med. Chem., 2004, 47, 6658-6661) and bosutinib (SKI-606), and metalloproteinase inhibitors like marimastat, inhibitors of urokinase plasminogen activator receptor function or antibodies to Heparanase] ;
  • immunotherapy approaches including for example ex-vivo and in- vivo approaches to increase the immunogenicity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor, approaches to decrease T-cell anergy, approaches using transfected immune cells such as cytokine-transfected dendritic cells, approaches using cytokine-transfected tumour cell lines and approaches using anti-idiotypic antibodies.
  • cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor
  • approaches to decrease T-cell anergy approaches using transfected immune cells such as cytokine-transfected dendritic cells
  • approaches using cytokine-transfected tumour cell lines approaches using anti-idiotypic antibodies.
  • PD-1 e.g. BMS-936558
  • CTLA4 e.g. ipilimumab and tremelimumab
  • (xi) gene therapy approaches including for example approaches to replace aberrant genes such as aberrant p53 or aberrant BRCA1 or BRCA2, GDEPT (gene-directed enzyme pro-drug therapy) approaches such as those using cytosine deaminase, thymidine kinase or a bacterial nitroreductase enzyme and approaches to increase patient tolerance to chemotherapy or radiotherapy such as multi-drug resistance gene therapy.
  • GDEPT gene-directed enzyme pro-drug therapy
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and an additional anti-tumour substance for the conjoint treatment of cancer are provided.
  • a combination suitable for use in the treatment of cancer comprising a compound of the Formula (I) or a pharmaceutically acceptable salt thereof and another anti-tumour agent, in particular any one of the anti tumour agents listed under (i) - (xi) above.
  • another anti-tumour agent in particular any one of the anti tumour agents listed under (i) - (xi) above.
  • the anti-tumour agent listed under (i)-(xi) above is the standard of care for the specific cancer to be treated; the person skilled in the art will understand the meaning of "standard of care”.
  • a compound of the Formula (I), or a pharmaceutically acceptable salt thereof in combination with another anti-tumour agent, in particular an anti-tumour agent selected from one listed under (i) - (xi) herein above.
  • a compound of the Formula (I), or a pharmaceutically acceptable salt thereof in combination with another anti-tumour agent, in particular an anti-tumour agent selected from one listed under (i) above.
  • a combination suitable for use in the treatment of cancer comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and a taxoid, such as for example taxol or taxotere, conveniently taxotere.
  • a compound of the Formula (I), or a pharmaceutically acceptable salt thereof in combination with another anti-tumour agent, in particular an anti-tumour agent selected from one listed under (ii) herein above.
  • a combination suitable for use in the treatment of cancer comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and any one of the antihormonal agents listed under (ii) above, for example any one of the anti-oestrogens listed in (ii) above, or for example an aromatase inhibitor listed in (ii) above.
  • a combination suitable for use in the treatment of cancer comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and an mTOR inhibitor, such as AZD2014 (see for example WO2008/023161).
  • a combination suitable for use in the treatment of cancer comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and a PDKa-inhibitor, such as the compound l-(4-(5-(5-amino-6- (5-tert-butyl- 1 ,3,4-oxadiazol-2-yl)pyrazin-2-yl)- 1 -ethyl- 1H- 1 ,2,4-triazol-3-yl)piperidin- 1 - yl)-3-hydroxypropan-l-one, or a pharmaceutically- acceptable salt thereof.
  • a PDKa-inhibitor such as the compound l-(4-(5-(5-amino-6- (5-tert-butyl- 1 ,3,4-oxadiazol-2-yl)pyrazin-2-yl)- 1 -ethyl- 1H- 1 ,2,4-triazol-3-yl)piperidin- 1 -
  • a combination suitable for use in the treatment of cancer comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and palbociclib.
  • an anti-tumour agent listed in (ii) above or an mTOR inhibitor (such as AZD2014), or a PB
  • administering the second component should not be such as to lose the beneficial effect of the combination.
  • a combination of two or more components is administered separately or sequential, it will be understood that the dosage regime for each component may be different to and independent of the other components. Conveniently, the
  • a pharmaceutical composition which comprises a compound of Formula (I) or a pharmaceutically acceptable salt thereof in combination with an anti-tumour agent selected from one listed under (i) - (xi) herein above, in association with a pharmaceutically acceptable diluent or carrier.
  • a pharmaceutical composition which comprises a compound of Formula (I) or a pharmaceutically acceptable salt thereof in combination with any one of antihormonal agents listed under (ii) above, for example any one of the anti-oestrogens listed in (ii) above, or for example an aromatase inhibitor listed in (ii) above in association with a pharmaceutically acceptable diluent or carrier.
  • an mTOR inhibitor such as AZD2014 (see for example WO2008/023161)
  • a compound of the Formula (I), or a pharmaceutically acceptable salt thereof comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and a PDK -inhibitor, such as the compound l-(4-(5-(5-amino-6-(5-tert-butyl-l,3,4- oxadiazol-2-yl)pyrazin-2-yl)- 1 -ethyl- 1H- l,2,4-triazol-3-yl)piperidin- l-yl)-3- hydroxypropan-l-one, in association with a pharmaceutically acceptable diluent or carrier.
  • a PDK -inhibitor such as the compound l-(4-(5-(5-amino-6-(5-tert-butyl-l,3,4- oxadiazol-2-yl)pyrazin-2-yl)- 1 -ethyl- 1H- l,2,4-tria
  • a pharmaceutical composition which comprises a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an anti-tumour agent selected from one listed under (i) - (xi) herein above, in association with a pharmaceutically acceptable diluent or carrier for use in treating cancer.
  • a pharmaceutical composition which comprises a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with any one of antihormonal agents listed under (ii) above, for example any one of the anti-oestrogens listed in (ii) above, or for example an aromatase inhibitor listed in (ii) above in association with a pharmaceutically acceptable diluent or carrier for use in treating cancer.
  • an mTOR inhibitor such as AZD2014 (see for example WO2008/023161)
  • a compound of the Formula (I), or a pharmaceutically acceptable salt thereof comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and a PDK -inhibitor, such as the compound l-(4-(5-(5-amino-6-(5-tert-butyl-l,3,4- oxadiazol-2-yl)pyrazin-2-yl)- 1 -ethyl- 1H- l,2,4-triazol-3-yl)piperidin- l-yl)-3- hydroxypropan-l-one, in association with a pharmaceutically acceptable diluent or carrier for use in treating cancer.
  • a PDK -inhibitor such as the compound l-(4-(5-(5-amino-6-(5-tert-butyl-l,3,4- oxadiazol-2-yl)pyrazin-2-yl)- 1 -ethyl- 1H- l,
  • an anti-tumour agent listed in (ii) above or an mTOR inhibitor (such as AZD2014), or a PB
  • a compound of the Formula (I) or a pharmaceutically acceptable salt thereof in combination with an anti- tumour agent selected from one listed under (i) - (xi) herein above, in the manufacture of a medicament for use in the treatment of cancer in a warm-blooded animal, such as man.
  • a compound of Formula (I) or a pharmaceutically acceptable salt thereof in combination with any one of antihormonal agents listed under (ii) above, for example any one of the anti-oestrogens listed in (ii) above, or for example an aromatase inhibitor listed in (ii) above in the manufacture of a medicament for use in the treatment of cancer in a warm-blooded animal, such as man.
  • a compound of the Formula (I), or a pharmaceutically acceptable salt thereof in combination with an mTOR inhibitor, such as AZD2014 (see for example WO2008/023161); in the manufacture of a medicament for use in the treatment of cancer in a warm-blooded animal, such as man.
  • an mTOR inhibitor such as AZD2014 (see for example WO2008/023161)
  • a compound of the Formula (I), or a pharmaceutically acceptable salt thereof in combination with a PBKcc-inhibitor, such as the compound l-(4-(5-(5-amino-6-(5-tert-butyl-l,3,4-oxadiazol-2-yl)pyrazin-2-yl)- l-ethyl-lH-l,2,4-triazol-3-yl)piperidin-l-yl)-3-hydroxypropan-l-one, in the manufacture of a medicament for use in the treatment of cancer in a warm-blooded animal, such as man.
  • a PBKcc-inhibitor such as the compound l-(4-(5-(5-amino-6-(5-tert-butyl-l,3,4-oxadiazol-2-yl)pyrazin-2-yl)- l-ethyl-lH-l,2,4-triazol-3-yl)piperidin-
  • a method of treating cancer in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an anti-tumour agent selected from one listed under (i) - (xi) herein above.
  • a method of treating cancer in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in combination with any one of antihormonal agents listed under (ii) above, for example any one of the anti-oestrogens listed in (ii) above, or for example an aromatase inhibitor listed in (ii) above.
  • a method of treating cancer in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an mTOR inhibitor, such as AZD2014 (see for example WO2008/023161).
  • warm-blooded animal such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a PBKcc-inhibitor, such as the compound l-(4-(5-(5-amino-6-(5-tert-butyl-l,3,4-oxadiazol-2-yl)pyrazin-2-yl)-l-ethyl- lH-l,2,4-triazol-3-yl)piperidin-l-yl)-3-hydroxypropan-l-one.
  • a PBKcc-inhibitor such as the compound l-(4-(5-(5-amino-6-(5-tert-butyl-l,3,4-oxadiazol-2-yl)pyrazin-2-yl)-l-ethyl- lH-l,2,4-triazol-3-yl)piperidin-l
  • a method of treating cancer in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with palbociclib.
  • compositions, uses and methods of treating cancer are methods for the treatment of breast or gynaecological cancers, such as cancer of the breast, endometrium, ovary or cervix, particularly breast cancer, such as ER+ve breast cancer.
  • kits comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof in combination with an anti- tumour agent selected from one listed under (i) - (xi) herein above.
  • kits comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof in combination with an anti- tumour agent selected from one listed under (i) or (ii) herein above.
  • an anti-tumour agent selected from one listed under (i) - (xi) herein above in a second unit dosage form;
  • an anti-tumour agent selected from one listed under (i) - (ii) herein above in a second unit dosage form;
  • an anti-tumour agent selected from an anti-tumour agent listed in (ii) above, an mTOR inhibitor (such as AZD2014), a PDK -inhibitor, such as the compound l-(4-(5-(5-amino- 6-(5-tert-butyl-l,3,4-oxadiazol-2-yl)pyrazin-2-yl)-l-ethyl-lH-l,2,4-triazol-3-yl)piperidin- l-yl)-3-hydroxypropan-l-one, and palbociclib, in a second unit dosage form; and c) container means for containing said first and second dosage forms.
  • an mTOR inhibitor such as AZD2014
  • a PDK -inhibitor such as the compound l-(4-(5-(5-amino- 6-(5-tert-butyl-l,3,4-oxadiazol-2-yl)pyrazin-2-yl
  • Combination therapy as described above may be added on top of standard of care therapy typically carried out according to its usual prescribing schedule.
  • the compounds of the Formula (I) are primarily of value as therapeutic agents for use in warm-blooded animals (including man), they are also useful whenever it is required to inhibit ER-cc. Thus, they are useful as pharmacological standards for use in the development of new biological tests and in the search for new pharmacological agents.
  • Another aspect of the present specification is based on identifying a link between the status of the gene encoding ERa and potential susceptibility to treatment with a compound of Formula (I).
  • ERa gene status may indicate that a patient is less likely to respond to exisiting hormone therapy (such as aromatase inhibitors), in part at least because some ERa mutations are though to arise as resistance mechanisms to existing treatments.
  • a SERD particularly a SERD which can be administered orally in potentially larger doses without excessive inconvenince, may then advantageously be used to treat patentients with ERa mutations who may be resistant to other therapies. This therefore provides opportunities, methods and tools for selecting patients for treatment with a compound of Formula (I), particularly cancer patients.
  • the present specification relates to patient selection tools and methods (including personalised medicine).
  • the selection is based on whether the tumour cells to be treated possess wild-type or mutant ERa gene.
  • the ERa gene status could therefore be used as a biomarker to indicate that selecting treatment with a SERD may be advantageous.
  • compounds of the Formula (I) as described herein are thought to be similarly active against wild-type and mutant ERa genes, at least those mutations in ERa gene identified at the date of filing this application.
  • Patient selection biomarkers that identify the patients most likely to respond to one agent over another are ideal in the treatment of cancer, since they reduce the unnecessary treatment of patients with non-responding tumours to the potential side effects of such agents.
  • a biomarker can be described as "a characteristic that is objectively measured and evaluated as an indicator of normal biologic processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention".
  • a biomarker is any identifiable and measurable indicator associated with a particular condition or disease where there is a correlation between the presence or level of the biomarker and some aspect of the condition or disease (including the presence of, the level or changing level of, the type of, the stage of, the susceptibility to the condition or disease, or the responsiveness to a drug used for treating the condition or disease). The correlation may be qualitative, quantitative, or both qualitative and quantitative.
  • a biomarker is a compound, compound fragment or group of compounds. Such compounds may be any compounds found in or produced by an organism, including proteins (and peptides), nucleic acids and other compounds.
  • Biomarkers may have a predictive power, and as such may be used to predict or detect the presence, level, type or stage of particular conditions or diseases (including the presence or level of particular microorganisms or toxins), the susceptibility (including genetic susceptibility) to particular conditions or diseases, or the response to particular treatments (including drug treatments). It is thought that biomarkers will play an increasingly important role in the future of drug discovery and development, by improving the efficiency of research and development programs. Biomarkers can be used as diagnostic agents, monitors of disease progression, monitors of treatment and predictors of clinical outcome. For example, various biomarker research projects are attempting to identify markers of specific cancers and of specific cardiovascular and immunological diseases. It is believed that the development of new validated biomarkers will lead both to significant reductions in healthcare and drug development costs and to significant improvements in treatment for a wide variety of diseases and conditions.
  • a biomarker may be required.
  • the marker may be measurable in surrogate and tumour tissues. Ideally these markers will also correlate with efficacy and thus could ultimately be used for patient selection.
  • Tumours which contain wild type ERa are believed to be susceptible to treatment with a compound of Formula (I), for example as a first- line treatment. Tumours may also respond to treatment with a compound of Formula (I) as a second- line, third-line or subsequent therapy and this may be useful, in particular, where the tumours contain mutant ERa and may thus be resistant to existing therapies such as AIs. A higher dosage of a compound of Formula (I) may be required in the resistant setting than in wild type tumours).
  • the specification provides a method of determining sensitivity of cells to a compound of Formula (I).
  • the method comprises determining the status of ERa gene in said cells.
  • a cell is defined as sensitive to a compound of Formula (I) if it inhibits the increase in cell number in a cell growth assay (either through inhibition of cell proliferation and /or through increased cell death). Methods of the specification are useful for predicting which cells are more likely to respond to a compound of Formula (I) by growth inhibition.
  • a sample "representative of the tumour” can be the actual tumour sample isolated, or may be a sample that has been further processed, e.g. a sample of PCR amplified nucleic acid from the tumour sample.
  • Allele refers to a particular form of a genetic locus, distinguished from other forms by its particular nucleotide or amino acid sequence.
  • Amplification reactions are nucleic acid reactions which result in specific amplification of target nucleic acids over non-target nucleic acids.
  • the polymerase chain reaction (PCR) is a well known amplification reaction.
  • Cancer is used herein to refer to neoplastic growth arising from cellular transformation to a neoplastic phenotype. Such cellular transformation often involves genetic mutation.
  • Gene is a segment of DNA that contains all the information for the regulated biosynthesis of an RNA product, including a promoter, exons, introns, and other sequence elements which may be located within 5' or 3' flanking regions (not within the transcribed portions of the gene) that control expression.
  • Gene status refers to whether the gene is wild type or not (i.e. mutant).
  • Label refers to a composition capable of producing a detectable signal indicative of the presence of the target polynucleotide in an assay sample. Suitable labels include radioisotopes, nucleotide chromophores, enzymes, substrates, fluorescent molecules, chemiluminescent moieties, magnetic particles, bioluminescent moieties, and the like. As such, a label is any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means.
  • Non- synonymous variation refers to a variation (variance) in or overlapping the coding sequence of a gene that result in the production of a distinct (altered) polypeptide sequence. These variations may or may not affect protein function and include missense variants (resulting in substitution of one amino acid for another), nonsense variants (resulting in a truncated polypeptide due to generation of a premature stop codon) and insertion/deletion variants.
  • “Synonymous variation” refers to a variation (variance) in the coding sequence of a gene that does not affect sequence of the encoded polypeptide. These variations may affect protein function indirectly (for example by altering expression of the gene), but, in the absence of evidence to the contrary, are generally assumed to be innocuous.
  • Nucleic acid refers to single stranded or double stranded DNA and RNA molecules including natural nucleic acids found in nature and/or modified, artificial nucleic acids having modified backbones or bases, as are known in the art.
  • Primer refers to a single stranded DNA oligonucleotide sequence capable of acting as a point of initiation for synthesis of a primer extension product which is complementary to the nucleic acid strand to be copied.
  • the length and sequence of the primer must be such that they are able to prime the synthesis of extension products.
  • a typical primer contains at least about 7 nucleotides in length of a sequence substantially complementary to the target sequence, but somewhat longer primers are preferred. Usually primers contain about 15-26 nucleotides, but longer or shorter primers may also be employed.
  • Polymorphic site is a position within a locus at which at least two alternative sequences are found in a population.
  • Polymorphism refers to the sequence variation observed in an individual at a polymorphic site. Polymorphisms include nucleotide substitutions, insertions, deletions and microsatellites and may, but need not, result in detectable differences in gene expression or protein function. In the absence of evidence of an effect on expression or protein function, common polymorphisms, including non-synonymous variants, are generally considered to be included in the definition of wild-type gene sequence. A catalog of human polymorphisms and associated annotation, including validation, observed frequencies, and disease association, is maintained by NCBI (dbSNP:
  • polymorphism when used in the context of solid state form of a compound, that is the crystalline or amorphous nature of a compound. The skilled person will understand the intended meaning by its context.
  • Probe refers to single stranded sequence- specific oligonucleotides which have a sequence that is exactly complementary to the target sequence of the allele to be detected.
  • Response is defined by measurements taken according to Response Evaluation Criteria in Solid Tumours (RECIST) involving the classification of patients into two main groups: those that show a partial response or stable disease and those that show signs of progressive disease.
  • “Stringent hybridisation conditions” refers to an overnight incubation at 42°C in a solution comprising 50% formamide, 5x SSC (750 mM NaCI, 75 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5x Denhardt's solution, 10% dextran sulphate, and 20 pg/ml denatured, sheared salmon sperm DNA, followed by washing the filters in O.lx SSC at about 65 °C.
  • “Survival” encompasses a patients' overall survival and progression-free survival.
  • OS Global System for disease
  • PFS progression-free survival
  • a method for selecting a patient for treatment with a compound of Formula (I) comprising providing a tumour cell containing sample from a patient; determining whether the ERa gene in the patient's tumour cell containing sample is wild type or mutant; and selecting a patient for treatment with a compound of Formula (I) based thereon.
  • the method may include or exclude the actual patient sample isolation step.
  • a method for selecting a patient for treatment with a compound of Formula (I) comprising determining whether the ERa gene in a tumour cell containing sample previously isolated from the patient is wild type or mutant; and selecting a patient for treatment with a compound of Formula (I) based thereon.
  • the patient is selected for treatment with a compound of Formula (I) if the tumour cell DNA has a mutant ERa gene.
  • a patient whose tumour cell DNA possesses a wild type ERa gene is selected for treatment with a compound of Formula (I).
  • a gene status of wild-type is meant to indicate normal or appropriate expression of the gene and normal function of the encoded protein.
  • mutant status is meant to indicate expression of a protein with altered function, consistent with the known roles of mutant ERa genes in cancer (as described herein). Any number of genetic or epigenetic alterations, including but not limited to mutation, amplification, deletion, genomic rearrangement, or changes in methylation profile, may result in a mutant status. However, if such alterations nevertheless result in appropriate expression of the normal protein, or a functionally equivalent variant, then the gene status is regarded as wild-type.
  • variants that typically would not result in a functional mutant gene status include synonymous coding variants and common polymorphisms (synonymous or non-synonymous).
  • gene status can be assessed by a functional assay, or it may be inferred from the nature of detected deviations from a reference sequence.
  • the wild-type or mutant status of the ERa gene is determined by the presence or absence of non-synonymous nucleic acid variations in the genes. Observed non-synonymous variations corresponding to known common
  • splice site variations that decrease recognition of an intron/exon junction during processing of pre- mRNA to mRNA. This can result in exon skipping or the inclusion of normally intronic sequence in spliced mRNA (intron retention or utilization of cryptic splice junctions). This can, in turn, result in the production of aberrant protein with insertions and/or deletions relative to the normal protein.
  • the gene has a mutant status if there is a variant that alters splice site recognition sequence at an intron/exon junction.
  • ESR1 ESR1 gene status, i.e. whether a particular ESRlgene is wild type or mutant, based on comparison of DNA or protein sequence with wild type.
  • sequences disclosed for ERa gene are representative sequences. In normal individuals there are two copies of each gene, a maternal and paternal copy, which will likely have some sequence differences, moreover within a population there will exist numerous allelic variants of the gene sequence.
  • Other sequences regarded as wild type include those that possess one or more synonymous changes to the nucleic acid sequence (which changes do not alter the encoded protein sequence), non-synonymous common polymorphisms (e.g. germ-line polymorphisms) which alter the protein sequence but do not affect protein function, and intronic non-splice- site sequence changes.
  • the gene status of ERa can be determined by determination of the nucleic acid sequence. This could be via direct sequencing of the full-length gene or analysis of specific sites within the gene, e.g. commonly mutated sites.
  • the patient's sample to be tested for the gene status can be any tumour tissue or tumour-cell containing sample obtained or obtainable from the individual.
  • the test sample is conveniently a sample of blood, mouth swab, biopsy, or other body fluid or tissue obtained from an individual.
  • Particular examples include: circulating tumour cells, circulating DNA in the plasma or serum, cells isolated from the ascites fluid of ovarian cancer patients, lung sputum for patients with tumours within the lung, a fine needle aspirate from a breast cancer patient, urine, peripheral blood, a cell scraping, a hair follicle, a skin punch or a buccal sample.
  • test sample may equally be a nucleic acid sequence corresponding to the sequence in the test sample, that is to say that all or a part of the region in the sample nucleic acid may firstly be amplified using any convenient technique e.g. polymerase chain reaction (PCR), before analysis.
  • the nucleic acid may be genomic DNA or fractionated or whole cell RNA.
  • the RNA is whole cell RNA and is used directly as the template for labelling a first strand cDNA using random primers or poly A primers.
  • the nucleic acid or protein in the test sample may be extracted from the sample according to standard methodologies (see Green & Sambrook,
  • the diagnostic methods of the specification can be undertaken using a sample previously taken from the individual or patient. Such samples may be preserved by freezing or fixed and embedded in formalin-paraffin or other media. Alternatively, a fresh tumour cell containing sample may be obtained and used.
  • tumours for treatment with a compound of Formula (I) have been described hereinbefore.
  • mutant ERa nucleic acids can be employed, in the context of the present specification, to select drug treatment. Since mutations in these genes occur at the DNA level, the methods of the specification can be based on detection of mutations or variances in genomic DNA, as well as transcripts and proteins themselves. It can be desirable to confirm mutations in genomic DNA by analysis of transcripts and/or polypeptides, in order to ensure that the detected mutation is indeed expressed in the subject.
  • allelic variation requires a mutation discrimination technique, optionally an amplification reaction (such as one based on polymerase chain reaction) and optionally a signal generation system.
  • mutation detection techniques available in the art and these may be used in combination with a signal generation system, of which there are numerous available in the art.
  • Many methods for the detection of allelic variation are reviewed by Nollau et al., Clin. Chem., 1997, 43, 1114-1120; Anderson SM. Expert Rev Mol Diagn., 2011, 11, 635- 642; Meyerson M.
  • determining the presence or absence of a particular variance or plurality of variances in the ERa gene in a patient with cancer can be performed in a variety of ways. Such tests are commonly performed using DNA or RNA collected from biological samples, e.g., tissue biopsies, urine, stool, sputum, blood, cells, tissue scrapings, breast aspirates or other cellular materials, and can be performed by a variety of methods including, but not limited to, PCR, hybridization with allele- specific probes, enzymatic mutation detection, chemical cleavage of mismatches, mass spectrometry or DNA sequencing, including minisequencing.
  • biological samples e.g., tissue biopsies, urine, stool, sputum, blood, cells, tissue scrapings, breast aspirates or other cellular materials
  • methods including, but not limited to, PCR, hybridization with allele- specific probes, enzymatic mutation detection, chemical cleavage of mismatches, mass spectrometry or DNA sequencing, including minis
  • Suitable mutation detection techniques include amplification refractory mutation system (ARMSTM), amplification refractory mutation system linear extension (ALEXTM), competitive oligonucleotide priming system (COPS), Taqman, Molecular Beacons, restriction fragment length polymorphism (RFLP), and restriction site based PCR and fluorescence resonance energy transfer (FRET) techniques.
  • ARMSTM amplification refractory mutation system
  • ALEXTM amplification refractory mutation system linear extension
  • COPS competitive oligonucleotide priming system
  • RFLP restriction fragment length polymorphism
  • FRET fluorescence resonance energy transfer
  • the method employed for determining the nucleotide(s) within a biomarker gene is selected from: allele- specific amplification (allele specific PCR) - such as amplification refractory mutation system (ARMS), sequencing, allelic discrimination assay, hybridisation, restriction fragment length polymorphism (RFLP) or oligonucleotide ligation assay (OLA).
  • allele-specific amplification allele specific PCR
  • ARMS amplification refractory mutation system
  • allelic discrimination assay hybridisation
  • RFLP restriction fragment length polymorphism
  • OLA oligonucleotide ligation assay
  • hybridization with allele specific probes can be conducted by: (1) allele specific oligonucleotides bound to a solid phase (e.g. glass, silicon, nylon membranes) with the labelled sample in solution, for example as in many DNA chip applications; or, (2) bound sample (often cloned DNA or PCR amplified DNA) and labelled oligonucleotides in solution (either allele specific or short so as to allow
  • Diagnostic tests may involve a panel of variances, often on a solid support, which enables the simultaneous determination of more than one variance.
  • Such hybridization probes are well known in the art (see, e.g., Green & Sambrook, Eds., Molecular Cloning: A Laboratory Manual, (2012, 4th edition, Vol. 1-3, ISBN 9781936113422), Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.) and may span two or more variance sites.
  • the detection of the presence or absence of at least one mutation provides for contacting ERa nucleic acid containing a putative mutation site with at least one nucleic acid probe.
  • the probe preferentially hybridizes with a nucleic acid sequence including a variance site and containing complementary nucleotide bases at the variance site under selective hybridization conditions.
  • Hybridization can be detected with a detectable label using labels known to one skilled in the art. Such labels include, but are not limited to radioactive, fluorescent, dye, and enzymatic labels.
  • the detection of the presence or absence of at least one mutation provides for contacting ERa nucleic acid containing a putative mutation site with at least one nucleic acid primer.
  • the primer preferentially hybridizes with a nucleic acid sequence including a variance site and containing complementary nucleotide bases at the variance site under selective hybridization conditions.
  • Oligonucleotides used as primers for specific amplification may carry the complementary nucleotide base to the mutation of interest in the centre of the molecule (so that amplification depends on differential hybridization; see, e.g., Gibbs, et al., 1989. Nucl. Acids Res., 17, 2437-248) or at the extreme 3'-terminus of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (see, e.g., Prossner, 1993, Tibtech, 11 238).
  • the detection of the presence or absence of at least one mutation comprises sequencing at least one nucleic acid sequence and comparing the obtained sequence with the known wild type nucleic acid sequence.
  • the presence or absence of at least one mutation comprises mass spectrometric determination of at least one nucleic acid sequence.
  • the detection of the presence or absence of at least one nucleic acid variance comprises performing a polymerase chain reaction (PCR).
  • PCR polymerase chain reaction
  • the target nucleic acid sequence containing the hypothetical variance is amplified and the nucleotide sequence of the amplified nucleic acid is determined.
  • Determining the nucleotide sequence of the amplified nucleic acid comprises sequencing at least one nucleic acid segment.
  • amplification products can be analysed using any method capable of separating the amplification products according to their size, including automated and manual gel electrophoresis, and the like.
  • Mutations in genomic nucleic acid are advantageously detected by techniques based on mobility shift in amplified nucleic acid fragments. For instance, Chen et al., Anal Biochem 1996, 239, 61-9, describe the detection of single-base mutations by a competitive mobility shift assay. Moreover, assays based on the technique of Marcelino et al.,
  • capillary heteroduplex analysis may be used to detect the presence of mutations based on mobility shift of duplex nucleic acids in capillary systems as a result of the presence of mismatches.
  • nucleic acids for analysis from samples generally requires nucleic acid amplification.
  • Many amplification methods rely on an enzymatic chain reaction (such as a polymerase chain reaction, a ligase chain reaction, or a self- sustained sequence replication) or from the replication of all or part of the vector into which it has been cloned.
  • the amplification according to the specification is an exponential amplification, as exhibited by for example the polymerase chain reaction.
  • PCR polymerase chain reaction
  • LAR ligase amplification reaction
  • GAS transcription-based amplification system
  • GAWTS genomic amplification with transcript sequencing
  • NASBA nucleic acid sequence-based amplification
  • PCR Polymerase Chain Reaction
  • PCR is a nucleic acid amplification method described inter alia in U.S. Pat. Nos. 4,683,195 and 4,683,202.
  • PCR consists of repeated cycles of DNA polymerase generated primer extension reactions.
  • the target DNA is heat denatured and two oligonucleotides, which bracket the target sequence on opposite strands of the DNA to be amplified, are hybridised. These oligonucleotides become primers for use with DNA polymerase.
  • the DNA is copied by primer extension to make a second copy of both strands. By repeating the cycle of heat denaturation, primer hybridisation and extension, the target DNA can be amplified a million fold or more in about two to four hours.
  • PCR is a molecular biology tool, which must be used in conjunction with a detection technique to determine the results of amplification.
  • An advantage of PCR is that it increases sensitivity by amplifying the amount of target DNA by 1 million to 1 billion fold in approximately 4 hours.
  • PCR can be used to amplify any known nucleic acid in a diagnostic context (Mok et al., Gvnaecologic Oncology, 1994, 52: 247-252,).
  • An allele specific amplification technique such as Amplification Refractory Mutation System (ARMSTM) (Newton et al., Nucleic Acids Res., 1989, 17, 2503-2516) can also be used to detect single base mutations. Under the appropriate PCR amplification conditions a single base mismatch located at the 3'-end of the primer is sufficient for preferential amplification of the perfectly matched allele (Newton et al., 1989, supra), allowing the discrimination of closely related species.
  • the basis of an amplification system using the primers described above is that oligonucleotides with a mismatched 3'-residue will not function as primers in the PCR under appropriate conditions. This amplification system allows genotyping solely by inspection of reaction mixtures after agarose gel electrophoresis.
  • Analysis of amplification products can be performed using any method capable of separating the amplification products according to their size, including automated and manual gel electrophoresis, mass spectrometry, and the like.
  • kits comprising degenerate primers to amplify a target nucleic acid in the ERa gene and instructions comprising; amplification protocol and analysis of the results.
  • the kit may alternatively also comprise buffers, enzymes, and containers for performing the amplification and analysis of the amplification products.
  • the kit may also be a component of a screening, or diagnostic kit comprising other tools such as DNA microarrays, or other supports.
  • the kit also provides one or more control templates, such as nucleic acids isolated from normal tissue sample, and/or a series of samples representing different variances in the reference genes.
  • the kit provides two or more primer pairs, each pair capable of amplifying a different region of the reference (ERa) gene (each region a site of potential variance) thereby providing a kit for analysis of expression of several gene variances in a biological sample in one reaction or several parallel reactions.
  • ERa reference gene
  • Primers in the kits may be labelled, for example fluorescently labelled, to facilitate detection of the amplification products and consequent analysis of the nucleic acid variances.
  • the kit may also allow for more than one variance to be detected in one analysis.
  • a combination kit will therefore comprise of primers capable of amplifying different segments of the reference gene.
  • the primers may be differentially labelled, for example using different fluorescent labels, so as to differentiate between the variances.
  • the specification provides a method of treating a patient suffering from cancer comprising: determining the mutant or wild type status of the ERa gene in the patient' s tumour cells and if the ERa gene is mutant, administering to the patient an effective amount of a compound of Formula (I).
  • Pharmacological effectiveness refers to the ability of the treatment to result in a desired biological effect in the patient.
  • Physiological safety refers to the level of toxicity, or other adverse physiological effects at the cellular, organ and/or organism level (often referred to as side-effects) resulting from administration of the treatment. "Less effective” means that the treatment results in a therapeutically significant lower level of pharmacological effectiveness and/or a therapeutically greater level of adverse physiological effects.
  • a compound of Formula (I) or a pharmaceutically acceptable salt thereof for treating cancers with tumour cells identified as harbouring mutant ERa gene According to another aspect of the specification there is provided a method of treating cancers with tumour cells identified as harbouring mutant ERa gene comprising administering an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • the specification relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula (I) for use in the prevention and treatment of cancer with tumour cells identified as harbouring a mutant ERa gene.
  • mutant forms of ERa determined/identified are at all positions across the gene.
  • tumours such as breast cancer
  • particular mutant forms of ERa determined/identified are those at positions Ser463Pro, Val543Glu, Leu536Arg, Tyr537Ser, Tyr537Asn and Asp538Gly.
  • the column used was Waters Acquity HSS T3 1.8 ⁇ 2.1 x50 mm
  • the column used was Waters Acquity BEH 1.7 ⁇ 2.1x50mm
  • LCMS was carried out using a Waters Alliance HT (2795) fitted with a Waters ZQ ESCi mass spectrometer and a Phenomenex Gemini -NX (50x2.1mm 5 ⁇ ) column at a flow rate of 1. lml/min 95%A to 95%B over 4 min with a 0.5 min hold.
  • the modifier is kept at a constant 5% C (50:50 acetonitrile: water 0.1% formic acid) or D (50:50 acetonitrile: water 0.1% ammonium hydroxide (0.88 SG) depending on whether it is an acidic or basic method.
  • n-BuLi in hexanes (1.6 M; 16.1 mL, 25.8 mmol) was added dropwise via syringe pump over 40 minutes to a stirred solution of l,3-dibromo-2-methylbenzene (6.13 g, 24.5 mmol) in THF (60 mL) at -78 °C.
  • tert-butyl (R)-4-methyl-l,2,3-oxathiazolidine- 3-carboxylate 2,2-dioxide (6.40 g, 27.0 mmol) was added in portions and the reaction was stirred for 30 minutes before being allowed to warm to 0 °C over -45 minutes.
  • the reaction was stirred at 0 °C for 30 minutes and then treated with a aqueous citric acid (IN; 40 mL) and the mixture was stirred under these conditions for 15 minutes.
  • the mixture was diluted with EtOAc, the phases separated and the aqueous phase was extracted with EtOAc (2 x 25 mL).
  • the combined organic layers were washed with saturated aqueous sodium chloride, dried over sodium sulfate and evaporated.
  • the resultant residue was treated with HC1 in dioxane (4N; 30 mL), stirred for 45 minutes then concentrated under reduced pressure.
  • the resulting residue was dissolved in water (100 mL) and extracted with diethyl ether (2 x 25 mL).
  • 2,6-Lutidine (1.5 mL, 13 mmol) was added to a stirred solution of 2,2-difluoropropan-l-ol (1.0 g, 10 mmol) in DCM (50 mL) at - 10 °C.
  • Trifluoromethanesulfonic anhydride in DCM (1 M; 11 mL, 11 mmol) was then added dropwise via syringe over 10 minutes. The solution was allowed to stir at - 10 °C for 2 hours and then washed with cold aqueous HC1 (IN; 2 x 40 mL) and saturated aqueous sodium bicarbonate (2 x 20 mL).
  • Aqueous hydrogen chloride solution (1 M; 68.4 mL, 68.4 mmol) was added to (R)-N-(l-(3- ((diphenylmethylene)amino)-2-methylphenyl)propan-2-yl)-2,2-difluoropropan-l-amine (6.95 g, 17.1 mmol) in DCM (75 mL) and the biphasic mixture stirred rapidly for 1 hour. The layers were separated and the aqueous layer was extracted with DCM (2 x 40 mL). The aqueous layer was then basified by addition of saturated aqueous sodium carbonate and extracted with DCM (3 x 50 mL).
  • aqueous layer was then basified by addition of sodium carbonate and extracted with EtOAc (3 x 75 mL). The combined basic extracts were washed with saturated aqueous sodium chloride (50 mL), dried over magnesium sulfate, filtered and concentrated. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 50% EtOAc in heptane.
  • Cerium (IV) ammonium nitrate (0.932 g, 1.70 mmol) was added in portions to a solution of 7-(4-((E)-3-(iert-butoxy)-3-oxoprop- l-en-l-yl)-2,6-difluorophenyl)-6- (2-fluoro-2-methylpropyl)-5-methyl-4,5,6,7-tetrahydro- lH-pyrazolo[4,3-f]isoquinoline- l- carboxylate (0.510 g, 0.85 mmol) in acetonitrile (6.8 mL) and water (1.7 mL).
  • Methylmagnesium bromide (3 M in diethyl ether; 0.167 mL, 0.50 mmol) was added to a solution of the yellow solid (200 mg, 0.33 mmol) in THF (3.2 mL) at 0 °C. The reaction was stirred for 30 min and then warmed to room temperature over 30 min before being quenched by addition of methanol (1 mL). The volatiles were evaporated and the resulting residue was stirred in HC1 in dioxane (4N; 4 mL) at room temperature for 30 mins. The solution was passed through an SCX-2 column, eluting with methanol and then with NH 3 in MeOH (1M).
  • N,N-Diethyl- 1, 1, 2,3,3, 3-hexafluoropropan-l -amine 9.45 mL, 52.1 mmol was added dropwise to a solution of methyl (R)-3-hydroxy-2-methylpropanoate (4.81 mL, 43.4 mmol) in DCM (40 mL) and a slight exotherm was noted.
  • the reaction was stirred for 1 hour at room temperature and then stirred at 40 °C for 5 hours before cooling to room temperature overnight.
  • the reaction mixture was poured onto ice and the layers were separated. The aqueous layer was extracted with DCM (2 x 75 mL) and the combined organic layers were dried over sodium sulfate, filtered and carefully concentrated at reduced pressure.
  • the reaction mixture was then heated at 90 °C for 2.5 hours before being cooled and filtered through celite.
  • the filter cake was washed with DCM and the filtrate was concentrated under reduced pressure.
  • the resulting residue was dissolved in DCM (10 mL) and aqueous HC1 (IN; 10 mL) was added.
  • the biphasic mixture was stirred vigorously for 60 min.
  • DCM (20 mL) and water (20 mL) were added and layers were separated.
  • the aqueous layer was then basified by addition of aqueous NaOH (IN) and extracted with DCM (3 x 50 mL). The combined organic extracts were dried over sodium sulfate, filtered and concentrated under reduced pressure.
  • the biphasic mixture was stirred vigorously and neutralized to pH 7 by the slow addition of cold saturated aqueous sodium bicarbonate.
  • the phases were separated and the organic layer was washed with saturated aqueous sodium bicarbonate (2 x 25 mL) and saturated aqueous sodium chloride (25 mL).
  • the organic layer was then dried over magnesium sulfate, filtered and concentrated under reduced pressure.
  • the resulting residue was purified by flash silica chromatography, elution gradient 5 to 50% EtOAc in hexanes.
  • Ceric (IV) ammonium nitrate (2.24 g, 4.08 mmol) was added to a solution of tert-b tyl (65 , ,8R)-7-(2-fluoro-2-methylpropyl)-6-(2-methoxy-4-((E)-3-methoxy-3-oxoprop-l-en-l- yl)phenyl)-8-methyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline-3-carboxylate (1.00 g, 1.81 mmol, mixture of 2 isomers, -2: 1 ratio,) in acetonitrile (9.28 mL) and water (2.32 mL) and the reaction was stirred at room temperature for 2 hours.
  • Methylmagnesium bromide in diethyl ether (2.5 M; 2.90 mL, 7.25 mmol) was added and the reaction was maintained under these conditions for 30 mins before being quenched by addition of water and saturated aqueous NH 4 C1.
  • the reaction was extracted with DCM (x3) and the crude mixture was dried over sodium sulfate and concentrated to dryness.
  • the resulting residue was stirred in HC1 in dioxane (4N; 5 mL) at room temperature for 1 hour. The mixture was then passed through an SCX-2 column, eluting with MeOH and then NH3 in MeOH (IN) to liberate the product.
  • Aqueous sodium hydroxide (2N; 0.98 mL, 1.95 mmol) was added dropwise to a solution of methyl (E)-3-(4-((65,8R)-7-(2-fluoro-3-methoxy-2-methylpropyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylate (188 mg, 0.39 mmol) in a mixture of THF (2.0 mL) and MeOH (0.98 mL) at room temperature. The reaction was stirred at room temperature for 3 hours.
  • the reaction was then diluted with water (50 mL) and the pH was adjusted was to pH 4.6 by addition of aqueous HC1 (2N). The mixture was then extracted with EtOAc (x2). The combined organic layers were washed with saturated aqueous sodium chloride, dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 8% MeOH in DCM.
  • Lithium aluminum hydride (4.40 g, 116 mmol) was added portionwise to a solution of diethyl 2-fluoro-2-methylmalonate (8.56 g, 44.5 mmol) in THF (225 mL) at 0 °C over a period of 40 minutes while maintaining an internal temperature of less than 5 °C. The reaction was then warmed to room temperature and stirred for 100 minutes. The reaction mixture was cooled to 0 °C and then quenched by dropwise addition of water (4.4 mL). The mixture was allowed to warm to room temperature and was then stirred under these conditions for 18 hours.
  • N,N-Dimethylpyridin-4-amine (0.349 g, 2.86 mmol) was added to a solution of 2-fluoro-2- methylpropane-l,3-diol (3.09 g, 28.6 mmol) and N-ethyl-N-isopropylpropan-2-amine (7.60 mL, 42.9 mmol) in DCM (111 mL) at room temperature under nitrogen.
  • Trifluoromethanesulfonic anhydride (1.09 mL, 6.45 mmol) was added to a solution of ((2- fluoro-3-methoxy-2-methylpropoxy)methanetriyl)tribenzene (2.24 g, 6.15 mmol) in DCM (28.6 mL) and the reaction was stirred under these conditions for 30 minutes.
  • Triethylsilane (1.08 mL, 6.76 mmol) was added and the reaction was stirred for a further 30 minutes. Volatile components were then evaporated to afford crude 2-fluoro-3- methoxy-2-methylpropyl trifluoromethanesulfonate (1.52 g, 6.00 mmol) as a purple oil. This material was dissolved in 1,4-dioxane (15.6 mL) and (R)- l-(3-bromo-2- methylphenyl)propan-2-amine (1.37 g, 6.00 mmol) and diisopropylethylamine (1.56 mL, 9.00 mmol) were added.
  • Pd 2 (dba)3 (0.089 g, 0.11 mmol) and sodium ie/t-butoxide (0.63 g, 6.6 mmol) were added to a degassed suspension of N-((R)- l-(3-bromo-2-methylphenyl)propan-2-yl)-2-fluoro-3- methoxy-2-methylpropan- 1 -amine (1.45 g, 4.36 mmol), diphenylmethanimine (0.81 mL, 4.8 mmol) and 2,2'-bis(diphenylphosphanyl)-l, l'-binaphthalene (0.136 g, 0.22 mmol) in toluene (16.6 mL).
  • the reaction was heated at 90 °C for 3 hours. After cooling, the reaction mixture was diluted with EtOAc and washed with water. The aqueous layer was extracted with EtOAc and the combined organic layers were dried over magnesium sulfate and concentrated under reduced pressure. The resulting residue was dissolved in EtOAc (80 mL) and aqueous HC1 (2N; 80 mL) was added. The biphasic mixture was stirred vigourously for 18 hours. The layers were separated and the organic layer was extracted with aqueous HC1 (2N). The combined aqueous layers were extracted with EtOAc. The aqueous acid layer was purified by ion exchange chromatography, using an 50 g SCX-2 column.
  • Aqueous HC1 (2N; 100 mL) was added and the biphasic mixture was stirred vigorously for 30 minutes. The layers were separated and the organic layer was extracted with aqueous HC1 (2N). The combined aqueous layers were then extracted with EtOAc (x2). The aqueous acid layer was then purified by ion exchange chromatography, using an 50 g SCX-2 column that had been pre- treated with first methanol and then water. After introduction of the crude aqueous acid layer, the SCX column was washed with water and MeOH prior to elution of the desired product with NH 3 in MeOH (1M). Product fractions were concentrated to dryness to afford crude product (1.2 g).
  • the reaction mixture was stirred at -17 °C for a further 30 minutes and then diluted with ice-cold EtOAc (24 mL).
  • the reaction mixture was stirred vigorously and neutralised by dropwise addition of ice cold saturated aqueous sodium bicarbonate (31 mL). After stirring for 1 hour, the layers were separated and the organic layer was washed with saturated aqueous sodium bicarbonate (2 x 50 mL) and saturated aqueous sodium chloride (50 mL). The combined aqueous layers were extracted with EtOAc (2 x 50 mL) and the combined organic layers were dried over magnesium sulfate, filtered and concentrated under reduced pressure.
  • the reaction mixture was stirred at -17 °C for a further 30 minutes and then diluted with ice-cold EtOAc (30 mL).
  • the reaction mixture was stirred vigorously and neutralised by dropwise addition of ice cold saturated aqueous sodium bicarbonate (39 mL) and stirred for 1 hour.
  • the layers were separated and the organic layer was washed with saturated aqueous sodium bicarbonate (2 x 50 mL) and saturated aqueous sodium chloride (50 mL).
  • the combined aqueous layers were extracted with EtOAc (2 x 50 mL) and the combined organic layers were dried over magnesium sulfate, filtered and concentrated under reduced pressure.
  • Methyl (E)-3-(3-fluoro-4-((8R)-7-((5)-3-fluoro-2-methylpropyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-5-methoxyphenyl)acrylate (260 mg, 0.55 mmol) was dissolved in MeOH (1.5 mL), water (4.5 mL) and THF (4.5 mL). Lithium hydroxide (265 mg, 11.1 mmol) was then added. The reaction was stirred at room temperature for 1.5 hours and then the solvent was removed under reduced pressure.
  • the aqueous layer was extracted with EtOAc (3 x 15 mL) and the combined organic layers were dried over sodium sulfate, filtered and concentrated under reduced pressure to an orange residue. This residue was stored in the freezer for 18 hours and then dissolved in EtOAc (20 mL), treated with aqueous HC1 (IN; 12 mL) and the biphasic mixture was stirred vigorously for 20 minutes. The layers were separated and the organic layer was extracted with aqueous HC1 (IN; 2 x 7 mL). The combined aqueous layers were extracted with DCM (10 mL). The aqueous layer was then basified with sodium carbonate and extracted with DCM (3 x 15 mL).
  • the biphasic mixture was stirred vigorously and neutralised by slow addition of cold saturated aqueous sodium bicarbonate (50 mL) resulting in gas evolution.
  • the phases were separated and the organic layers were washed with saturated aqueous sodium bicarbonate (25 mL) and saturated aqueous sodium chloride (20 mL).
  • the organic layer was dried over magnesium sulfate, filtered and concentrated under reduced pressure.
  • Example 8 (E)-3-(4-((6 ⁇ ,8R)-7- ,2-difluoropropyl)-6,8-dimethyl-6,7,8,9-tetrahvdro- 3H-pyrazolor4,3-f1iso uinolin-6-yl)-3-fluoro-5-methoxyphenyl)acrylic acid
  • the resulting solid was treated with HCl in dioxane (4N; 0.24 mL, 0.98 mmol) and the mixture was stirred for another 1.5 hours before being concentrated under reduced pressure.
  • the resulting solid was partitioned between EtOAc and water and the pH was adjusted to ⁇ pH 5 with saturated aqueous sodium bicarbonate. The phases were separated and the aqueous phase was extracted with EtOAc (2 x 3 mL). The combined organic layers were washed with saturated aqueous sodium chloride, dried over magnesium sulfate, filtered and concentrated under reduced pressure.
  • the crude product was purified by flash silica chromatography, elution gradient 1 to 10% MeOH in DCM. Fractions containing the desired product were evaporated to dryness.
  • the vial was sealed and dioxane (0.5 mL) was added via syringe.
  • the mixture was stirred at ambient temperature for 2 minutes and then heated under microwave conditions at 100 °C for 30 minutes.
  • the reaction was heated in the microwave for a further 30 minutes at 125 °C before the reaction was cooled and dimethylacetamide (0.1 mL) plus further portions of dichloro [ ⁇ , ⁇ - bis(di- tert- butylphosphino)ferrocene]palladium(II) (1.7 mg, 2.6 ⁇ ) and tert-butyl acrylate (0.012 mL, 0.08 mmol) were added.
  • the mixture was sparged with argon for 5 minutes and again heated under microwave conditions at 125 °C for 30 minutes.
  • the cooled mixture was diluted with EtOAc and washed with saturated aqueous sodium chloride (x2).
  • the organic layer was dried over magnesium sulfate, filtered and concentrated under reduced pressure.
  • the crude product was purified by flash silica chromatography, elution gradient 0 to 30% EtOAc in hexanes.
  • Example 9 contains about 1.0% of ISOMER 2.
  • 3,4-Dihydro-2H-pyran (5.36 mL, 58.57 mmol) was added to a solution of 4-bromo- lH- indazole (5.77 g, 29.3 mmol) and 4-methylbenzenesulfonic acid (0.176 g, 1.02 mmol) in ethyl acetate (60 mL) and the mixture was heated at 70 °C for 16 hours. The mixture was cooled, added to saturated aqueous sodium bicarbonate (50 mL) and the phases were separated.
  • n-Butyllithium (1.6 M; 21.7 mL, 34.6 mmol) was added dropwise over 5 minutes to an oven-dried flask containing a solution of 4-bromo-l-(tetrahydro-2H-pyran-2-yl)-lH- indazole (9.28 g, 33.0 mmol) in THF (100 mL) at -78 °C. After stirring for 30 minutes, 2,2-dimethyloxirane (5.86 mL, 66.00 mmol) was added, followed by boron trifluoride diethyl etherate (4.07 mL, 33.0 mmol) dropwise.
  • the resulting solution was heated under reflux conditions with stirring for 3 hours. Further portions of iert-butyl acrylate (0.71 mL, 4.8 mmol) and l,l'-bis(di-tert-butylphosphino)ferrocene palladium dichloride (0.209 g, 0.32 mmol) were added and the reaction was stirred under reflux conditions for a further 1 hour. The reaction mixture was concentrated to dryness; the resulting residue was dissolved in EtOAc (150 mL) and aqueous citric acid (0.5 M; 100 mL). The mixture was filtered through a pad of celite and the organic layer was washed with water (100 mL).
  • the organic layer was dried over magnesium sulfate, filtered and adsorbed onto silica gel (5 g).
  • the resulting powder was purified by flash silica chromatography, elution gradient 10 to 25% (3: 1 EtOAc:EtOH) in heptane.
  • the reaction mixture was then diluted with EtOAc (150 mL) and water (50 mL) and the mixture was filtered through a pad of diatomaceous earth. The organic layer was dried over magnesium sulfate, filtered and adsorbed onto silica gel (3 g). The resulting powder was purified by flash silica chromatography, elution gradient 10 to 25% (3: 1 EtOAc:EtOH) in heptane.

Abstract

The specification relates to compounds of Formula (I) and pharmaceutically acceptable salts thereof. The specification also relates to processes and intermediates used for their preparation, pharmaceutical compositions containing them and their use in the treatment of cell proliferative disorders.

Description

INDAZOLE DERIVATIVES THAT DOWN-REGULATE THE ESTROGEN
RECEPTOR AND POSSESS ANTI-CANCER ACTIVITY
The specification relates to certain indazole compounds and pharmaceutically acceptable salts thereof that selectively down-regulate the estrogen receptor and possess anti-cancer activity. The specification also relates to use of said indazole compounds and pharmaceutically acceptable salts thereof in methods of treatment of the human or animal body, for example in prevention or treatment of cancer. The specification also relates to processes and intermediate compounds involved in the preparation of said indazole compounds and to pharmaceutical compositions containing them.
Estrogen receptor alpha (ERcc, ESR1, NR3A) and estrogen receptor beta (ER , ESR2, NR3b) are steroid hormone receptors which are members of the large nuclear receptor family. Structured similarly to all nuclear receptors, ERcc is composed of six functional domains (named A-F) (Dahlman- Wright, et al., Pharmacol. Rev., 2006, 58:773- 781) and is classified as a ligand-dependent transcription factor because after its association with the specific ligand, (the female sex steroid hormone 17b estradiol (E2)), the complex binds to genomic sequences, named Estrogen Receptor Elements (ERE) and interacts with co-regulators to modulate the transcription of target genes. The ERcc gene is located on 6q25.1 and encodes a 595AA protein and multiple isoforms can be produced due to alternative splicing and translational start sites. In addition to the DNA binding domain (Domain C) and the ligand binding domain (Domain E) the receptor contains a N- terminal (A/B) domain, a hinge (D) domain that links the C and E domains and a C- terminal extension (F domain). While the C and E domains of ERcc and ER are quite conserved (96% and 55% amino acid identity respectively) conservation of the A/B, D and F domains is poor (below 30% amino acid identity). Both receptors are involved in the regulation and development of the female reproductive tract and in addition play roles in the central nervous system, cardiovascular system and in bone metabolism. The genomic action of ERs occurs in the nucleus of the cell when the receptor binds EREs directly (direct activation or classical pathway) or indirectly (indirect activation or non-classical pathway). In the absence of ligand, ERs are associated with heat shock proteins, Hsp90 and Hsp70, and the associated chaperone machinery stabilizes the ligand binding domain (LBD) making it accessible to ligand. Liganded ER dissociates from the heat shock proteins leading to a conformational change in the receptor that allows dimerisation, DNA binding, interaction with co-activators or co-repressors and modulation of target gene expression. In the non-classical pathway, AP-1 and Sp-1 are alternative regulatory DNA sequences used by both isoforms of the receptor to modulate gene expression. In this example, ER does not interact directly with DNA but through associations with other DNA bound transcription factors e.g. c-Jun or c-Fos (Kushner et al., Pure Applied Chemistry 2003, 75: 1757-1769). The precise mechanism whereby ER affects gene transcription is poorly understood but appears to be mediated by numerous nuclear factors that are recruited by the DNA bound receptor. The recruitment of co-regulators is primarily mediated by two protein surfaces, AF2 and AF1 which are located in E-domain and the A/B domain respectively. AF1 is regulated by growth factors and its activity depends on the cellular and promoter environment whereas AF2 is entirely dependent on ligand binding for activity. Although the two domains can act independently, maximal ER transcriptional activity is achieved through synergistic interactions via the two domains (Tzukerman, et al., Mol. Endocrinology, 1994, 8:21-30). Although ERs are considered transcription factors they can also act through non-genomic mechanisms as evidenced by rapid ER effects in tissues following E2 administration in a timescale that is considered too fast for a genomic action. It is still unclear if receptors responsible for the rapid actions of estrogen are the same nuclear ERs or distinct G-protein coupled steroid receptors (Warner, et al., Steroids 2006 71_:91-95) but an increasing number of E2 induced pathways have been identified e.g. MAPK/ERK pathway and activation of endothelial nitric oxide synthase and PI3K/Akt pathway. In addition to ligand dependent pathways, ERcc has been shown to have ligand independent activity through AF-1 which has been associated with stimulation of MAPK through growth factor signalling e.g. insulin like growth factor 1 (IGF-1) and epidermal growth factor (EGF). Activity of AF-1 is dependent on
phosphorylation of Serl 18 and an example of cross-talk between ER and growth factor signalling is the phosphorylation of Ser 118 by MAPK in response to growth factors such as IGF-1 and EGF (Kato, et al., Science, 1995, 270: 1491-1494).
A large number of structurally distinct compounds have been shown to bind to ER. Some compounds such as endogenous ligand E2, act as receptor agonists whereas others competitively inhibit E2 binding and act as receptor antagonists. These compounds can be divided into 2 classes depending on their functional effects. Selective estrogen receptor modulators (SERMs) such as tamoxifen have the ability to act as both receptor agonists and antagonists depending on the cellular and promoter context as well as the ER isoform targeted. For example tamoxifen acts as an antagonist in breast but acts as a partial agonist in bone, the cardiovascular system and uterus. All SERMs appear to act as AF2 antagonists and derive their partial agonist characteristics through AFl. A second group, fulvestrant being an example, are classified as full antagonists and are capable of blocking estrogen activity via the complete inhibition of AFl and AF2 domains through induction of a unique conformation change in the ligand binding domain (LBD) on compound binding which results in complete abrogation of the interaction between helix 12 and the remainder of the LBD, blocking co-factor recruitment (Wakeling, et al., Cancer Res., 1991, 51:3867-3873; Pike, et al., Structure, 2001, 9: 145-153).
Intracellular levels of ERa are down-regulated in the presence of E2 through the ubiquitin/proteosome (Ub/26S) pathway. Polyubiquitinylation of liganded ERa is catalysed by at least three enzymes; the ubiquitin- activating enzyme El activated ubiquitin is conjugated by E2 with lysine residues through an isopeptide bond by E3 ubiquitin ligase and polyubiquitinated ERa is then directed to the proteosome for degradation. Although ER-dependent transcription regulation and proteosome-mediated degradation of ER are linked (Lonard, et al., Mol. Cell, 2000 5:939-948), transcription in itself is not required for ERa degradation and assembly of the transcription initiation complex is sufficient to target ERa for nuclear proteosomal degradation. This E2 induced degradation process is believed to necessary for its ability to rapidly activate transcription in response to requirements for cell proliferation, differentiation and metabolism (Stenoien, et al., Mol. Cell Biol., 2001, 21:4404-4412). Fulvestrant is also classified as a selective estrogen receptor down- regulator (SERD), a subset of antagonists that can also induce rapid down-regulation of ERa via the 26S proteosomal pathway. In contrast a SERM such as tamoxifen can increase ERa levels although the effect on transcription is similar to that seen for a SERD.
Approximately 70% of breast cancers express ER and/or progesterone receptors implying the hormone dependence of these tumour cells for growth. Other cancers such as ovarian and endometrial are also thought to be dependent on ERa signalling for growth. Therapies for such patients can inhibit ER signalling either by antagonising ligand binding to ER e.g. tamoxifen which is used to treat early and advanced ER positive breast cancer in both pre and post menopausal setting; antagonising and down-regulating ERa e.g. fulvestrant which is used to treat breast cancer in women which have progressed despite therapy with tamoxifen or aromatase inhibitors; or blocking estrogen synthesis e.g.
aromatase inhibitors which are used to treat early and advanced ER positive breast cancer. Although these therapies have had an enormously positive impact on breast cancer treatment, a considerable number of patients whose tumours express ER display de novo resistance to existing ER therapies or develop resistance to these therapies over time. Several distinct mechanisms have been described to explain resistance to first-time tamoxifen therapy which mainly involve the switch from tamoxifen acting as an antagonist to an agonist, either through the lower affinity of certain co-factors binding to the tamoxifen-ERa complex being off-set by over-expression of these co-factors, or through the formation of secondary sites that facilitate the interaction of the tamoxifen-ERa complex with co-factors that normally do not bind to the complex. Resistance could therefore arise as a result of the outgrowth of cells expressing specific co-factors that drive the tamoxifen-ERa activity. There is also the possibility that other growth factor signalling pathways directly activate the ER receptor or co-activators to drive cell proliferation independently of ligand signalling.
More recently, mutations in ESR1 have been identified as a possible resistance mechanism in metastatic ER-positive patient derived tumour samples and patient-derived xenograft models (PDX) at frequencies varying from 17-25%. These mutations are predominantly, but not exclusively, in the ligand-binding domain leading to mutated functional proteins; examples of the amino acid changes include Ser463Pro, Val543Glu, Leu536Arg, Tyr537Ser, Tyr537Asn and Asp538Gly, with changes at amino acid 537 and 538 constituting the majority of the changes currently described. These mutations have been undetected previously in the genomes from primary breast samples characterised in the Cancer Genome Atlas database. Of 390 primary breast cancer samples positive for ER expression not a single mutation was detected in ESR1 (Cancer Genome Atlas Network, 2012 Nature 490: 61-70). The ligand binding domain mutations are thought to have developed as a resistance response to aromatase inhibitor endocrine therapies as these mutant receptors show basal transcriptional activity in the absence of estradiol. The crystal structure of ER, mutated at amino acids 537 and 538, showed that both mutants favoured the agonist conformation of ER by shifting the position of helix 12 to allow co-activator recruitment and thereby mimicking agonist activated wild type ER. Published data has shown that endocrine therapies such as tamoxifen and fulvestrant can still bind to ER mutant and inhibit transcriptional activation to some extent and that fulvestrant is capable of degrading Try537Ser but that higher doses may be needed for full receptor inhibition (Toy et al., Nat. Genetics 2013, 45: 1439-1445; Robinson et al., Nat. Genetics 2013, 45: 144601451; Li, S. et al. Cell Rep. 4, 1116-1130 (2013). It is therefore feasible that certain compounds of the Formula (I) or pharmaceutically acceptable salts thereof (as described hereinafter) will be capable of down-regulating and antagonising mutant ER although it is not known at this stage whether ESR1 mutations are associated with an altered clinical outcome.
Regardless of which resistance mechanism or combination of mechanisms takes place, many are still reliant on ER-dependent activities and removal of the receptor through a SERD mechanism offers the best way of removing the ERcc receptor from the cell.
Fulvestrant is currently the only SERD approved for clinical use, yet despite its
mechanistic properties, the pharmacological properties of the drug have limited its efficacy due to the current limitation of a 500mg monthly dose which results in less than 50% turnover of the receptor in patient samples compared to the complete down-regulation of the receptor seen in in vitro breast cell line experiments (Wardell, et al., Biochem. Pharm., 2011, 82: 122-130). Hence there is a need for new ER targeting agents that have the required pharmaceutical properties and SERD mechanism to provide enhanced benefit in the early, metastatic and acquired resistance setting.
The compounds of the specification have been found to possess potent anti-tumour activity, being useful in inhibiting the uncontrolled cellular proliferation which arises from malignant disease. The compounds of the specification provide an anti-tumour effect by, as a minimum, acting as SERDs.
According to one aspect of the specification there is provided a compound of the Formula (I):
Figure imgf000007_0001
or a pharmaceutically acceptable salt thereof, wherein:
R1 is fluoro or methoxy;
R2, R3 and R4 are each independently hydrogen or fluoro;
R5 is hydrogen or methyl;
R6 is hydrogen or methyl;
R7 is methyl, CHF2 or cyclopropyl;
R8 is hydrogen or methyl;
R9 is hydrogen, fluoro, CH2OH, CH2OMe, CH2F or CHF2;
R10 is hydrogen, methyl or fluoro; and
R11 is hydrogen, methyl, fluoro or CH2F; or
R10 and R11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring.
In one embodiment there is provided a compound of Formula (I) as defined above.
In one embodiment there is provided a pharmaceutically acceptable salt of a compound of Formula (I).
The compounds of Formula (I) have two or more chiral centres and it will be recognised that the compound of Formula (I) may be prepared, isolated and/or supplied with or without the presence, in addition, of one or more of the other possible enantiomeric and/or diastereomeric isomers of the compound of Formula (I) in any relative proportions. The preparation of enantioenriched/ enantiopure and/or diastereoenriched/ diastereopure compounds may be carried out by standard techniques of organic chemistry that are well known in the art, for example by synthesis from enantioenriched or enantiopure starting materials, use of an appropriate enantioenriched or enantiopure catalyst during synthesis, and/or by resolution of a racemic or partially enriched mixture of stereoisomers, for example via chiral chromatography.
For use in a pharmaceutical context it may be preferable to provide a compound of Formula (I) or a pharmaceutically acceptable salt thereof without large amounts of the other stereoisomeric forms being present.
Accordingly, in one embodiment there is provided a composition comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof, optionally together with one or more of the other stereoisomeric forms of the compound of Formula (I) or pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) or pharmaceutically acceptable salt thereof is present within the composition with a diastereomeric excess (%de) of > 90%.
In a further embodiment the %de in the above-mentioned composition is > 95%.
In a further embodiment the %de in the above-mentioned composition is > 98%.
In a further embodiment the %de in the above-mentioned composition is > 99%.
In a further embodiment there is provided a composition comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof, optionally together with one or more of the other stereoisomeric forms of the compound of Formula (I) or
pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) or pharmaceutically acceptable salt thereof is present within the composition with an enantiomeric excess (%ee) of > 90%.
In a further embodiment the %ee in the above-mentioned composition is > 95%.
In a further embodiment the %ee in the above-mentioned composition is > 98%.
In a further embodiment the %ee in the above-mentioned composition is > 99%.
In a further embodiment there is provided a composition comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof, optionally together with one or more of the other stereoisomeric forms of the compound of Formula (I) or
pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) or pharmaceutically acceptable salt thereof is present within the composition with an enantiomeric excess (%ee) of > 90% and a diastereomeric excess (%de) of > 90%. In further embodiments of the above-mentioned composition the %ee and %de may take any combination of values as listed below:
• The %ee is <5% and the %de is > 80%.
• The %ee is <5% and the %de is > 90%.
• The %ee is <5% and the %de is > 95%.
• The %ee is <5% and the %de is > 98%.
• The %ee is > 95% and the %de is > 95%.
• The %ee is > 98% and the %de is > 98%.
• The %ee is > 99% and the %de is > 99%.
In a further embodiment there is provided a pharmaceutical composition which comprises a compound of the Formula (I) or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable diluent or carrier.
In one embodiment there is provided a pharmaceutical composition which comprises a compound of the Formula (I) or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable diluent or carrier, optionally further comprising one or more of the other stereoisomeric forms of the compound of Formula (I) or pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) or pharmaceutically acceptable salt thereof is present within the composition with an enantiomeric excess (%ee) of > 90%.
In a further embodiment the %ee in the above-mentioned composition is > 95%.
In a further embodiment the %ee in the above-mentioned composition is > 98%.
In a further embodiment the %ee in the above-mentioned composition is > 99%.
In one embodiment there is provided a pharmaceutical composition which comprises a compound of the Formula (I) or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable diluent or carrier, optionally further comprising one or more of the other stereoisomeric forms of the compound of Formula (I) or pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) or pharmaceutically acceptable salt thereof is present within the composition with a diastereomeric excess (%de) of > 90%.
In a further embodiment the %de in the above-mentioned composition is > 95%.
In a further embodiment the %de in the above-mentioned composition is > 98%. In a further embodiment the %de in the above-mentioned composition is > 99%.
In one embodiment there is provided a pharmaceutical composition which comprises a compound of the Formula (I) or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable diluent or carrier, optionally further comprising one or more of the other stereoisomeric forms of the compound of Formula (I) or pharmaceutically acceptable salt thereof, wherein the compound of Formula (I) or pharmaceutically acceptable salt thereof is present within the composition with an enantiomeric excess (%ee) of > 90% and a diastereomeric excess (%de) of > 90%.
In further embodiments of the above-mentioned pharmaceutical composition the %ee and %de may take any combination of values as listed below:
• The %ee is > 95% and the %de is > 95%.
• The %ee is > 98% and the %de is > 98%.
• The %ee is > 99% and the %de is > 99%.
The compounds of Formula (I) and pharmaceutically acceptable salts thereof may be prepared, used or supplied in amorphous form, crystalline form, or semicrystalline form and any given compound of Formula (I) or pharmaceutically acceptable salt thereof may be capable of being formed into more than one crystalline / polymorphic form, including hydrated (e.g. hemi-hydrate, a mono-hydrate, a di-hydrate, a tri-hydrate or other stoichiometry of hydrate) and/or solvated forms. It is to be understood that the present specification encompasses any and all such solid forms of the compound of Formula (I) and pharmaceutically acceptable salts thereof.
In one embodiment there is provided a compound of Formula (I):
Figure imgf000010_0001
or a pharmaceutically acceptable salt thereof, wherein:
R1 is fluoro or methoxy;
R2, R3 and R4 are each independently hydrogen or fluoro;
R5 is hydrogen or methyl;
R6 is hydrogen or methyl;
R7 is methyl, CHF2 or cyclopropyl;
R8 is hydrogen or methyl;
R9 is hydrogen, fluoro, CH2OH, CH2OMe, CH2F or CHF2;
R10 is hydrogen, methyl or fluoro; and
R11 is hydrogen, methyl, fluoro or CH2F; or
R10 and R11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring;
provided that the compound of formula (I) is not:
(E)-3-[3,5-difluoro-4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydro- pyrazolo[4,3-/]isoquinolin-6-yl]phenyl]prop-2-enoic acid;
(E)-3-[3,5-difluoro-4-[7-(2-fluoro-2-methyl-propyl)-6,8-dimethyl-8,9-dihydro-3H- pyrazolo[4,3-/]isoquinolin-6-yl]phenyl]prop-2-enoic acid;
(E)-3-[4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoic acid;
(£T)-3-[4-[7-(2,2-difluoropropyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3-/]isoquinolin-6- yl]-3-methoxy-phenyl]prop-2-enoic acid;
(E)-3-[3-fluoro-4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-5-methoxy-phenyl]prop-2-enoic acid; or
(E)-3-[4-[7-(2,2-difluoropropyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3-/]isoquinolin-6- yl]-3-fluoro-5-methoxy-phenyl]prop-2-enoic acid;
or enantiomeric or diastereomeric isomers thereof.
In one embodiment R1 is methoxy.
In one embodiment R1 is fluoro.
In one embodiment R4 is fluoro.
In one embodiment R4 is hydrogen.
In one embodiment R1 is methoxy and R4 is fluoro.
In one embodiment R1 is methoxy and R4 is hydrogen. In one embodiment R1 is fluoro and R4 is fluoro.
In one embodiment R2 is hydrogen.
In one embodiment R2 is fluoro.
In one embodiment R3 is hydrogen.
In one embodiment R3 is fluoro.
In one embodiment R2 is hydrogen and R3 is hydrogen.
In one embodiment R1 is methoxy and R2 is fluoro.
In one embodiment R1 is methoxy and R3 is fluoro.
In one embodiment R1 is methoxy, R2 is hydrogen, R3 is hydrogen and R4 is fluoro.
In one embodiment R1 is methoxy and R2, R3 and R4 are hydrogen.
In one embodiment R1 is fluoro, R2 is hydrogen, R3 is hydrogen and R4 is fluoro.
In one embodiment R5 is hydrogen.
In one embodiment R5 is methyl.
In one embodiment R6 is hydrogen.
In one embodiment R6 is methyl.
In one embodiment R7 is methyl or CHF2.
In one embodiment R7 is methyl.
In one embodiment R6 is hydrogen and R7 is methyl.
In one embodiment R6 is hydrogen and R7 is CHF2.
In one embodiment R6 is methyl and R7 is methyl.
In one embodiment R8 is hydrogen.
In one embodiment R9 is fluoro.
In one embodiment R9 is CH2OH, CH2OMe, CH2F or CHF2 and R10 is methyl.
In one embodiment R9 is CH2OH, CH2OMe, CH2F or CHF2 and R10 is fluoro.
In one embodiment R9 is CH2OH, CH2OMe, CH2F or CHF2 and R10 and R11 taken together with the carbon atom to which they are attached form a cyclopropyl ring. In one embodiment the group -CH(R8)-C(R9)(R10)(Rn) in the compound of Formula (I) is selected from the group consisting of:
In another embodiment the group -CH(R8)-C(R9)(R10)(Rn) in the compound of Formula (I) is selected from the group consisting of:
Figure imgf000013_0002
In one embodiment the group -CH(R8)-C(R9)(R10)(Rn) in the compound of Formula (I) is selected from the group consisting of:
Figure imgf000014_0001
In another embodiment the group -CH(R8)-C(R9)(R10)(Rn) in the compound of Formula (I) is selected from the group consisting of:
Figure imgf000014_0002
In one embodiment there is provided a compound of Formula (I) or a pharmaceutically acceptable salt thereof, wherein;
R1 is methoxy;
R2 and R3 are hydrogen;
R4 is hydrogen or fluoro;
R5 is hydrogen or methyl;
R6 is hydrogen or methyl;
R7 is methyl, CHF2 or cyclopropyl;
R8 is hydrogen or methyl;
R9 is hydrogen, fluoro, CH2OH, CH2OMe, CH2F or CHF2;
R10 is hydrogen, methyl or fluoro; and
R11 is hydrogen, methyl, fluoro or CH2F; or R10 and R11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring.
In one embodiment there is provided a compound of Formula (I) or a
pharmaceutically acceptable salt thereof, wherein;
R1 is methoxy;
R2 and R3 are hydrogen;
R4 is hydrogen or fluoro;
R5 is hydrogen or methyl;
R6 is hydrogen or methyl;
R7 is methyl, CHF2 or cyclopropyl;
R8 is hydrogen or methyl;
R9 is hydrogen, fluoro, CH2OH, CH2OMe, CH2F or CHF2;
R10 is hydrogen, methyl or fluoro; and
R11 is hydrogen, methyl, fluoro or CH2F; or
R10 and R11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring;
provided that the compound of formula (I) is not:
(E)-3-[4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoic acid;
(E)-3-[4-[7-(2,2-difluoropropyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3-/]isoquinolin-6- yl]-3-methoxy-phenyl]prop-2-enoic acid;
(E)-3-[3-fluoro-4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-5-methoxy-phenyl]prop-2-enoic acid; or
(E)-3-[4-[7-(2,2-difluoropropyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3-/]isoquinolin-6- yl]-3-fluoro-5-methoxy-phenyl]prop-2-enoic acid;
or enantiomeric or diastereomeric isomers thereof.
In one embodiment there is provided a compound of Formula (I) or a
pharmaceutically acceptable salt thereof, wherein;
R1 is methoxy;
R2, R3 and R4 are hydrogen;
R5 is hydrogen or methyl;
R6 is hydrogen or methyl; R7 is methyl, CHF2 or cyclopropyl;
R8 is hydrogen or methyl;
R9 is hydrogen, fluoro, CH2OH, CH2OMe, CH2F or CHF2;
R10 is hydrogen, methyl or fluoro; and
R11 is hydrogen, methyl, fluoro or CH2F; or
R10 and R11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring.
In one embodiment there is provided a compound of Formula (I) or a
pharmaceutically acceptable salt thereof, wherein;
R1 is methoxy;
R2, R3 and R4 are hydrogen;
R5 is hydrogen or methyl;
R6 is hydrogen or methyl;
R7 is methyl, CHF2 or cyclopropyl;
R8 is hydrogen or methyl;
R9 is hydrogen, fluoro, CH2OH, CH2OMe, CH2F or CHF2;
R10 is hydrogen, methyl or fluoro; and
R11 is hydrogen, methyl, fluoro or CH2F; or
R10 and R11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring;
provided that the compound of Formula (I) is not:
(E)-3-[4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoic acid;
(E)-3-[4-[7-(2,2-difluoropropyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3-/]isoquinolin-6- yl]-3-methoxy-phenyl]prop-2-enoic acid;
or enantiomeric or diastereomeric isomers thereof.
In one embodiment there is provided a compound of Formula (I) or a
pharmaceutically acceptable salt thereof, wherein;
R1 is methoxy;
R2 and R3 are hydrogen;
R4 is hydrogen or fluoro;
R5 is methyl; R6 is hydrogen or methyl;
R7 is methyl, CHF2 or cyclopropyl;
R8 is hydrogen or methyl;
R9 is hydrogen, fluoro, CH2OH, CH2OMe, CH2F or CHF2;
R10 is hydrogen, methyl or fluoro; and
R11 is hydrogen, methyl, fluoro or CH2F; or
R10 and R11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring.
In one embodiment there is provided a compound of Formula (I) or a
pharmaceutically acceptable salt thereof, wherein;
R1 is fluoro;
R2 and R3 are hydrogen;
R4 is fluoro;
R5 is hydrogen or methyl;
R6 is hydrogen or methyl;
R7 is methyl, CHF2 or cyclopropyl;
R8 is hydrogen or methyl;
R9 is hydrogen, fluoro, CH2OH, CH2OMe, CH2F or CHF2;
R10 is hydrogen, methyl or fluoro; and
R11 is hydrogen, methyl, fluoro or CH2F; or
R10 and R11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring;
provided that the compound of formula (I) is not:
(E)-3-[3,5-difluoro-4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydro- pyrazolo[4,3-/]isoquinolin-6-yl]phenyl]prop-2-enoic acid;
(E)-3-[3,5-difluoro-4-[7-(2-fluoro-2-methyl-propyl)-6,8-dimethyl-8,9-dihydro-3H- pyrazolo[4,3-/]isoquinolin-6-yl]phenyl]prop-2-enoic acid;
or enantiomeric or diastereomeric isomers thereof.
In another aspect of the application there is provided a compound of Formula (IA):
Figure imgf000018_0001
R5 is hydrogen or methyl;
R6 is hydrogen or methyl;
R7 is methyl, CHF2 or cyclopropyl;
R8 is hydrogen or methyl;
R9 is hydrogen, fluoro, CH2OH, CH2OMe, CH2F or CHF2;
R10 is hydrogen, methyl or fluoro; and
R11 is hydrogen, methyl, fluoro or CH2F; or
R10 and R11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring;
provided that the compound of formula (IA) is not:
(E)-3-[3,5-difluoro-4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydro- pyrazolo[4,3-/]isoquinolin-6-yl]phenyl]prop-2-enoic acid;
(E)-3-[3,5-difluoro-4-[7-(2-fluoro-2-methyl-propyl)-6,8-dimethyl-8,9-dihydro-3H- pyrazolo[4,3-/]isoquinolin-6-yl]phenyl]prop-2-enoic acid;
(E)-3-[4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoic acid; (E)-3-[4-[7-(2,2-difluoropropyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- |isoquinolin-6- yl]-3-methoxy-phenyl]prop-2-enoic acid;
(E)-3-[3-fluoro-4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-5-methoxy-phenyl]prop-2-enoic acid; or
(i¾-3-[4-[7-(2,2-difluoropropyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- |isoquinolin-6- yl]-3-fluoro-5-methoxy-phenyl]prop-2-enoic acid;
or enantiomeric or diastereomeric isomers thereof.
In one embodiment there is provided a compound of Formula (IA) or a
pharmaceutically acceptable salt thereof, wherein:
Ring Y is selected from:
Figure imgf000019_0001
R5 is hydrogen or methyl;
R6 is hydrogen or methyl;
R7 is methyl, CHF2 or cyclopropyl;
R8 is hydrogen or methyl;
R9 is hydrogen, fluoro, CH2OH, CH2OMe, CH2F or CHF2;
R10 is hydrogen, methyl or fluoro; and
R11 is hydrogen, methyl, fluoro or CH2F; or
R10 and R11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring.
In one embodiment there is provided a compound of Formula (IA) or a
pharmaceutically acceptable salt thereof, wherein:
Ring Y is selected from:
Figure imgf000019_0002
R5 is hydrogen or methyl;
R6 is hydrogen or methyl;
R7 is methyl, CHF2 or cyclopropyl; R is hydrogen or methyl;
R9 is hydrogen, f uoro, CH2OH, CH2OMe, CH2F or CHF2;
R10 is hydrogen, methyl or fluoro; and
R11 is hydrogen, methyl, fluoro or CH2F; or
R10 and R11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring;
provided that the compound of formula (IA) is not:
(E)-3-[4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoic acid;
(E)-3-[4-[7-(2,2-difluoropropyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3-/]isoquinolin-6- yl]-3-methoxy-phenyl]prop-2-enoic acid;
(E)-3-[3-fluoro-4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-5-methoxy-phenyl]prop-2-enoic acid; or
(E)-3-[4-[7-(2,2-difluoropropyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3-/]isoquinolin-6- yl]-3-fluoro-5-methoxy-phenyl]prop-2-enoic acid;
or enantiomeric or diastereomeric isomers thereof.
g Y in the compound of Formula (IA) is selected from:
Figure imgf000020_0001
In another aspect of the application there is provided a compound of Formula (IB)
Figure imgf000020_0002
(IB)
or a pharmaceutically acceptable salt thereof, wherein: R5 is hydrogen or methyl;
R6 is hydrogen or methyl;
R7 is methyl, CHF2 or cyclopropyl; and
R12 is selected from the group consisting
Figure imgf000021_0001
In one embodiment there is provided a compound of Formula (IB), or a pharmaceutically acceptable salt thereof, wherein;
R5 is hydrogen or methyl;
R6 is hydrogen and R7 is CHF2 or cyclopropyl; or
R6 is methyl and R7 is methyl; and
R12 is selected from the group consisting of:
Figure imgf000021_0002
In one embodiment there is provided a compound of Formula (IB), or a pharmaceutically acceptable salt thereof, wherein;
R5 is hydrogen or methyl;
R6 is hydrogen or methyl;
R7 is methyl, CHF2 or cyclopropyl; and
Figure imgf000022_0001
In another aspect of the application there is provided a compound of Formula (IC)
Figure imgf000022_0002
(IC)
or a pharmaceutically acceptable salt thereof, wherein:
R4 is hydrogen or fluoro;
R5 is hydrogen or methyl;
R6 is hydrogen or methyl;
R7 is methyl, CHF2 or cyclopropyl; and
R12 is selected from the group consisting of:
Figure imgf000023_0001
provided that when R5 is h dro en, R6 is hydrogen and R7 is methyl, then R12 is not:
Figure imgf000023_0002
In one embodiment there is provided a compound of Formula (IC), or a pharmaceutically acceptable salt thereof, wherein;
R4 is hydrogen or fluoro;
R5 is hydrogen or methyl;
R6 is hydrogen or methyl;
R7 is methyl, CHF2 or cyclopropyl; and
R12 is selected from the group consisting of:
Figure imgf000023_0003
In one embodiment there is provided a compound of Formula (IC), or a
pharmaceutically acceptable salt thereof, wherein;
R4 is hydrogen or fluoro;
R5 is hydrogen or methyl;
R6 is hydrogen;
R7 is methyl; and
R12 is selected from the group consisting of:
Figure imgf000024_0001
In another aspect of the application there is provided a compound of Formula (ID):
Figure imgf000024_0002
wherein:
R4 is hydrogen or fluoro; R6 is hydrogen or methyl;
R7 is methyl, CHF2 or cyclopropyl; and
R12 is selected from the group consisting of:
Figure imgf000025_0001
In one embodiment of the application there is provided a compound of Formula (I), or a pharmaceutically acceptable salt thereof, selected from:
(E)-3-(4-((6S,8R)-7-((S)-3-fluoro-2-methylpropyl)-6,8-dimethyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylic acid;
(E)-3-(4-((6S,8R)-7-(2,3-difluoro-2-methylpropyl)-8-methyl-6,7,8,9-tetrahydro- 3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylic acid;
(E)-3-(4-((6S,8R)-7-(3-fluoro-2-(methoxymethyl)propyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylic acid;
(E)-3-(4-((6S,8R)-7-(2,3-difluoro-2-(methoxymethyl)propyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylic acid;
(E)-3-(4-((6S,8R)-7-((l-(fluoromethyl)cyclopropyl)methyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylic acid;
(E)-3-(3-methoxy-4-((6S,8R)-7-((l-(methoxymethyl)cyclopropyl)methyl)-8- methyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)phenyl)acrylic acid;
(E)-3-(4-((65,8R)-7-(2,2-difluoropropyl)-8-methyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3,5-difluorophenyl)acrylic acid;
(E)-3-(3,5-difluoro-4-((65,8R)-7-(2-fluoro-2-methylpropyl)-6,8-dimethyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)phenyl)acrylic acid; (E)-3-(4-((65,8R)-7-((5)-3-fluoro-2-methylpropyl)-8-methyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylic acid;
(E)-3-(4-((65,8R)-7-(2-fluoro-2-methylpropyl)-6,8-dimethyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylic acid;
(E)-3-(4-((65,8R)-7-(2-fluoro-3-methoxy-2-methylpropyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f|isoquinolin-6-yl)-3-methoxyphenyl)acrylic acid;
(E)-3-(3-fluoro-4-((65,8R)-7-((5)-3-fluoro-2-methylpropyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f|isoquinolin-6-yl)-5-methoxyphenyl)acrylic acid;
(E)-3-(4-((65,8R)-7-(2,2-difluoropropyl)-6,8-dimethyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3-fluoro-5-methoxyphenyl)acrylic acid;
(E)-3-(3-fluoro-4-(7-((5)-3-fluoro-2-methylpropyl)-8,8-dimethyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f|isoquinolin-6-yl)-5-methoxyphenyl)acrylic acid;
(E)-3-(4-((65,85)-8-(difluoromethyl)-7-(2-fluoro-2-methylpropyl)-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f|isoquinolin-6-yl)-3,5-difluorophenyl)acrylic acid; and
(E)-3-(4-((65,85)-8-(difluoromethyl)-7-(2-fluoro-2-methylpropyl)-6-methyl- 6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3,5-difluorophenyl)acrylic acid.
In another embodiment of the application there is provided a compound of Formula (I), or a pharmaceutically acceptable salt thereof, selected from:
(E)-3-(4-((65,8R)-7-(2,2-difluoropropyl)-8-methyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3,5-difluorophenyl)acrylic acid;
(E)-3-(3,5-difluoro-4-((65,8R)-7-(2-fluoro-2-methylpropyl)-6,8-dimethyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)phenyl)acrylic acid;
(E)-3-(4-((65,8R)-7-((5)-3-fluoro-2-methylpropyl)-8-methyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylic acid;
(E)-3-(4-((65,8R)-7-(2-fluoro-2-methylpropyl)-6,8-dimethyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylic acid;
(E)-3-(4-((65,8R)-7-(2-fluoro-3-methoxy-2-methylpropyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylic acid;
(E)-3-(3-fluoro-4-((65,8R)-7-((5)-3-fluoro-2-methylpropyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-5-methoxyphenyl)acrylic acid;
(E)-3-(4-((65,8R)-7-(2,2-difluoropropyl)-6,8-dimethyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3-fluoro-5-methoxyphenyl)acrylic acid; (E)-3-(3-fluoro-4-(7-((5)-3-fluoro-2-methylpropyl)-8,8-dimethyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-5-methoxyphenyl)acrylic acid;
(E)-3-(4-((65,85)-8-(difluoromethyl)-7-(2-fluoro-2-methylpropyl)-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3,5-difluorophenyl)acrylic acid; and
(E)-3-(4-((65,85)-8-(difluoromethyl)-7-(2-fluoro-2-methylpropyl)-6-methyl- 6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3,5-difluorophenyl)acrylic acid.
In another embodiment of the application there is provided a compound of Formula (I), or a pharmaceutically acceptable salt thereof, selected from:
(E)-3-[4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoic acid;
(E)-3-[4-[7-(2,2-difluoropropyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoic acid;
(E)-3-[3-fluoro-4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9- tetrahydropyrazolo[4,3-/]isoquinolin-6-yl]-5-methoxy-phenyl]prop-2-enoic acid; and
(E)-3-[4-[7-(2,2-difluoropropyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-3-fluoro-5-methoxy-phenyl]prop-2-enoic acid
In further embodiments there is provided a compound of Formula (I), which is obtainable by the methods described in the 'Examples' section hereinafter.
The present specification is intended to include all isotopes of atoms occurring in the present compounds. Isotopes will be understood to include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include tritium and deuterium. Isotopes of carbon include 13C and 14C. In a particular embodiment, R5 is deuterium.
A suitable pharmaceutically acceptable salt of a compound of the Formula (I) is, for example, an acid addition salt or a base addition salt. For example, a base addition salt may be formed using a metal salt, for example an alkali or alkaline earth metal salt such as a sodium, potassium, lithium, calcium or magnesium salt, or an ammonium salt, or a salt with an organic base such as methylamine, dimethylamine, trimethylamine, piperidine, morpholine or tris-(2-hydroxyethyl)amine. Further suitable pharmaceutically acceptable salts of a compound of the Formula (I) may be other metal salts, such as zinc, or other such metal cations known in the art. Further suitable pharmaceutically acceptable salts of a compound of the Formula (I) may be salts formed with a base such as one of the following: calcium acetate, diethylamine, ethanolamine, ethylenediamine, hydroxyethylpyrrolidine, magnesium acetate, meglumine, piperazine, potassium hydroxide, sodium hydroxide, t- butylamine, triethanolamine, tris(hydroxymethyl)-aminomethane (Tris) or N,N- diethylethanolamine .
In one embodiment there is provided a pharmaceutically acceptable salt of a compound of Formula (I) which is a salt comprising a metal cation, an ammonium cation or a salt formed with an organic base.
A further suitable pharmaceutically acceptable salt of a compound of the Formula (I) is, for example, a salt formed within the human or animal body after administration of a compound of the Formula (I) to said human or animal body.
A suitable pharmaceutically acceptable salt of a compound of the Formula (I) may also be, for example, an acid-addition salt of a compound of the Formula (I), for example an acid-addition salt with an inorganic or organic acid such as hydrochloric acid, hydrobromic acid, sulphuric acid or trifluoroacetic acid. Pharmaceutically acceptable salts of a compound of the Formula (I) may also be an acid-addition salt with an acid such as one of the following: acetic acid, adipic acid, benzene sulfonic acid, benzoic acid, cinnamic acid, citric acid, D,L-lactic acid, ethane disulfonic acid, ethane sulfonic acid, fumaric acid, L-tartaric acid, maleic acid, malic acid, malonic acid, methane sulfonic acid, napadisylic acid, phosphoric acid, saccharin, succinic acid, /7-toluenesulfonic acid or toluene sulfonic acid.
The compound of Formula (I) or pharmaceutically acceptable salt thereof may be prepared as a co-crystal solid form. It is to be understood that a pharmaceutically acceptable co-crystal of a compound of the Formula (I) or pharmaceutically acceptable salts thereof, form an aspect of the present specification.
It is to be understood that a suitable pharmaceutically acceptable pro-drug of a compound of the Formula (I) also forms an aspect of the present specification.
Accordingly, the compounds of the specification may be administered in the form of a prodrug, which is a compound that is broken down in the human or animal body to release a compound of the specification. A pro-drug may be used to alter the physical properties and/or the pharmacokinetic properties of a compound of the specification. A pro-drug can be formed when the compound of the specification contains a suitable group or substituent to which a property-modifying group can be attached. Examples of pro-drugs include in- vivo cleavable ester or amide derivatives that may be formed at the carboxy group in a compound of the Formula (I).
Accordingly, one aspect of the present specification includes those compounds of Formula (I) as defined hereinbefore when made available by organic synthesis and when made available within the human or animal body by way of cleavage of a pro-drug thereof. Accordingly, the present specification includes those compounds of the Formula (I) that are produced by organic synthetic means and also such compounds that are produced in the human or animal body by way of metabolism of a precursor compound, that is a compound of the Formula (I) may be a synthetically-produced compound or a metabolically- produced compound.
A suitable pharmaceutically acceptable pro-drug of a compound of the Formula (I) is one that is based on reasonable medical judgement as being suitable for administration to the human or animal body without undesirable pharmacological activities and without undue toxicity.
Various forms of pro-drug have been described, for example in the following documents :- a) Methods in Enzymology, Vol. 42, p. 309-396, edited by K. Widder, et al.
(Academic Press, 1985);
b) Design of Pro-drugs, edited by H. Bundgaard, (Elsevier, 1985);
c) A Textbook of Drug Design and Development, edited by Krogsgaard-Larsen and H. Bundgaard, Chapter 5 "Design and Application of Pro-drugs", by H. Bundgaard p. 113-191 (1991);
d) H. Bundgaard, Advanced Drug Delivery Reviews, 8, 1-38 (1992);
e) H. Bundgaard, et al., Journal of Pharmaceutical Sciences, 77, 285 (1988);
f) N. Kakeya, et al., Chem. Pharm. Bull, 32, 692 (1984);
g) T. Higuchi and V. Stella, "Pro-Drugs as Novel Delivery Systems", A.C.S.
Symposium Series, Volume 14; and
h) E. Roche (editor), "Bioreversible Carriers in Drug Design", Pergamon Press, 1987.
A suitable pharmaceutically acceptable pro-drug of a compound of the Formula (I) that possesses a carboxy group is, for example, an in-vivo cleavable ester thereof. An in- vivo cleavable ester of a compound of the Formula (I) containing a carboxy group is, for example, a pharmaceutically acceptable ester which is cleaved in the human or animal body to produce the parent acid. Suitable pharmaceutically acceptable esters for a carboxy group include (l-6C)alkyl esters such as methyl, ethyl and ie/t-butyl, (l-6C)alkoxymethyl esters such as methoxymethyl esters, (l-6C)alkanoyloxymethyl esters such as
pivaloyloxymethyl esters, 3-phthalidyl esters, (3-8C)cycloalkylcarbonyloxy-(l-6C)alkyl esters such as cyclopentylcarbonyloxymethyl and 1-cyclohexylcarbonyloxyethyl esters, 2- oxo-l,3-dioxolenylmethyl esters such as 5-methyl-2-oxo-l,3-dioxolen-4-ylmethyl esters and (l-6C)alkoxycarbonyloxy-(l-6C)alkyl esters such as methoxycarbonyloxymethyl and 1 -methoxycarbonyloxyethyl esters .
A suitable pharmaceutically acceptable pro-drug of a compound of the Formula (I) which have a carboxy group is for example an in-vivo cleavable amide such as a N-Ci-e alkyl and N,N-di-(Ci-6alkyl)amide such as N-methyl, N-ethyl, N-propyl, N,N-dimethyl, N- ethyl-N-methyl or N,N-diethylamide.
The in-vivo effects of a compound of the Formula (I) may be exerted in part by one or more metabolites that are formed within the human or animal body after administration of a compound of the Formula (I). As stated hereinbefore, the in-vivo effects of a compound of the Formula (I) may also be exerted by way of metabolism of a precursor compound (a pro-drug).
For the avoidance of doubt it is to be understood that where in this specification a group is qualified by 'hereinbefore defined' or 'defined herein' the said group
encompasses the first occurring and broadest definition as well as each and all of the alternative definitions for that group.
Another aspect of the present specification provides a process for preparing a compound of the Formula (I), or a pharmaceutically acceptable salt thereof. Necessary starting materials may be obtained by standard procedures of organic chemistry. The preparation of such starting materials is described in conjunction with the following representative process variants and within the accompanying Examples. Alternatively, necessary starting materials are obtainable by analogous procedures to those illustrated which are within the ordinary skill of an organic chemist.
Compounds of Formula (I) may be prepared by hydrolysis of a compound of Formula (II), wherein R1, R2, R3, R4, R5, R6, R7, R8, R9, R10 and R11 are as defined in the compound of Formula (I) and R12 is (1-6C) alkyl. Hydrolysis is conveniently carried out in the presence of a suitable base (such as aqueous sodium hydroxide) or acid (such as hydrochloric acid) in a suitable solvent (such as THF/methanol or DCM) and at a suitable temperature, for example at room temperature.
Figure imgf000031_0001
Unless otherwise stated, (1-6C) alkyl groups containing the requisite number of carbon atoms can be branched or unbranched. Examples of suitable (1-6C) alkyl groups include methyl, ethyl, n-propyl, i-propyl, n-butyl, sec -butyl, t-butyl, n-pentyl, i-pentyl, neo- pentyl, n-hexyl and i-hexyl, such as methyl and t-butyl.
Accordingly, a compound of Formula (II), or a salt thereof, is a useful intermediate towards the preparation of the compound of Formula (I) or a pharmaceutically acceptable salt thereof.
Therefore, in one aspect there is provided a compound of Formula (II), as shown hereinabove, or a salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, R8, R9, R10 and R11 are as defined in the compound of Formula (I) and R12 is (1-6C) alkyl, provided that the compound of formula (II) is not:
Methyl (E)-3-[3,5-difluoro-4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydro- pyrazolo[4,3-/]isoquinolin-6-yl]phenyl]prop-2-enoate;
Methyl (E)-3-[4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoate;
Methyl (E)-3-[4-[7-(2,2-difluoropropyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoate; Methyl (E)-3-[3-fluoro-4-[7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9- tetrahydropyrazolo[4,3-/]isoquinolin-6-yl]-5-methoxy-phenyl]prop-2-enoate; or
Methyl (E)-3-[4-[7-(2,2-difluoropropyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-3-fluoro-5-methoxy-phenyl]prop-2-enoate;
or enantiomeric or diastereomeric isomers thereof.
Compounds of formula (II) may be made by, for example: a Heck type coupling reaction of a compound of formula (III), where L is for example a halogen (such as Br), a trifluoromethanesulfonyl (triflate) group, a boronic acid or a boronate ester and P is optionally an appropriate protecting group (for example THP or Boc that may be subsequently removed by treatment with acid), with an acrylate ester of formula (IV), where R12 is (1-6C) alkyl, using a suitable metal catalyst (for example Pd- 118) in a suitable solvent (for example 1,4-dioxane) in the presence of a suitable base (for example diisopropylethylamine) and at a suitable temperature (such as 100- 130°C).
Figure imgf000032_0001
(Ill) (IV)
Compounds of formula (III) where R5 is methyl, may be made from, for example, compounds of formula (V) by oxidation with a suitable reagent (for example
bis(trifluoracetoxy)-iodobenzene or eerie ammonium nitrate) and treatment with an organometallic reagent (for example methyl magnesium bromide) in a suitable solvent (for example THF) at a low temperature (typically -80 to -60 °C)
Figure imgf000033_0001
(V)
Compounds of formula (V) may be prepared by, for example, reaction of an aniline of Formula (VI) with suitable reagents to effect the construction of an indazole such as inorganic nitrite (such as sodium nitrite) in organic acid (such as propionic acid) optionally in the presence of water, at low temperature (typically -20 to 0 °C); or alternatively an acid anhydride (such as acetic anhydride) in the presence of a suitable base (such as potassium acetate) together with organic nitrite (such as isopentyl nitrite) optionally in the presence of a crown ether (such as 18-crown-6) in a suitable solvent (such as chloroform) at a suitable temperature (such as 70 °C). In such a process L is for example a halogen (such as Br) or a trifluoromethanesulfonyl (triflate) group or a boronic acid or a boronate ester or alternatively an acrylate ester.
Figure imgf000033_0002
(VI)
Compounds of formula (VI) may be made by reaction of a compound of formula (VII) with a compound of formula (VIII) under conditions known in the art as suitable for Pictet-Spengler reactions, such as in the presence of acid (such as acetic acid) and in a suitable solvent (for example toluene or water) and a suitable temperature (such as 60- 100°C). In such a process L is for example a halogen (such as Br) or a trifluoromethanesulfonyl (triflate) group or a boronic acid or a boronate ester or alternatively an acrylate ester.
Figure imgf000034_0001
(VII) (VIII)
Compounds of formula (VII) may be prepared by functional group interconversions known to those skilled in the art of organic synthesis, for example by the amination of aryl halides, such as the aryl bromide of formula (IX), using a protected amine (such as diphenylmethanimine) in the presence of a suitable catalyst and ligand (such as bis(dibenzylideneacetone)palladium(0) and rac-2,2'-bis(diphenylphosphino)- 1,1'- binaphthyl), in the presence of a suitable base (such as sodium iert-butoxide), in a suitable solvent (such as toluene) and at a suitable temperature (such as 80-100°C); followed by deprotection of the protected amine using, for example, acid (such as hydrochloric acid) in a suitable solvent (such as DCM) at a suitable temperature (such as ambient temperature).
Figure imgf000034_0002
Compounds of formula (IX) may be prepared by
a) reaction of a compound of formula (X) with an aldehyde of formula (XI), in a suitable solvent (for example THF) in the presence of a suitable reducing agent (such as sodium triacetoxyborohydride) and at a suitable temperature (such as 20-30°C);
b) (i) reaction of a compound of formula (X) with an acid of formula (XII) under standard amide bond forming conditions (for example in the presence of an amide coupling reagent (such as HATU) and a suitable base (such as triethylamine) in a suitable solvent (such as DMF)), followed by (ii) reduction of the resultant amide bond using a suitable reducing agent (such as borane) in a suitable solvent (such as THF) at a suitable temperature (such as 60-70 °C);
reaction of a compound of formula (X) with a compound of formula (XIII), wherein LG is a suitable leaving group (for example a halogen atom (such as bromo or chloro) or trifluoromethanesulfone), in the presence of a suitable base (such as
diisopropylethylamine) in a suitable solvent (for example DCM or dioxane) and at a suitable temperature (such as 20-85 °C).
Figure imgf000035_0001
(X) (XI) (XII) (XIII)
Compounds of formula (X) may be prepared by a number of methods known to the art the synthesis of chiral amines notably;
a) Metal-halogen exchange on an aryl bromide using, for example, n-butyllithium in THF at -78°C, followed by ring-opening of a sulfamidate of formula (XIV) according to the scheme shown below:
Figure imgf000035_0002
(XIV)
b) Phase transfer alkylation in the presence of a chiral catalyst, for example using (15,25,45,5R)-2-((R)-(allyloxy)(quinolin-4-yl)methyl)- l-(anthracen-9-ylmethyl)-5- vinylquinuclidin-l-ium bromide in toluene/KOH at 0°C, followed by functional group manipulation:
Figure imgf000035_0003
Alternatively, compounds of formula (V) may be prepared by alkylation of compounds of formula (XV) with a compound of formula (XVI), wherein LG is a suitable leaving group (for example a halogen atom (such as bromo or chloro) or
trifluoromethanesulfone), in the presence of a suitable base (such as
diisopropylethylamine) in a suitable solvent (for example DCM or 1,4-dioxane) and at a suitable temperature (such as 20-85 °C).
Figure imgf000036_0001
(XV) (XVI)
Compounds of formula (XV) may be made by reaction of a compound of formula (XVII) with a compound of formula (VIII) under conditions known in the art as suitable for Pictet-Spengler reactions, such as in the presence of acid (such as acetic acid or trifluoroacetic acid) at a suitable temperature (such as 60-150°C) optionally with microwave irradiation. In such a process L is for example a halogen (such as Br) or a trifluoromethanesulfonyl (triflate) group or a boronic acid or a boronate ester or alternatively an acrylate ester.
Figure imgf000036_0002
(XVII) (VIII)
It is to be understood that other permutations of the process steps in the process variants described above are also possible.
When a pharmaceutically acceptable salt of a compound of the Formula (I) is required it may be obtained by, for example, reaction of said compound with a suitable acid or suitable base. When a pharmaceutically acceptable pro-drug of a compound of the Formula (I) is required, it may be obtained using a conventional procedure. For example, an in-vivo cleavable ester of compound of the Formula (I) may be obtained by, for example, reaction of a compound of the Formula (I) containing a carboxy group with a pharmaceutically acceptable alcohol. Further information on pro-drugs has been provided hereinbefore.
It will also be appreciated that, in some of the reactions mentioned hereinbefore, it may be necessary or desirable to protect any sensitive groups in the compounds. The instances where protection is necessary or desirable, and suitable methods for protection, are known to those skilled in the art. Conventional protecting groups may be used in accordance with standard practice (for illustration see T.W. Green, Protective Groups in Organic Synthesis, John Wiley and Sons, 1991). Thus, if reactants include groups such as amino, carboxy or hydroxy, it may be desirable to protect the group in some of the reactions mentioned herein.
A suitable protecting group for an amino or alkylamino group is, for example, an acyl group, for example an alkanoyl group such as acetyl, an alkoxycarbonyl group, for example a methoxycarbonyl, ethoxycarbonyl or i-butoxycarbonyl group, an
arylmethoxycarbonyl group, for example benzyloxycarbonyl, or an aroyl group, for example benzoyl. The deprotection conditions for the above protecting groups necessarily vary with the choice of protecting group. Thus, for example, an acyl group such as an alkanoyl or alkoxycarbonyl group or an aroyl group may be removed for example, by hydrolysis with a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide. Alternatively an acyl group such as a i-butoxycarbonyl group may be removed, for example, by treatment with a suitable acid as hydrochloric, sulphuric or phosphoric acid or trifluoroacetic acid and an arylmethoxycarbonyl group such as a benzyloxycarbonyl group may be removed, for example, by hydrogenation over a catalyst such as palladium-on-carbon, or by treatment with a Lewis acid for example boron tris(trifluoroacetate). A suitable alternative protecting group for a primary amino group is, for example, a phthaloyl group which may be removed by treatment with an alkylamine, for example dimethylaminopropylamine, or with hydrazine.
A suitable protecting group for a hydroxy group is, for example, an acyl group, for example an alkanoyl group such as acetyl, an aroyl group, for example benzoyl, or an arylmethyl group, for example benzyl. The deprotection conditions for the above protecting groups will necessarily vary with the choice of protecting group. Thus, for example, an acyl group such as an alkanoyl or an aroyl group may be removed, for example, by hydrolysis with a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide. Alternatively an arylmethyl group such as a benzyl group may be removed, for example, by hydrogenation over a catalyst such as palladium-on- carbon.
A suitable protecting group for a carboxy group is, for example, an esterifying group, for example a methyl or an ethyl group which may be removed, for example, by hydrolysis with a base such as sodium hydroxide, or for example a i-butyl group which may be removed, for example, by treatment with an acid, for example an organic acid such as trifluoroacetic acid, or for example a benzyl group which may be removed, for example, by hydrogenation over a catalyst such as palladium-on-carbon.
The protecting groups may be removed at any convenient stage in the synthesis using conventional techniques well known in the chemical art.
Certain of the intermediates defined herein are novel and these are provided as further features of the specification.
Biological Assays-
The following assays were used to measure the effects of the compounds of the present specification.
ERq binding assay
The ability of compounds to bind to isolated Estrogen Receptor Alpha Ligand binding domain (ER alpha - LBD (GST)) was assessed in competition assays using a LanthaScreen™ Time-Resolved Fluorescence Resonance Energy Transfer (TR-FRET) detection end-point. For the LanthaScreen TR-FRET endpoint, a suitable fluorophore (Fluormone ES2, ThermoFisher, Product code P2645) and recombinant human Estrogen Receptor alpha ligand binding domain, residues 307-554 (expressed and purified in-house) were used to measure compound binding. The assay principle is that ER alpha -LBD (GST) is added to a fluorescent ligand to form a receptor/fluorophore complex. A terbium- labelled anti-GST antibody (Product code PV3551) is used to indirectly label the receptor by binding to its GST tag, and competitive binding is detected by a test compounds' ability to displace the fluorescent ligand resulting in a loss of TR-FRET signal between the Tb- anti-GST antibody and the tracer. The assay was performed as follows with all reagent additions carried out using the Beckman Coulter BioRAPTR FRD microfluidic
workstation :-
1. Acoustic dispense 120nL of the test compound into a black low volume 384 well assay plates.
2. Prepare lx ER alpha -LBD/Tb-antiGST Ab in ES2 screening buffer and
incubate for 15 minutes.
3. Dispense
Figure imgf000039_0001
of the lx AR-LBD/Tb-anti-GST Ab reagent into each well of the assay plate followed by 6 μΐ Fluorophore reagent into each well of the assay plate
4. Cover the assay plate to protect the reagents from light and evaporation, and incubate at room temperature for 4 hours.
5. Excite at 337nm and measure the fluorescent emission signal of each well at 490nm and 520nm using the BMG PheraSTAR.
Compounds were dosed directly from a compound source microplate containing serially diluted compound (4 wells containing lOmM, O. lmM, ImM and lOnM final compound respectively) to an assay microplate using the Labcyte Echo 550. The Echo 550 is a liquid handler that uses acoustic technology to perform direct microplate-to-microplate transfers of DMSO compound solutions and the system can be programmed to transfer multiple small nL volumes of compound from the different source plate wells to give the desired serial dilution of compound in the assay which is then back-filled to normalise the DMSO concentration across the dilution range. In total 120nL of compound plus DMSO is added to each well and compounds were tested in a 12-point concentration response format over a final compound concentration range of 10, 2.917, 1.042, 0.2083, 0.1, 0.0292, 0.0104, 0.002083, 0.001, 0.0002917, 0.0001042, Ο.ΟΟΟΟΙμΜ respectively. TR-FRET dose response data obtained with each compound was exported into a suitable software package (such as Origin or Genedata) to perform curve fitting analysis. Competitive ER alpha binding was expressed as an IC50 value. This was determined by calculation of the concentration of compound that was required to give a 50% reduction in tracer compound binding to ER alpha-LBD. MCF-7 ER down regulation assay
The ability of compounds to down-regulate Estrogen Receptor (ER) numbers was assessed in a cell based immuno-fluorescence assay using the MCF-7 human ductal carcinoma breast cell line. MCF-7 cells were revived directly from a cryovial (approx 5 x 106 cells) in Assay Medium (phenol red free Dulbecco's Modified Eagle's medium
(DMEM) (Sigma D5921) containing 2mM L-Glutamine and 5% (v/v) Charcoal/Dextran treated foetal calf serum. Cells were syringed once using a sterile 18G x 1.5inch (1.2 x 40mm) broad gauge needle and cell density was measured using a Coulter Counter (Beckman). Cells were further diluted in Assay Medium to a density of 3.75 x 104 cells per mL and 40μϊ^ per well added to transparent bottomed, black, tissue culture treated 384 well plates (Costar, No. 3712) using a Thermo Scientific Matrix WellMate or Thermo
Multidrop. Following cell seeding, plates were incubated overnight at 37°C, 5% C02 (Liconic carousel incubator). Test data was generated using the LabCyte Echo™ model 555 compound reformatter which is part of an automated workcell (Integrated Echo 2 workcell). lOmM compound stock solutions of the test compounds were used to generate a 384 well compound dosing plate (Labcyte P-05525-CV1). 40μΛ of each of the lOmM compound stock solutions was dispensed into the first quadrant well and then 1: 100 stepwise serial dilutions in DMSO were performed using a Hydra II (MATRIX UK) liquid handling unit to give 40μL· of diluted compound into quadrant wells 2 (O. lmM), 3 (ΙμΜ) and 4 (0.0 ΙμΜ), respectively. 40μΛ of DMSO added to wells in row P on the source plate allow for DMSO normalisation across the dose range. To dose the control wells 40μL· of DMSO was added to row 01 and 40μΙ, of ΙΟΟμΜ fulvestrant in DMSO was added to row 03 on the compound source plate. The Echo uses acoustic technology to perform direct microplate-to-microplate transfers of DMSO compound solutions to assay plates. The system can be programmed to transfer volumes as low as 2.5 nL in multiple increments between microplates and in so doing generates a serial dilution of compound in the assay plate which is then back-filled to normalise the DMSO concentration across the dilution range. Compounds were dispensed onto the cell plates with a compound source plate prepared as above producing a 12pt duplicate 3μΜ to 3pM dose range with 3 fold dilutions and one final 10 fold dilution using the Integrated Echo 2 workcell. The maximum signal control wells were dosed with DMSO to give a final concentration of 0.3% and the minimum signal control wells were dosed with fulvestrant to give a final concentration of ΙΟΟηΜ accordingly. Plates were further incubated for 18-22 hours at 37°C, 5% C02 and then fixed by the addition of 20μί of 11.1 % (v/v) formaldehyde solution (in phosphate buffered saline (PBS)) giving a final formaldehyde concentration of 3.7% (v/v). Cells were fixed at room temperature for 20 mins before being washed two times with 250μί
PBS/Proclin (PBS with a Biocide preservative) using a BioTek platewasher, 40μL· of PBS/Proclin was then added to all wells and the plates stored at 4°C. The fixing method described above was carried out on the Integrated Echo 2 workcell. Immunostaining was performed using an automated AutoElisa workcell. The PBS/Proclin was aspirated from all wells and the cells permeabilised with 40μϊ^ PBS containing 0.5% Tween™ 20 (v/v) for 1 hour at room temperature. The plates were washed three times in 250μί of PBS/0.05% (v/v) Tween 20 with Proclin (PBST with a Biocide preservative) and then 20μL· of ERa (SP1) Rabbit monoclonal antibody (Thermofisher) 1: 1000 in PBS/Tween™/3% (w/v) Bovine Serum Albumin was added. The plates were incubated overnight at 4°C (Liconic carousel incubator) and then washed three times in 250μί of PBS/0.05% (v/v) Tween™ 20 with Proclin (PBST). The plates were then incubated with 20μΕΛνε11 of a goat anti-rabbit IgG AlexaFluor 594 or goat anti-rabbit AlexaFluor 488 antibody (Molecular Probes) with Hoechst at 1:5000 in PBS/Tween™/3% (w/v) Bovine Serum Albumin for lhour at room temperature. The plates were then washed three times in 250μί of PBS/0.05% (v/v) Tween™ 20 with Proclin (PBST with a Biocide preservative). 20μϊ^ of PBS was added to each well and the plates covered with a black plate seal and stored at 4°C before being read. Plates were read using a Cellomics Arrayscan reading the 594nm (24hr time point) or 488nm (5hr timepoint) fluorescence to measure the ERa receptor level in each well. The mean total intensity was normalized for cell number giving the total intensity per cell. The data was exported into a suitable software package (such as Origin) to perform curve fitting analysis. Down-regulation of the ERa receptor was expressed as an IC50 value and was determined by calculation of the concentration of compound that was required to give a 50% reduction of the average maximum Total Intensity signal.
The following data were generated for the Examples (the data below may be a result from a single experiment or an average of two or more experiments): Table A
Figure imgf000042_0001
1 Compounds tested in the ER down regulation assay show downregulation values (>90 ) in the assay unless otherwise stated, in which case the % downregulation is shown in brackets.
Human Hepatocvte assay
The metabolic stability of compounds in human hepatocytes was assed using the following protocol:
1. Prepare 10 mM stock solutions of compound and control compounds in
appropriate solvent (DMSO). Place incubation medium (L-15Medium) in a 37 °C water bath, and allow warming for at least 15 minutes prior to use.
2. Add 80 of acetonitrile to each well of the 96-well deep well plate (quenching plate).
3. In a new 96-well plate, dilute the 10 mM test compounds and the control compounds to 100 μΜ by combining 198 of acetonitrile and 2 of 10 mM stock.
4. Remove a vial of cryopreserved (less than -150 °C) human hepatocytes
(LiverPool™ 10 Donor Human hepatocytes obtained from Celsis IVT. Chicago, IL (Product No. SO 1205)) from storage, ensuring that vials remain at cryogenic temperatures until thawing process ensues. As quickly as possible, thaw the cells by placing the vial in a 37 °C water bath and gently shaking the vials. Vials should remain in water bath until all ice crystals have dissolved and are no longer visible. After thawing is completed, spray vial with 70% ethanol, transfer the vial to a bio- safety cabinet.
5. Open the vial and pour the contents into the 50 mL conical tube containing thawing medium. Place the 50 mL conical tube into a centrifuge and spin at 100 g for 10 minutes. Upon completion of spin, aspirate thawing medium and resuspend hepatocytes in enough incubation medium to yield ~1.5xl06 cells/mL.
6. Using Cellometer® Vision, count cells and determine the viable cell density. Cells with poor viability (<80% viability) are not acceptable for use. Dilute cells with incubation medium to a working cell density of l.OxlO6 viable cells/mL.
7. Transfer 247.5 of hepatocytes into each well of a 96-well cell culture plate. Place the plate on Eppendorf Thermomixer Comfort plate shaker to allow the hepatocytes to warm for 10 minutes.
8. Add 2.5μΕ of 100 μΜ test compound or control compounds into an incubation well containing cells, mix to achieve a homogenous suspension at 0.5 min, which when achieved, will define the 0.5 min time point. At the 0.5 min time, transfer 20 μΐ^ incubated mixture to wells in a "Quenching plate" followed by vortexing.
9. Incubate the plate at 37 °C at 900 rpm on an Eppendorf Thermomixer Comfort plate shaker. At 5, 15, 30, 45, 60, 80, 100 and 120 min, mix the incubation system and transfer samples of 20 μΐ^ incubated mixture at each time point to wells in a separate "Quenching plate" followed by vortexing.
10. Centrifuge the quenching plates for 20 minutes at 4,000 rpm. 4 different
compounds are pooled into one cassette and used for LC/MS/MS analysis.
All calculations are carried out using Microsoft Excel. Peak areas are determined from extracted ion chromato grams. In vitro intrinsic clearance (in vitro Clint, in L/min/106 cells) of parent compound is determined by regression analysis of the Ln percent parent disappearance vs. time curve. The in vitro intrinsic clearance (in vitro Clint, in L/min/106 cells) is determined from the slope value using the following equation: in vitro Clint =kV/N
V = incubation volume (0.25 mL); number of hepatocytes per well (0.25x10 6 cells).
Figure imgf000044_0001
According to a further aspect of the specification there is provided a pharmaceutical composition, which comprises a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore in association with a pharmaceutically acceptable diluent or carrier.
Suitable pharmaceutically acceptable excipients for a tablet formulation include, for example, inert diluents, granulating and disintegrating agents, binding agents, lubricating agents, preservative agents and antioxidants. A further suitable pharmaceutically acceptable excipient may be a chelating agent. Tablet formulations may be uncoated or coated either to modify their disintegration and the subsequent absorption of the active ingredient within the gastrointestinal tract, or to improve their stability and/or appearance, in either case, using conventional coating agents and procedures well known in the art.
Compositions for oral use may alternatively be in the form of hard gelatin capsules in which the active ingredient is mixed with an inert solid diluent, or as soft gelatin capsules in which the active ingredient is mixed with water or an oil.
Aqueous suspensions generally contain the active ingredient in finely powdered form together with one or more suspending agents, dispersing or wetting agents. The aqueous suspensions may also contain one or more preservatives, anti-oxidants, colouring agents, flavouring agents, and/or sweetening agents. Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil or in a mineral oil. The oily suspensions may also contain a thickening agent. Sweetening agents such as those set out above, and flavouring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant.
Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water generally contain the active ingredient together with a dispersing or wetting agent, suspending agent and one or more preservatives. Additional excipients such as sweetening, flavouring and colouring agents, may also be present.
The pharmaceutical compositions of the specification may also be in the form of oil-in- water emulsions. The oily phase may be a vegetable oil or a mineral oil or a mixture of any of these. The emulsions may also contain sweetening, flavouring and preservative agents.
Syrups and elixirs may be formulated with sweetening agents, and may also contain a demulcent, preservative, flavouring and/or colouring agent.
The pharmaceutical compositions may also be in the form of a sterile injectable aqueous or oily suspension, which may be formulated according to known procedures using one or more of the appropriate dispersing or wetting agents and suspending agents, which have been mentioned above. A sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent system.
Compositions for administration by inhalation may be in the form of a conventional pressurised aerosol arranged to dispense the active ingredient either as an aerosol containing finely divided solid or liquid droplets. Conventional aerosol propellants such as volatile fluorinated hydrocarbons or hydrocarbons may be used and the aerosol device is conveniently arranged to dispense a metered quantity of active ingredient. Dry powder inhalers may also be suitable.
For further information on formulation the reader is referred to Chapter 25.2 in Volume 5 of Comprehensive Medicinal Chemistry (Corwin Hansch; Chairman of Editorial Board), Pergamon Press 1990.
The amount of active ingredient that is combined with one or more excipients to produce a single dosage form will necessarily vary depending upon the host treated and the particular route of administration. For example, oral administration to humans will generally require, for example, from 1 mg to 2 g of active agent (more suitably from lOOmg to 2g, for example from 250 mg to 1.8g, such as from 500mg to 1.8g, particularly from 500mg to 1.5g, conveniently from 500mg to lg) to be administered compounded with an appropriate and convenient amount of excipients which may vary from about 3 to about 98 percent by weight of the total composition. It will be understood that, if a large dosage is required, multiple dosage forms may be required, for example two or more tablets or capsules, with the dose of active ingredient divided conveniently between them. Typically, unit dosage forms will contain about 10 mg to 0.5 g of a compound of this specification, although a unit dosage form may contain up to lg. Conveniently, a single solid dosage form may contain between 1 and 300mg of active ingredient.
The size of the dose for therapeutic or prophylactic purposes of compounds of the present specification will naturally vary according to the nature and severity of the disease state, the age and sex of the animal or patient and the route of administration, according to well known principles of medicine.
In using compounds of the present specification for therapeutic or prophylactic purposes it will generally be administered so that a daily dose in the range, for example, 1 mg/kg to 100 mg/kg body weight is received, given if required in divided doses. In general, lower doses will be administered when a parenteral route is employed. Thus, for example, for intravenous administration, a dose in the range, for example, 1 mg/kg to 25 mg/kg body weight will generally be used. Similarly, for administration by inhalation, a dose in the range, for example, 1 mg/kg to 25 mg/kg body weight will be used. Oral administration is however preferred, particularly in tablet form.
In one aspect of the specification, compounds of the present specification or pharmaceutically acceptable salts thereof, are administered as tablets comprising lOmg to 300mg of the compound of Formula (I) (or a pharmaceutically acceptable salt thereof), wherein one or more tablets are administered as required to achieve the desired dose.
As stated above, it is known that signalling through ERa causes tumourigenesis by one or more of the effects of mediating proliferation of cancer and other cells, mediating angiogenic events and mediating the motility, migration and invasiveness of cancer cells. We have found that the compounds of the present specification possess potent anti-tumour activity which it is believed is obtained by way of antagonism and down-regulation of ERa that is involved in the signal transduction steps which lead to the proliferation and survival of tumour cells and the invasiveness and migratory ability of metastasising tumour cells.
Accordingly, the compounds of the present specification may be of value as anti- tumour agents, in particular as selective inhibitors of the proliferation, survival, motility, dissemination and invasiveness of mammalian cancer cells leading to inhibition of tumour growth and survival and to inhibition of metastatic tumour growth. Particularly, the compounds of the present specification may be of value as anti-proliferative and anti- invasive agents in the containment and/or treatment of solid tumour disease. Particularly, the compounds of the present specification may be useful in the prevention or treatment of those tumours which are sensitive to inhibition of ERa and that are involved in the signal transduction steps which lead to the proliferation and survival of tumour cells and the migratory ability and invasiveness of metastasising tumour cells. Further, the compounds of the present specification may be useful in the prevention or treatment of those tumours which are mediated alone or in part by antagonism and down-regulation of ERa, i.e. the compounds may be used to produce an ERa inhibitory effect in a warm-blooded animal in need of such treatment.
According to a further aspect of the specification there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore for use as a medicament in a warm-blooded animal such as man.
According to a further aspect of the specification, there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore for use in the production of an anti-proliferative effect in a warm-blooded animal such as man.
According to a further aspect of the specification there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore for use in a warm-blooded animal such as man as an anti-invasive agent in the containment and/or treatment of solid tumour disease.
According to a further aspect of the specification, there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, for the production of an anti-proliferative effect in a warm-blooded animal such as man.
According to a further aspect of the specification there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, in the manufacture of a medicament for use in the production of an antiproliferative effect in a warm-blooded animal such as man.
According to a further aspect of the specification there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, in the manufacture of a medicament for use in a warm-blooded animal such as man as an anti-invasive agent in the containment and/or treatment of solid tumour disease.
According to a further aspect of the specification there is provided a method for producing an anti-proliferative effect in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
According to a further aspect of the specification there is provided a method for producing an anti-invasive effect by the containment and/or treatment of solid tumour disease in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
According to a further aspect of the specification, there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, for use in the prevention or treatment of cancer in a warm-blooded animal such as man.
According to a further aspect of the specification there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore in the manufacture of a medicament for use in the prevention or treatment of cancer in a warm-blooded animal such as man.
According to a further aspect of the specification there is provided a method for the prevention or treatment of cancer in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
According to a further aspect of the specification, there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore for use in the prevention or treatment of solid tumour disease in a warm-blooded animal such as man.
According to a further aspect of the specification there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, in the manufacture of a medicament for use in the prevention or treatment of solid tumour disease in a warm-blooded animal such as man.
According to a further aspect of the specification there is provided a method for the prevention or treatment of solid tumour disease in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
According to a further aspect of the specification there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, for use in the prevention or treatment of those tumours which are sensitive to inhibition of ERa that are involved in the signal transduction steps which lead to the proliferation, survival, invasiveness and migratory ability of tumour cells.
According to a further aspect of the specification there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, in the manufacture of a medicament for use in the prevention or treatment of those tumours which are sensitive to inhibition of ERa that are involved in the signal transduction steps which lead to the proliferation, survival, invasiveness and migratory ability of tumour cells.
According to a further aspect of the specification there is provided a method for the prevention or treatment of those tumours which are sensitive to inhibition of ERa that are involved in the signal transduction steps which lead to the proliferation, survival, invasiveness and migratory ability of tumour cells which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
According to a further aspect of the specification there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore for use in providing an inhibitory effect on ERa. According to a further aspect of the specification there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore in the manufacture of a medicament for use in providing an inhibitory effect on ERa.
According to a further aspect of the specification there is also provided a method for providing an inhibitory effect on ERa which comprises administering an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
According to a further aspect of the specification there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, for use in providing a selective inhibitory effect on ERa.
According to a further aspect of the specification there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, in the manufacture of a medicament for use in providing a selective inhibitory effect on ERa.
According to a further aspect of the specification there is also provided a method for providing a selective inhibitory effect on ERa which comprises administering an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
Described herein are compounds that can bind to ERa ligand binding domain and are selective estrogen receptor degraders. In biochemical and cell based assays the compounds of the present specification are shown to be potent estrogen receptor binders and reduce cellular levels of ERa and may therefore be useful in the treatment of estrogen sensitive diseases or conditions (including diseases that have developed resistance to endocrine therapies), i.e. for use in the treatment of cancer of the breast and gynaecological cancers (including endometrial, ovarian and cervical) and cancers expressing ERa mutated proteins which may be de novo mutations or have arisen as a result of treatment with a prior endocrine therapy such as an aromatase inhibitor.
According to a further aspect of the specification there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, for use in the treatment of breast or gynaecological cancers. According to a further aspect of the specification there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, for use in the treatment of cancer of the breast, endometrium, ovary or cervix.
According to a further aspect of the specification there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, for use in the treatment of cancer of the breast.
According to a further aspect of the specification there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, for use in the treatment of cancer of the breast, wherein the cancer has developed resistance to one or more other endocrine therapies.
According to a further aspect of the specification there is provided a method for treating breast or gynaecological cancers, which comprises administering an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
According to a further aspect of the specification there is provided a method for treating cancer of the breast, endometrium, ovary or cervix, which comprises administering an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
According to a further aspect of the specification there is provided a method for treating breast cancer, which comprises administering an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
According to a further aspect of the specification there is provided a method for treating breast cancer, wherein the cancer has developed resistance to one or more other endocrine therapies, which comprises administering an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
According to a further aspect of the specification there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, in the manufacture of a medicament for use in the treatment of breast or gynaecological cancers.
According to a further aspect of the specification there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, in the manufacture of a medicament for use in the treatment of cancer of the breast, endometrium, ovary or cervix.
According to a further aspect of the specification there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, in the manufacture of a medicament for use in the treatment of breast cancer.
According to a further aspect of the specification there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, in the manufacture of a medicament for use in the treatment of breast cancer, wherein the cancer has developed resistance to one or more other endocrine therapies.
In one feature of the specification, the cancer to be treated is breast cancer. In a further aspect of this feature, the breast cancer is Estrogen Receptor +ve (ER+ve). In one embodiment of this aspect, the compound of Formula (I) is dosed in combination with another anticancer agent, such as an anti-hormonal agent as defined herein.
According to a further aspect of the specification there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, for use in the treatment of ER+ve breast cancer.
According to a further aspect of the specification there is provided a method for treating ER+ve breast cancer, which comprises administering an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore.
According to a further aspect of the specification there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, as defined herein before in the manufacture of a medicament for use in the treatment of ER+ve breast cancer.
As stated hereinbefore, the in-vivo effects of a compound of the Formula (I) may be exerted in part by one or more metabolites that are formed within the human or animal body after administration of a compound of the Formula (I).
The present specification therefore also contemplates a method for inhibiting ER-cc in a patient, comprising administering to a patient an amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, effective in inhibiting ER-cc in the patient. The present specification therefore also contemplates a method for inhibiting ER-cc in a patient, comprising administering to a patient an amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, effective in inhibiting ER-cc in the patient.
The anti-cancer treatment defined herein may be applied as a sole therapy or may involve, in addition to the compounds of the specification, conventional surgery or radiotherapy or chemotherapy. Such chemotherapy may include one or more of the following categories of anti-tumour agents :-
(i) other antiproliferative/antineoplastic drugs and combinations thereof, as used in medical oncology, such as alkylating agents (for example cis-platin, oxaliplatin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan, temozolamide and nitrosoureas); antimetabolites (for example gemcitabine and antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, and hydroxyurea); antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin- C, dactinomycin and mithramycin); antimitotic agents (for example vinca alkaloids like vincristine, vinblastine, vindesine and vinorelbine and taxoids like taxol and taxotere and polokinase inhibitors); and topoisomerase inhibitors (for example epipodophyllotoxins like etoposide and teniposide, amsacrine, topotecan and camptothecin);
(ii) antihormonal agents such as antioestrogens (for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene), progestogens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrozole, vorazole and exemestane);
(iii) inhibitors of growth factor function and their downstream signalling pathways:
included are Ab modulators of any growth factor or growth factor receptor targets, reviewed by Stern et al. Critical Reviews in Oncologv/Haematology, 2005, 54, ppl 1-29); also included are small molecule inhibitors of such targets, for example kinase inhibitors - examples include the anti-erbB2 antibody trastuzumab [Herceptin™], the anti-EGFR antibody panitumumab, the anti-EGFR antibody cetuximab [Erbitux, C225] and tyrosine kinase inhibitors including inhibitors of the erbB receptor family, such as epidermal growth factor family receptor (EGFR/erbBl) tyrosine kinase inhibitors such as gefitinib or erlotinib, erbB2 tyrosine kinase inhibitors such as lapatinib, and mixed erbl/2 inhibitors such as afatanib; similar strategies are available for other classes of growth factors and their receptors, for example inhibitors of the hepatocyte growth factor family or their receptors including c-met and ron; inhibitors of the insulin and insulin growth factor family or their receptors (IGFR, IR) inhibitors of the platelet-derived growth factor family or their receptors (PDGFR), and inhibitors of signalling mediated by other receptor tyrosine kinases such as c-kit, AnLK, and CSF-1R; also included are modulators which target signalling proteins in the PI3-kinase signalling pathway, for example, inhibitors of PI3-kinase isoforms such as PDK-oc β/γ and ser / thr kinases such as AKT, mTOR (such as AZD2014), PDK, SGK, PI4K or PIP5K; also included are inhibitors of serine/threonine kinases not listed above, for example raf inhibitors such as vemurafenib, MEK inhibitors such as selumetinib (AZD6244), Abl inhibitors such as imatinib or nilotinib, Btk inhibitors such as ibrutinib, Syk inhibitors such as fostamatinib, aurora kinase inhibitors (for example AZD1152), inhibitors of other ser/thr kinases such as JAKs, STATs and IRAK4, and cyclin dependent kinase inhibitors for example inhibitors of CDK1, CDK7, CDK9 and CDK4/6 such as palbociclib;
iv) modulators of DNA damage signalling pathways, for example PARP inhibitors (e.g. Olaparib), ATR inhibitors or ATM inhibitors;
v) modulators of apoptotic and cell death pathways such as Bel family modulators (e.g. ABT-263 / Navitoclax, ABT-199);
(vi) antiangiogenic agents such as those which inhibit the effects of vascular endothelial growth factor, [for example the anti-vascular endothelial cell growth factor antibody bevacizumab (Avastin™) and for example, a VEGF receptor tyrosine kinase inhibitor such as sorafenib, axitinib, pazopanib, sunitinib and vandetanib (and compounds that work by other mechanisms (for example linomide, inhibitors of integrin νβ3 function and angio statin)];
(vii) vascular damaging agents, such as Combretastatin A4;
(viii) anti-invasion agents, for example c-Src kinase family inhibitors like (dasatinib, L Med. Chem., 2004, 47, 6658-6661) and bosutinib (SKI-606), and metalloproteinase inhibitors like marimastat, inhibitors of urokinase plasminogen activator receptor function or antibodies to Heparanase] ;
(ix) immunotherapy approaches, including for example ex-vivo and in- vivo approaches to increase the immunogenicity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor, approaches to decrease T-cell anergy, approaches using transfected immune cells such as cytokine-transfected dendritic cells, approaches using cytokine-transfected tumour cell lines and approaches using anti-idiotypic antibodies. Specific examples include
monoclonal antibodies targeting PD-1 (e.g. BMS-936558) or CTLA4 (e.g. ipilimumab and tremelimumab);
(x) Antisense or RNAi based therapies, for example those which are directed to the targets listed.
(xi) gene therapy approaches, including for example approaches to replace aberrant genes such as aberrant p53 or aberrant BRCA1 or BRCA2, GDEPT (gene-directed enzyme pro-drug therapy) approaches such as those using cytosine deaminase, thymidine kinase or a bacterial nitroreductase enzyme and approaches to increase patient tolerance to chemotherapy or radiotherapy such as multi-drug resistance gene therapy.
Accordingly, in one embodiment there is provided a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and an additional anti-tumour substance for the conjoint treatment of cancer.
According to this embodiment there is provided a combination suitable for use in the treatment of cancer comprising a compound of the Formula (I) or a pharmaceutically acceptable salt thereof and another anti-tumour agent, in particular any one of the anti tumour agents listed under (i) - (xi) above. In particular, the anti-tumour agent listed under (i)-(xi) above is the standard of care for the specific cancer to be treated; the person skilled in the art will understand the meaning of "standard of care".
Therefore in a further embodiment there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with another anti-tumour agent, in particular an anti-tumour agent selected from one listed under (i) - (xi) herein above.
In a further embodiment there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with another anti-tumour agent, in particular an anti-tumour agent selected from one listed under (i) above.
In a further embodiment there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and any one of the anti-tumour agents listed under (i) above. In a further embodiment there is provided a combination suitable for use in the treatment of cancer comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and a taxoid, such as for example taxol or taxotere, conveniently taxotere.
In a further embodiment there is provided a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with another anti-tumour agent, in particular an anti-tumour agent selected from one listed under (ii) herein above.
In a further embodiment there is provided a combination suitable for use in the treatment of cancer comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and any one of the antihormonal agents listed under (ii) above, for example any one of the anti-oestrogens listed in (ii) above, or for example an aromatase inhibitor listed in (ii) above.
In a further embodiment there is provided a combination suitable for use in the treatment of cancer comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and an mTOR inhibitor, such as AZD2014 (see for example WO2008/023161).
Figure imgf000056_0001
AZD2014
In a further embodiment there is provided a combination suitable for use in the treatment of cancer comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and a PDKa-inhibitor, such as the compound l-(4-(5-(5-amino-6- (5-tert-butyl- 1 ,3,4-oxadiazol-2-yl)pyrazin-2-yl)- 1 -ethyl- 1H- 1 ,2,4-triazol-3-yl)piperidin- 1 - yl)-3-hydroxypropan-l-one, or a pharmaceutically- acceptable salt thereof.
In a further embodiment there is provided a combination suitable for use in the treatment of cancer comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and palbociclib.
In one aspect the above combination of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, with an anti-tumour agent listed in (ii) above, or an mTOR inhibitor (such as AZD2014), or a PBK-cc inhibitor (such as the compound l-(4-(5- (5-amino-6-(5-tert-butyl-l,3,4-oxadiazol-2-yl)pyrazin-2-yl)-l-ethyl-lH-l,2,4-triazol-3- yl)piperidin-l-yl)-3-hydroxypropan-l-one) or palbociclib, is suitable for use in the treatment of breast or gynaecological cancers, such as cancer of the breast, endometrium, ovary or cervix, particularly breast cancer, such as ER+ve breast cancer.
Herein, where the term "combination" is used it is to be understood that this refers to simultaneous, separate or sequential administration. In one embodiment "combination" refers to simultaneous administration. In another embodiment "combination" refers to separate administration. In a further embodiment "combination" refers to sequential administration. Where the administration is sequential or separate, the delay in
administering the second component should not be such as to lose the beneficial effect of the combination. Where a combination of two or more components is administered separately or sequential, it will be understood that the dosage regime for each component may be different to and independent of the other components. Conveniently, the
compounds of the present specification are dosed once daily.
According to a further embodiment there is provided a pharmaceutical composition which comprises a compound of Formula (I) or a pharmaceutically acceptable salt thereof in combination with an anti-tumour agent selected from one listed under (i) - (xi) herein above, in association with a pharmaceutically acceptable diluent or carrier.
According to a further embodiment there is provided a pharmaceutical composition which comprises a compound of Formula (I) or a pharmaceutically acceptable salt thereof in combination with any one of antihormonal agents listed under (ii) above, for example any one of the anti-oestrogens listed in (ii) above, or for example an aromatase inhibitor listed in (ii) above in association with a pharmaceutically acceptable diluent or carrier.
In a further embodiment there is provided a pharmaceutical composition
comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and an mTOR inhibitor, such as AZD2014 (see for example WO2008/023161); in association with a pharmaceutically acceptable diluent or carrier.
In a further embodiment there is provided a pharmaceutical composition
comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and a PDK -inhibitor, such as the compound l-(4-(5-(5-amino-6-(5-tert-butyl-l,3,4- oxadiazol-2-yl)pyrazin-2-yl)- 1 -ethyl- 1H- l,2,4-triazol-3-yl)piperidin- l-yl)-3- hydroxypropan-l-one, in association with a pharmaceutically acceptable diluent or carrier.
In a further embodiment there is provided a pharmaceutical composition
comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and palbociclib in association with a pharmaceutically acceptable diluent or carrier.
According to a further embodiment there is provided a pharmaceutical composition which comprises a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an anti-tumour agent selected from one listed under (i) - (xi) herein above, in association with a pharmaceutically acceptable diluent or carrier for use in treating cancer.
According to a further embodiment there is provided a pharmaceutical composition which comprises a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with any one of antihormonal agents listed under (ii) above, for example any one of the anti-oestrogens listed in (ii) above, or for example an aromatase inhibitor listed in (ii) above in association with a pharmaceutically acceptable diluent or carrier for use in treating cancer.
In a further embodiment there is provided a pharmaceutical composition
comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and an mTOR inhibitor, such as AZD2014 (see for example WO2008/023161); in association with a pharmaceutically acceptable diluent or carrier for use in treating cancer.
In a further embodiment there is provided a pharmaceutical composition
comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and a PDK -inhibitor, such as the compound l-(4-(5-(5-amino-6-(5-tert-butyl-l,3,4- oxadiazol-2-yl)pyrazin-2-yl)- 1 -ethyl- 1H- l,2,4-triazol-3-yl)piperidin- l-yl)-3- hydroxypropan-l-one, in association with a pharmaceutically acceptable diluent or carrier for use in treating cancer.
In a further embodiment there is provided a pharmaceutical composition
comprising a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, and palbociclib in association with a pharmaceutically acceptable diluent or carrier for use in treating cancer.
In one aspect the above pharmaceutical compositions of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, with an anti-tumour agent listed in (ii) above, or an mTOR inhibitor (such as AZD2014), or a PBK-cc inhibitor (such as the compound l-(4-(5-(5-amino-6-(5-tert-butyl-l,3,4-oxadiazol-2-yl)pyrazin-2-yl)-l-ethyl-lH- l,2,4-triazol-3-yl)piperidin-l-yl)-3-hydroxypropan-l-one) or palbociclib, is suitable for use in the treatment of breast or gynaecological cancers, such as cancer of the breast, endometrium, ovary or cervix, particularly breast cancer, such as ER+ve breast cancer.
According to another embodiment there is provided the use of a compound of the Formula (I) or a pharmaceutically acceptable salt thereof in combination with an anti- tumour agent selected from one listed under (i) - (xi) herein above, in the manufacture of a medicament for use in the treatment of cancer in a warm-blooded animal, such as man.
According to a further embodiment there is provided the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in combination with any one of antihormonal agents listed under (ii) above, for example any one of the anti-oestrogens listed in (ii) above, or for example an aromatase inhibitor listed in (ii) above in the manufacture of a medicament for use in the treatment of cancer in a warm-blooded animal, such as man.
In a further embodiment there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an mTOR inhibitor, such as AZD2014 (see for example WO2008/023161); in the manufacture of a medicament for use in the treatment of cancer in a warm-blooded animal, such as man.
In a further embodiment there is provided the use of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a PBKcc-inhibitor, such as the compound l-(4-(5-(5-amino-6-(5-tert-butyl-l,3,4-oxadiazol-2-yl)pyrazin-2-yl)- l-ethyl-lH-l,2,4-triazol-3-yl)piperidin-l-yl)-3-hydroxypropan-l-one, in the manufacture of a medicament for use in the treatment of cancer in a warm-blooded animal, such as man.
In a further embodiment there is provided the use a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with palbociclib in the manufacture of a medicament for use in the treatment of cancer in a warm-blooded animal, such as man.
In one aspect the above uses of a compound of the Formula (I), or a
pharmaceutically acceptable salt thereof, in combination with an anti-tumour agent listed in (ii) above, or an mTOR inhibitor (such as AZD2014), or a PI3K-CC inhibitor (such as the compound l-(4-(5-(5-amino-6-(5-tert-butyl-l,3,4-oxadiazol-2-yl)pyrazin-2-yl)-l-ethyl-lH- l,2,4-triazol-3-yl)piperidin-l-yl)-3-hydroxypropan-l-one) or palbociclib, is suitable for use in the manufacture of a medicament for the treatment of breast or gynaecological cancers, such as cancer of the breast, endometrium, ovary or cervix, particularly breast cancer, such as ER+ve breast cancer.
Therefore in an additional feature, there is provided a method of treating cancer in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an anti-tumour agent selected from one listed under (i) - (xi) herein above.
According to a further embodiment there is provided a method of treating cancer in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in combination with any one of antihormonal agents listed under (ii) above, for example any one of the anti-oestrogens listed in (ii) above, or for example an aromatase inhibitor listed in (ii) above.
In a further embodiment there is provided a method of treating cancer in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with an mTOR inhibitor, such as AZD2014 (see for example WO2008/023161).
In a further embodiment there provided a method of treating cancer in a
warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a PBKcc-inhibitor, such as the compound l-(4-(5-(5-amino-6-(5-tert-butyl-l,3,4-oxadiazol-2-yl)pyrazin-2-yl)-l-ethyl- lH-l,2,4-triazol-3-yl)piperidin-l-yl)-3-hydroxypropan-l-one.
In a further embodiment there is provided a method of treating cancer in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in combination with palbociclib.
In one aspect the above combinations, pharmaceutical compositions, uses and methods of treating cancer, are methods for the treatment of breast or gynaecological cancers, such as cancer of the breast, endometrium, ovary or cervix, particularly breast cancer, such as ER+ve breast cancer.
According to a further embodiment there is provided a kit comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof in combination with an anti- tumour agent selected from one listed under (i) - (xi) herein above.
According to a further embodiment there is provided a kit comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof in combination with an anti- tumour agent selected from one listed under (i) or (ii) herein above.
According to a further embodiment there is provided a kit comprising:
a) a compound of Formula (I) or a pharmaceutically acceptable salt thereof in a first unit dosage form;
b) an anti-tumour agent selected from one listed under (i) - (xi) herein above in a second unit dosage form; and
c) container means for containing said first and second dosage forms.
According to a further embodiment there is provided a kit comprising:
a) a compound of Formula (I) or a pharmaceutically acceptable salt thereof in a first unit dosage form;
b) an anti-tumour agent selected from one listed under (i) - (ii) herein above in a second unit dosage form; and
c) container means for containing said first and second dosage forms.
According to a further embodiment there is provided a kit comprising:
a) a compound of the Formula (I), or a pharmaceutically acceptable salt thereof, in a first unit dosage form;
b) an anti-tumour agent selected from an anti-tumour agent listed in (ii) above, an mTOR inhibitor (such as AZD2014), a PDK -inhibitor, such as the compound l-(4-(5-(5-amino- 6-(5-tert-butyl-l,3,4-oxadiazol-2-yl)pyrazin-2-yl)-l-ethyl-lH-l,2,4-triazol-3-yl)piperidin- l-yl)-3-hydroxypropan-l-one, and palbociclib, in a second unit dosage form; and c) container means for containing said first and second dosage forms.
Combination therapy as described above may be added on top of standard of care therapy typically carried out according to its usual prescribing schedule.
Although the compounds of the Formula (I) are primarily of value as therapeutic agents for use in warm-blooded animals (including man), they are also useful whenever it is required to inhibit ER-cc. Thus, they are useful as pharmacological standards for use in the development of new biological tests and in the search for new pharmacological agents.
Personalised Healthcare
Another aspect of the present specification is based on identifying a link between the status of the gene encoding ERa and potential susceptibility to treatment with a compound of Formula (I). In particular, ERa gene status may indicate that a patient is less likely to respond to exisiting hormone therapy (such as aromatase inhibitors), in part at least because some ERa mutations are though to arise as resistance mechanisms to existing treatments. A SERD, particularly a SERD which can be administered orally in potentially larger doses without excessive inconvenince, may then advantageously be used to treat patentients with ERa mutations who may be resistant to other therapies. This therefore provides opportunities, methods and tools for selecting patients for treatment with a compound of Formula (I), particularly cancer patients. The present specification relates to patient selection tools and methods (including personalised medicine). The selection is based on whether the tumour cells to be treated possess wild-type or mutant ERa gene. The ERa gene status could therefore be used as a biomarker to indicate that selecting treatment with a SERD may be advantageous. For the avoidance of doubt, compounds of the Formula (I) as described herein are thought to be similarly active against wild-type and mutant ERa genes, at least those mutations in ERa gene identified at the date of filing this application.
There is a clear need for biomarkers that will enrich for or select patients whose tumours will respond to treatment with a SERD, such as a compound of Formula (I).
Patient selection biomarkers that identify the patients most likely to respond to one agent over another are ideal in the treatment of cancer, since they reduce the unnecessary treatment of patients with non-responding tumours to the potential side effects of such agents.
A biomarker can be described as "a characteristic that is objectively measured and evaluated as an indicator of normal biologic processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention". A biomarker is any identifiable and measurable indicator associated with a particular condition or disease where there is a correlation between the presence or level of the biomarker and some aspect of the condition or disease (including the presence of, the level or changing level of, the type of, the stage of, the susceptibility to the condition or disease, or the responsiveness to a drug used for treating the condition or disease). The correlation may be qualitative, quantitative, or both qualitative and quantitative. Typically a biomarker is a compound, compound fragment or group of compounds. Such compounds may be any compounds found in or produced by an organism, including proteins (and peptides), nucleic acids and other compounds.
Biomarkers may have a predictive power, and as such may be used to predict or detect the presence, level, type or stage of particular conditions or diseases (including the presence or level of particular microorganisms or toxins), the susceptibility (including genetic susceptibility) to particular conditions or diseases, or the response to particular treatments (including drug treatments). It is thought that biomarkers will play an increasingly important role in the future of drug discovery and development, by improving the efficiency of research and development programs. Biomarkers can be used as diagnostic agents, monitors of disease progression, monitors of treatment and predictors of clinical outcome. For example, various biomarker research projects are attempting to identify markers of specific cancers and of specific cardiovascular and immunological diseases. It is believed that the development of new validated biomarkers will lead both to significant reductions in healthcare and drug development costs and to significant improvements in treatment for a wide variety of diseases and conditions.
In order to optimally design clinical trials and to gain the most information from these trials, a biomarker may be required. The marker may be measurable in surrogate and tumour tissues. Ideally these markers will also correlate with efficacy and thus could ultimately be used for patient selection.
Thus, the technical problem underlying this aspect of the present specification is the identification of means for stratification of patients for treatment with a compound of Formula (I). The technical problem is solved by provision of the embodiments
characterized in the claims and/or description herein.
Tumours which contain wild type ERa are believed to be susceptible to treatment with a compound of Formula (I), for example as a first- line treatment. Tumours may also respond to treatment with a compound of Formula (I) as a second- line, third-line or subsequent therapy and this may be useful, in particular, where the tumours contain mutant ERa and may thus be resistant to existing therapies such as AIs. A higher dosage of a compound of Formula (I) may be required in the resistant setting than in wild type tumours).
The specification provides a method of determining sensitivity of cells to a compound of Formula (I). The method comprises determining the status of ERa gene in said cells. A cell is defined as sensitive to a compound of Formula (I) if it inhibits the increase in cell number in a cell growth assay (either through inhibition of cell proliferation and /or through increased cell death). Methods of the specification are useful for predicting which cells are more likely to respond to a compound of Formula (I) by growth inhibition.
A sample "representative of the tumour" can be the actual tumour sample isolated, or may be a sample that has been further processed, e.g. a sample of PCR amplified nucleic acid from the tumour sample.
Definitions:
In this Personalised Healthcare section:
"Allele" refers to a particular form of a genetic locus, distinguished from other forms by its particular nucleotide or amino acid sequence.
"Amplification reactions" are nucleic acid reactions which result in specific amplification of target nucleic acids over non-target nucleic acids. The polymerase chain reaction (PCR) is a well known amplification reaction.
"Cancer" is used herein to refer to neoplastic growth arising from cellular transformation to a neoplastic phenotype. Such cellular transformation often involves genetic mutation.
"Gene" is a segment of DNA that contains all the information for the regulated biosynthesis of an RNA product, including a promoter, exons, introns, and other sequence elements which may be located within 5' or 3' flanking regions (not within the transcribed portions of the gene) that control expression.
"Gene status" refers to whether the gene is wild type or not (i.e. mutant).
"Label" refers to a composition capable of producing a detectable signal indicative of the presence of the target polynucleotide in an assay sample. Suitable labels include radioisotopes, nucleotide chromophores, enzymes, substrates, fluorescent molecules, chemiluminescent moieties, magnetic particles, bioluminescent moieties, and the like. As such, a label is any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means.
"Non- synonymous variation" refers to a variation (variance) in or overlapping the coding sequence of a gene that result in the production of a distinct (altered) polypeptide sequence. These variations may or may not affect protein function and include missense variants (resulting in substitution of one amino acid for another), nonsense variants (resulting in a truncated polypeptide due to generation of a premature stop codon) and insertion/deletion variants.
"Synonymous variation" refers to a variation (variance) in the coding sequence of a gene that does not affect sequence of the encoded polypeptide. These variations may affect protein function indirectly (for example by altering expression of the gene), but, in the absence of evidence to the contrary, are generally assumed to be innocuous.
"Nucleic acid" refers to single stranded or double stranded DNA and RNA molecules including natural nucleic acids found in nature and/or modified, artificial nucleic acids having modified backbones or bases, as are known in the art.
"Primer" refers to a single stranded DNA oligonucleotide sequence capable of acting as a point of initiation for synthesis of a primer extension product which is complementary to the nucleic acid strand to be copied. The length and sequence of the primer must be such that they are able to prime the synthesis of extension products. A typical primer contains at least about 7 nucleotides in length of a sequence substantially complementary to the target sequence, but somewhat longer primers are preferred. Usually primers contain about 15-26 nucleotides, but longer or shorter primers may also be employed.
"Polymorphic site" is a position within a locus at which at least two alternative sequences are found in a population.
"Polymorphism" refers to the sequence variation observed in an individual at a polymorphic site. Polymorphisms include nucleotide substitutions, insertions, deletions and microsatellites and may, but need not, result in detectable differences in gene expression or protein function. In the absence of evidence of an effect on expression or protein function, common polymorphisms, including non-synonymous variants, are generally considered to be included in the definition of wild-type gene sequence. A catalog of human polymorphisms and associated annotation, including validation, observed frequencies, and disease association, is maintained by NCBI (dbSNP:
http://www.ncbi.nlm.nih.gov/proiects/SNP/). Please note that the term "polymorphism" when used in the context of gene sequences should not be confused with the term
"polymorphism" when used in the context of solid state form of a compound, that is the crystalline or amorphous nature of a compound. The skilled person will understand the intended meaning by its context.
"Probe" refers to single stranded sequence- specific oligonucleotides which have a sequence that is exactly complementary to the target sequence of the allele to be detected.
"Response" is defined by measurements taken according to Response Evaluation Criteria in Solid Tumours (RECIST) involving the classification of patients into two main groups: those that show a partial response or stable disease and those that show signs of progressive disease.
"Stringent hybridisation conditions" refers to an overnight incubation at 42°C in a solution comprising 50% formamide, 5x SSC (750 mM NaCI, 75 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5x Denhardt's solution, 10% dextran sulphate, and 20 pg/ml denatured, sheared salmon sperm DNA, followed by washing the filters in O.lx SSC at about 65 °C.
"Survival" encompasses a patients' overall survival and progression-free survival.
"Overall survival" (OS) is defined as the time from the initiation of drug administration to death from any cause. "Progression-free survival" (PFS) is defined as the time from the initiation of drug administration to first appearance of progressive disease or death from any cause.
According to one aspect of the specification there is provided a method for selecting a patient for treatment with a compound of Formula (I), the method comprising providing a tumour cell containing sample from a patient; determining whether the ERa gene in the patient's tumour cell containing sample is wild type or mutant; and selecting a patient for treatment with a compound of Formula (I) based thereon.
The method may include or exclude the actual patient sample isolation step. Thus, according to one aspect of the specification there is provided a method for selecting a patient for treatment with a compound of Formula (I), the method comprising determining whether the ERa gene in a tumour cell containing sample previously isolated from the patient is wild type or mutant; and selecting a patient for treatment with a compound of Formula (I) based thereon.
In one embodiment, the patient is selected for treatment with a compound of Formula (I) if the tumour cell DNA has a mutant ERa gene. In other embodiments, a patient whose tumour cell DNA possesses a wild type ERa gene is selected for treatment with a compound of Formula (I).
For the purpose of this specification, a gene status of wild-type is meant to indicate normal or appropriate expression of the gene and normal function of the encoded protein. In contrast, mutant status is meant to indicate expression of a protein with altered function, consistent with the known roles of mutant ERa genes in cancer (as described herein). Any number of genetic or epigenetic alterations, including but not limited to mutation, amplification, deletion, genomic rearrangement, or changes in methylation profile, may result in a mutant status. However, if such alterations nevertheless result in appropriate expression of the normal protein, or a functionally equivalent variant, then the gene status is regarded as wild-type. Examples of variants that typically would not result in a functional mutant gene status include synonymous coding variants and common polymorphisms (synonymous or non-synonymous). As discussed below, gene status can be assessed by a functional assay, or it may be inferred from the nature of detected deviations from a reference sequence.
In certain embodiments the wild-type or mutant status of the ERa gene is determined by the presence or absence of non-synonymous nucleic acid variations in the genes. Observed non-synonymous variations corresponding to known common
polymorphisms with no annotated functional effects do not contribute to a gene status of mutant.
Other variations in the ERa gene that signify mutant status include splice site variations that decrease recognition of an intron/exon junction during processing of pre- mRNA to mRNA. This can result in exon skipping or the inclusion of normally intronic sequence in spliced mRNA (intron retention or utilization of cryptic splice junctions). This can, in turn, result in the production of aberrant protein with insertions and/or deletions relative to the normal protein. Thus, in other embodiments, the gene has a mutant status if there is a variant that alters splice site recognition sequence at an intron/exon junction. For ESR1, reference sequences are available for the gene (GenBank accession number: NG_008493), mRNA (GenBank accession number: NM_000125), and protein (GenBank accession number: NP_000116 or Swiss-Prot accession: P03372). A person of skill in the art will be able to determine the ESR1 gene status, i.e. whether a particular ESRlgene is wild type or mutant, based on comparison of DNA or protein sequence with wild type.
It will be apparent that the gene and mRNA sequences disclosed for ERa gene are representative sequences. In normal individuals there are two copies of each gene, a maternal and paternal copy, which will likely have some sequence differences, moreover within a population there will exist numerous allelic variants of the gene sequence. Other sequences regarded as wild type include those that possess one or more synonymous changes to the nucleic acid sequence (which changes do not alter the encoded protein sequence), non-synonymous common polymorphisms (e.g. germ-line polymorphisms) which alter the protein sequence but do not affect protein function, and intronic non-splice- site sequence changes.
There are numerous techniques available to the person skilled in the art to determine the gene status of ERa. The gene status can be determined by determination of the nucleic acid sequence. This could be via direct sequencing of the full-length gene or analysis of specific sites within the gene, e.g. commonly mutated sites.
Samples
The patient's sample to be tested for the gene status can be any tumour tissue or tumour-cell containing sample obtained or obtainable from the individual. The test sample is conveniently a sample of blood, mouth swab, biopsy, or other body fluid or tissue obtained from an individual. Particular examples include: circulating tumour cells, circulating DNA in the plasma or serum, cells isolated from the ascites fluid of ovarian cancer patients, lung sputum for patients with tumours within the lung, a fine needle aspirate from a breast cancer patient, urine, peripheral blood, a cell scraping, a hair follicle, a skin punch or a buccal sample.
It will be appreciated that the test sample may equally be a nucleic acid sequence corresponding to the sequence in the test sample, that is to say that all or a part of the region in the sample nucleic acid may firstly be amplified using any convenient technique e.g. polymerase chain reaction (PCR), before analysis. The nucleic acid may be genomic DNA or fractionated or whole cell RNA. In particular embodiments the RNA is whole cell RNA and is used directly as the template for labelling a first strand cDNA using random primers or poly A primers. The nucleic acid or protein in the test sample may be extracted from the sample according to standard methodologies (see Green & Sambrook,
Eds., Molecular Cloning: A Laboratory Manual, (2012, 4th edition, Vol. 1-3, ISBN
9781936113422), Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.)
The diagnostic methods of the specification can be undertaken using a sample previously taken from the individual or patient. Such samples may be preserved by freezing or fixed and embedded in formalin-paraffin or other media. Alternatively, a fresh tumour cell containing sample may be obtained and used.
The methods of the specification can be applied using cells from any tumour. Suitable tumours for treatment with a compound of Formula (I) have been described hereinbefore.
Methods for Detection of Nucleic Acids
The detection of mutant ERa nucleic acids can be employed, in the context of the present specification, to select drug treatment. Since mutations in these genes occur at the DNA level, the methods of the specification can be based on detection of mutations or variances in genomic DNA, as well as transcripts and proteins themselves. It can be desirable to confirm mutations in genomic DNA by analysis of transcripts and/or polypeptides, in order to ensure that the detected mutation is indeed expressed in the subject.
It will be apparent to the person skilled in the art that there are a large number of analytical procedures which may be used to detect the presence or absence of variant nucleotides at one or more positions in a gene. In general, the detection of allelic variation requires a mutation discrimination technique, optionally an amplification reaction (such as one based on polymerase chain reaction) and optionally a signal generation system. There are a multitude of mutation detection techniques available in the art and these may be used in combination with a signal generation system, of which there are numerous available in the art. Many methods for the detection of allelic variation are reviewed by Nollau et al., Clin. Chem., 1997, 43, 1114-1120; Anderson SM. Expert Rev Mol Diagn., 2011, 11, 635- 642; Meyerson M. et al., Nat Rev Genet, 2010, 11, 685-696; and in standard textbooks, for example "Laboratory Protocols for Mutation Detection", Ed. by U. Landegren, Oxford University Press, 1996 and "PCR", 2nd Edition by Newton & Graham, BIOS Scientific Publishers Limited, 1997.
As noted above, determining the presence or absence of a particular variance or plurality of variances in the ERa gene in a patient with cancer can be performed in a variety of ways. Such tests are commonly performed using DNA or RNA collected from biological samples, e.g., tissue biopsies, urine, stool, sputum, blood, cells, tissue scrapings, breast aspirates or other cellular materials, and can be performed by a variety of methods including, but not limited to, PCR, hybridization with allele- specific probes, enzymatic mutation detection, chemical cleavage of mismatches, mass spectrometry or DNA sequencing, including minisequencing.
Suitable mutation detection techniques include amplification refractory mutation system (ARMS™), amplification refractory mutation system linear extension (ALEX™), competitive oligonucleotide priming system (COPS), Taqman, Molecular Beacons, restriction fragment length polymorphism (RFLP), and restriction site based PCR and fluorescence resonance energy transfer (FRET) techniques.
In particular embodiments the method employed for determining the nucleotide(s) within a biomarker gene is selected from: allele- specific amplification (allele specific PCR) - such as amplification refractory mutation system (ARMS), sequencing, allelic discrimination assay, hybridisation, restriction fragment length polymorphism (RFLP) or oligonucleotide ligation assay (OLA).
In particular embodiments, hybridization with allele specific probes can be conducted by: (1) allele specific oligonucleotides bound to a solid phase (e.g. glass, silicon, nylon membranes) with the labelled sample in solution, for example as in many DNA chip applications; or, (2) bound sample (often cloned DNA or PCR amplified DNA) and labelled oligonucleotides in solution (either allele specific or short so as to allow
sequencing by hybridization). Diagnostic tests may involve a panel of variances, often on a solid support, which enables the simultaneous determination of more than one variance. Such hybridization probes are well known in the art (see, e.g., Green & Sambrook, Eds., Molecular Cloning: A Laboratory Manual, (2012, 4th edition, Vol. 1-3, ISBN 9781936113422), Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.) and may span two or more variance sites.
Thus, in one embodiment, the detection of the presence or absence of at least one mutation provides for contacting ERa nucleic acid containing a putative mutation site with at least one nucleic acid probe. The probe preferentially hybridizes with a nucleic acid sequence including a variance site and containing complementary nucleotide bases at the variance site under selective hybridization conditions. Hybridization can be detected with a detectable label using labels known to one skilled in the art. Such labels include, but are not limited to radioactive, fluorescent, dye, and enzymatic labels.
In another embodiment, the detection of the presence or absence of at least one mutation provides for contacting ERa nucleic acid containing a putative mutation site with at least one nucleic acid primer. The primer preferentially hybridizes with a nucleic acid sequence including a variance site and containing complementary nucleotide bases at the variance site under selective hybridization conditions.
Oligonucleotides used as primers for specific amplification may carry the complementary nucleotide base to the mutation of interest in the centre of the molecule (so that amplification depends on differential hybridization; see, e.g., Gibbs, et al., 1989. Nucl. Acids Res., 17, 2437-248) or at the extreme 3'-terminus of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (see, e.g., Prossner, 1993, Tibtech, 11 238).
In yet another embodiment, the detection of the presence or absence of at least one mutation comprises sequencing at least one nucleic acid sequence and comparing the obtained sequence with the known wild type nucleic acid sequence.
Alternatively, the presence or absence of at least one mutation comprises mass spectrometric determination of at least one nucleic acid sequence.
In one embodiment, the detection of the presence or absence of at least one nucleic acid variance comprises performing a polymerase chain reaction (PCR). The target nucleic acid sequence containing the hypothetical variance is amplified and the nucleotide sequence of the amplified nucleic acid is determined. Determining the nucleotide sequence of the amplified nucleic acid comprises sequencing at least one nucleic acid segment. Alternatively, amplification products can be analysed using any method capable of separating the amplification products according to their size, including automated and manual gel electrophoresis, and the like.
Mutations in genomic nucleic acid are advantageously detected by techniques based on mobility shift in amplified nucleic acid fragments. For instance, Chen et al., Anal Biochem 1996, 239, 61-9, describe the detection of single-base mutations by a competitive mobility shift assay. Moreover, assays based on the technique of Marcelino et al.,
BioTechniques 1999, 26, 1134-1148 are available commercially.
In a particular example, capillary heteroduplex analysis may be used to detect the presence of mutations based on mobility shift of duplex nucleic acids in capillary systems as a result of the presence of mismatches.
Generation of nucleic acids for analysis from samples generally requires nucleic acid amplification. Many amplification methods rely on an enzymatic chain reaction (such as a polymerase chain reaction, a ligase chain reaction, or a self- sustained sequence replication) or from the replication of all or part of the vector into which it has been cloned. Preferably, the amplification according to the specification is an exponential amplification, as exhibited by for example the polymerase chain reaction.
Many target and signal amplification methods have been described in the literature, for example, general reviews of these methods in Landegren, U. , et al., Science, 1988 242, 229-237 and Lewis, R., Genetic Engineering News 1990, 10, 54-55. These amplification methods can be used in the methods of our specification, and include polymerase chain reaction (PCR), PCR in situ, ligase amplification reaction (LAR), ligase hybridisation, QP bacteriophage replicase, transcription-based amplification system (TAS), genomic amplification with transcript sequencing (GAWTS), nucleic acid sequence-based amplification (NASBA) and in situ hybridisation. Primers suitable for use in various amplification techniques can be prepared according to methods known in the art.
Polymerase Chain Reaction (PCR) PCR is a nucleic acid amplification method described inter alia in U.S. Pat. Nos. 4,683,195 and 4,683,202. PCR consists of repeated cycles of DNA polymerase generated primer extension reactions. The target DNA is heat denatured and two oligonucleotides, which bracket the target sequence on opposite strands of the DNA to be amplified, are hybridised. These oligonucleotides become primers for use with DNA polymerase. The DNA is copied by primer extension to make a second copy of both strands. By repeating the cycle of heat denaturation, primer hybridisation and extension, the target DNA can be amplified a million fold or more in about two to four hours. PCR is a molecular biology tool, which must be used in conjunction with a detection technique to determine the results of amplification. An advantage of PCR is that it increases sensitivity by amplifying the amount of target DNA by 1 million to 1 billion fold in approximately 4 hours. PCR can be used to amplify any known nucleic acid in a diagnostic context (Mok et al., Gvnaecologic Oncology, 1994, 52: 247-252,).
An allele specific amplification technique such as Amplification Refractory Mutation System (ARMS™) (Newton et al., Nucleic Acids Res., 1989, 17, 2503-2516) can also be used to detect single base mutations. Under the appropriate PCR amplification conditions a single base mismatch located at the 3'-end of the primer is sufficient for preferential amplification of the perfectly matched allele (Newton et al., 1989, supra), allowing the discrimination of closely related species. The basis of an amplification system using the primers described above is that oligonucleotides with a mismatched 3'-residue will not function as primers in the PCR under appropriate conditions. This amplification system allows genotyping solely by inspection of reaction mixtures after agarose gel electrophoresis.
Analysis of amplification products can be performed using any method capable of separating the amplification products according to their size, including automated and manual gel electrophoresis, mass spectrometry, and the like.
The methods of nucleic acid isolation, amplification and analysis are routine for one skilled in the art and examples of protocols can be found, for example, Green & Sambrook, Eds., Molecular Cloning: A Laboratory Manual, (2012, 4th edition, Vol. 1-3, ISBN 9781936113422), Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.) Particularly useful protocol source for methods used in PCR amplification is PCR (Basics: From Background to Bench) by M. J. McPherson, S. G. Mailer, R. Beynon, C. Howe, Springer Verlag; 1st edition (October 15, 2000), ISBN: 0387916008.
The present specification also provides predictive and diagnostic kits comprising degenerate primers to amplify a target nucleic acid in the ERa gene and instructions comprising; amplification protocol and analysis of the results. The kit may alternatively also comprise buffers, enzymes, and containers for performing the amplification and analysis of the amplification products. The kit may also be a component of a screening, or diagnostic kit comprising other tools such as DNA microarrays, or other supports. Preferably, the kit also provides one or more control templates, such as nucleic acids isolated from normal tissue sample, and/or a series of samples representing different variances in the reference genes.
In one embodiment, the kit provides two or more primer pairs, each pair capable of amplifying a different region of the reference (ERa) gene (each region a site of potential variance) thereby providing a kit for analysis of expression of several gene variances in a biological sample in one reaction or several parallel reactions.
Primers in the kits may be labelled, for example fluorescently labelled, to facilitate detection of the amplification products and consequent analysis of the nucleic acid variances. The kit may also allow for more than one variance to be detected in one analysis. A combination kit will therefore comprise of primers capable of amplifying different segments of the reference gene. The primers may be differentially labelled, for example using different fluorescent labels, so as to differentiate between the variances.
In another aspect, the specification provides a method of treating a patient suffering from cancer comprising: determining the mutant or wild type status of the ERa gene in the patient' s tumour cells and if the ERa gene is mutant, administering to the patient an effective amount of a compound of Formula (I).
As used herein, the terms "effective" and "effectiveness" includes both
pharmacological effectiveness and physiological safety. Pharmacological effectiveness refers to the ability of the treatment to result in a desired biological effect in the patient. Physiological safety refers to the level of toxicity, or other adverse physiological effects at the cellular, organ and/or organism level (often referred to as side-effects) resulting from administration of the treatment. "Less effective" means that the treatment results in a therapeutically significant lower level of pharmacological effectiveness and/or a therapeutically greater level of adverse physiological effects.
According to another aspect of the specification there is provided the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof to treat a cancer patient whose tumour cells have been identified as possessing a mutant ERa gene.
According to another aspect of the specification there is provided a compound of Formula (I) or a pharmaceutically acceptable salt thereof for treating cancers with tumour cells identified as harbouring mutant ERa gene According to another aspect of the specification there is provided a method of treating cancers with tumour cells identified as harbouring mutant ERa gene comprising administering an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
In still further embodiments, the specification relates to a pharmaceutical composition comprising a compound of Formula (I) for use in the prevention and treatment of cancer with tumour cells identified as harbouring a mutant ERa gene.
For all the aspects above, mutant forms of ERa determined/identified are at all positions across the gene.
For all the aspects above, using tumours such as breast cancer as an example, particular mutant forms of ERa determined/identified are those at positions Ser463Pro, Val543Glu, Leu536Arg, Tyr537Ser, Tyr537Asn and Asp538Gly.
Examples
The specification will now be illustrated in the following Examples in which, generally:
(i) operations were carried out at ambient temperature, i.e. in the range 17 to 25°C and under an atmosphere of an inert gas such as nitrogen unless otherwise stated;
(ii) evaporations were carried out by rotary evaporation or utilising Genevac equipment or Biotage vlO evaporator in vacuo and work-up procedures were carried out after removal of residual solids by filtration;
(iii) flash chromatography purifications were performed on an
automated Teledyne Isco CombiFlash® Rf or Teledyne Isco CombiFlash® Companion® using prepacked RediSep Rf Gold™ Silica Columns (20-40 μιη, spherical particles), GraceResolv™ Cartridges (Davisil® silica) or Silicycle cartridges (40 - 63 μιη).
(iv) preparative chromatography was performed on a Gilson prep HPLC instrument with UV collection;
(v) chiral preparative chromatography was performed on a Gilson instrument with UV collection (233 injector / fraction collector, 333 & 334 pumps, 155 UV detector) or a Varian Prep Star instrument (2 x SD1 pumps, 325 UV detector, 701 fraction collector) pump running with Gilson 305 injection;
(vi) yields, where present, are not necessarily the maximum attainable; (vii) in general, the structures of end-products of the Formula (I) were confirmed by nuclear magnetic resonance (NMR) spectroscopy; NMR chemical shift values were measured on the delta scale [proton magnetic resonance spectra were determined using a Bruker Avance 500 (500 MHz) or Bruker Avance 400 (400 MHz) instrument];
measurements were taken at ambient temperature unless otherwise specified; the following abbreviations have been used: s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; dd, doublet of doublets; ddd, doublet of doublet of doublet; dt, doublet of triplets; bs, broad signal
(viii) in general, end-products of the Formula (I) were also characterised by mass spectroscopy following liquid chromatography (LCMS or UPLC); UPLC was carried out using a Waters UPLC fitted with Waters SQ mass spectrometer (Column temp 40, UV = 220-300nm, Mass Spec = ESI with positive/negative switching) at a flow rate of lml/min using a solvent system of 97% A + 3% B to 3% A to 97% B over 1.50mins (total runtime with equilibration back to starting conditions etc 1.70min), where A = 0.1% formic acid in water (for acid work) or 0.1% ammonia in water (for base work) B = acetonitrile. For acid analysis the column used was Waters Acquity HSS T3 1.8μιη 2.1 x50 mm, for base analysis the column used was Waters Acquity BEH 1.7μιη 2.1x50mm; LCMS was carried out using a Waters Alliance HT (2795) fitted with a Waters ZQ ESCi mass spectrometer and a Phenomenex Gemini -NX (50x2.1mm 5μιη) column at a flow rate of 1. lml/min 95%A to 95%B over 4 min with a 0.5 min hold. The modifier is kept at a constant 5% C (50:50 acetonitrile: water 0.1% formic acid) or D (50:50 acetonitrile: water 0.1% ammonium hydroxide (0.88 SG) depending on whether it is an acidic or basic method.
(ix) ion exchange purification was generally performed using a SCX-2 (Biotage, Propylsulfonic acid functionalized silica. Manufactured using a trifunctional silane. Non end-capped) cartridge.
(x) intermediate purity was assessed by thin layer chromatographic, mass spectral, HPLC (high performance liquid chromatography) and/or NMR analysis;
(xi) the following abbreviations have been used:- aq. aqueous
n-BuLi n-butyl lithium
tBuOH tert-butanol
CDC deutero-chloroform Cone. concentrated
DCM dichloromethane
DIPEA diisopropylethylamine
DMA N,N-dimethylacetamide
DMAP dimethylaminopyridine
DMSO dimethyl sulphoxide
EtOH ethanol
EtOAc ethyl acetate
MeOH methanol
rt/RT room temperature
sat. saturated
sol. solution
THF tetrahydrofuran
Example 1 : (E)-3-(4-((6^,8R)-7- ,2-difluoropropyl)-8-methyl-6J,8,9-tetrahvdro-3H- Pyrazolor4,3-f1iso uinolin-6-yl)- -difluorophenyl)acrylic acid
Figure imgf000077_0001
Aqueous sodium hydroxide (2 M; 18.69 mL, 37.38 mmol) was added slowly to a solution of methyl (E)-3-(4-((65,8R)-7-(2,2-dif uoropropyl)-8-methyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3,5-difluorophenyl)acrylate (3.45 g, 7.48 mmol) in methanol (40 mL) and THF (20 mL). The reaction mixture was stirred at room
temperature for 1 hour. The reaction mixture was diluted with water (150 mL) and adjusted to ~pH 6 with aqueous HC1 (2 M). The mixture was extracted with EtOAc (3 x 150 mL) and the combined organic layers were washed with saturated aqueous sodium chloride (30 mL), dried over magnesium sulfate, filtered and concentrated. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 75% EtOAc in heptane. Product fractions were evaporated to dryness to afford (E)-3-(4-((65',8R)-7-(2,2- difluoropropyl)-8-methyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3,5- difluorophenyl)acrylic acid (2.45 g, 73%) as a white solid. XH NMR (500 MHz, CDC13, 27 °C) 1.10 (3H, d), 1.42 (3H, t), 2.63 (1H, q), 2.95 (1H, dd), 3.10 (1H, dt), 3.47 (1H, dd), 3.64 - 3.73 (1H, m), 5.38 (1H, s), 6.43 (1H, d), 6.80 (1H, d), 7.02 (2H, d), 7.21 (1H, d), 7.62 (1H, d), 8.09 (1H, d). m/z: ES+ [M+H]+ 448.
The methyl (E)-3-(4-((65,8R)-7-(2,2-difluoropropyl)-8-methyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3,5-difluorophenyl)acrylate used as starting material was prepared as follows;
Preparation of (R)-l-(3-bromo-2-methylphenyl)propan-2-amine
Figure imgf000078_0001
n-BuLi in hexanes (1.6 M; 16.1 mL, 25.8 mmol) was added dropwise via syringe pump over 40 minutes to a stirred solution of l,3-dibromo-2-methylbenzene (6.13 g, 24.5 mmol) in THF (60 mL) at -78 °C. After 30 minutes, tert-butyl (R)-4-methyl-l,2,3-oxathiazolidine- 3-carboxylate 2,2-dioxide (6.40 g, 27.0 mmol) was added in portions and the reaction was stirred for 30 minutes before being allowed to warm to 0 °C over -45 minutes. The reaction was stirred at 0 °C for 30 minutes and then treated with a aqueous citric acid (IN; 40 mL) and the mixture was stirred under these conditions for 15 minutes. The mixture was diluted with EtOAc, the phases separated and the aqueous phase was extracted with EtOAc (2 x 25 mL). The combined organic layers were washed with saturated aqueous sodium chloride, dried over sodium sulfate and evaporated. The resultant residue was treated with HC1 in dioxane (4N; 30 mL), stirred for 45 minutes then concentrated under reduced pressure. The resulting residue was dissolved in water (100 mL) and extracted with diethyl ether (2 x 25 mL). The aqueous phase was then basified by addition of solid sodium carbonate and extracted with DCM (3 x 30 mL). The combined DCM extracts were dried over magnesium sulfate, filtered and concentrated under reduced pressure to afford (R)- l-(3-bromo-2-methylphenyl)propan-2-amine (4.0 g, 71%) as light yellow oil. The material was used in the next step without further purification. XH NMR (300 MHz, CDC , 27 °C) 1.15 (3H, d), 1.98 (2H, br s), 2.42 (3H, s), 2.60 - 2.69 (1H, m), 2.74 - 2.84 (1H, m), 3.11 - 3.24 (1H, m), 6.95 - 7.03 (1H, m), 7.07 - 7.13 (1H, m), 7.45 (1H, dd). m/z: ES+ [M+H]+ 228.
Preparation of 2,2-difluoropropyl trifluoromethanesulfonate
Figure imgf000079_0001
2,6-Lutidine (1.5 mL, 13 mmol) was added to a stirred solution of 2,2-difluoropropan-l-ol (1.0 g, 10 mmol) in DCM (50 mL) at - 10 °C. Trifluoromethanesulfonic anhydride in DCM (1 M; 11 mL, 11 mmol) was then added dropwise via syringe over 10 minutes. The solution was allowed to stir at - 10 °C for 2 hours and then washed with cold aqueous HC1 (IN; 2 x 40 mL) and saturated aqueous sodium bicarbonate (2 x 20 mL). The combined organic layers were dried over magnesium sulfate, filtered and concentrated under reduced pressure (bath temperature at 25 °C) to afford 2,2-difluoropropyl trifluoromethanesulfonate (1.4 g, 61 %) as a pale pink oil. The material was used without further purification. XH NMR (300 MHz, CDCb, 27 °C) 1.76 (3H, t), 4.51 (2H, t).
Preparation of (R)-N-(l -(3-bromo-2-methylphenyl)propan-2-yl)-2,2-difluoropropan-
1 -amine
Figure imgf000079_0002
A solution of 2,2-difluoropropyl trifluoromethanesulfonate (1.34 g, 4.99 mmol) in DCM (3 mL) was added dropwise to a stirred solution of (R)-l-(3-bromo-2-methylphenyl)propan-2- amine (1.15 g, 4.54 mmol) and DIEA (0.951 mL, 5.44 mmol) in DCM (17 mL). The reaction was stirred at ambient temperature for 14 hours and then heated under microwave conditions at 90 °C for 1.5 hours. The mixture was cooled, diluted with DCM (20 mL) and washed with water (2 x 30 mL). The combined aqueous phases were extracted with DCM (1 x 20 mL) and the combined organic layers were combined, dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 3 to 25% EtOAc in hexanes. Fractions containing the desired product were combined and concentrated under reduced pressure to afford (R)-N-(l-(3-bromo-2-methylphenyl)propan-2-yl)-2,2-difluoropropan- l- amine (0.93 g, 67%) as a pale yellow oil. lH NMR (300 MHz, CDC13, 27 °C) 1.08 (3H, d), 1.60 (3H, app t), 2.42 (3H, s), 2.65 (1H, dd), 2.79 - 3.09 (m, 4 H), 6.90 - 7.02 (1H, m), 7.04 - 7.16 (1H, m), 7.44 (1H, d). NH not observed, m/z: ES+ [M+] 306.
Preparation of (R)-N-(l -(3-((diphenylmethylene)amino)-2-methylphenyl)propan-2- -2,2-difluoropropan-l-amine
Figure imgf000080_0001
(R)-N-(l-(3-Bromo-2-methylphenyl)propan-2-yl)-2,2-difluoropropan-l-amine (5.66 g, 18.5 mmol), diphenylmethanimine (3.41 mL, 20.3 mmol), sodium 2-methylpropan-2-olate (2.66 g, 27.7 mmol) and 2,2'-bis(diphenylphosphanyl)- l,l'-binaphthalene (0.46 g, 0.74 mmol) were added to a round bottom flask and suspended in toluene (80 mL). The solvent was evacuated and back-filled with nitrogen (3x) and then Pd2(dba)3 (0.34 g, 0.37 mmol) was added. The reaction was heated at 90 °C for 3 hr. After cooling, the reaction mixture was filtered through celite, the solids were washed with DCM and the filtrate was evaporated to a red-brown oil. The resulting residue was dissolved in DCM (100 mL), washed with water (2 x 50 mL), the combined aqueous phases were extracted with DCM (2 x 40 mL). The combined organic layers were dried over magnesium sulfate, filtered and the filtrate concentrated to a brown oil. The oil was purified by flash silica
chromatography, elution gradient 0 to 25% EtOAc in heptane. Product fractions were evaporated to dryness to afford (R)-N-(l-(3-((diphenylmethylene)amino)-2- methylphenyl)propan-2-yl)-2,2-difluoropropan- l -amine (6.92 g, 92%) as a clear yellow oil. XH NMR (500 MHz, CDCI3, 27 °C) 0.99 (3H, d), 1.57 (3H, t), 2.15 (3H, s), 2.58 (1H, dd), 2.72 (1H, dd), 2.76 - 2.93 (3H, m), 6.33 (1H, dd), 6.72 (1H, dd), 6.82 - 6.87 (1H, m), 7.03 - 7.08 (2H, m), 7.19 - 7.25 (3H, m), 7.41 (2H, ddt), 7.44 - 7.50 (1H, m), 7.76 - 7.81 (2H, m). m/z: ES+ [M+H]+ 407. reparation of (R)-3-(2-((2,2-difluoropropyl)amino)propyl)-2-methylaniline
Figure imgf000081_0001
Aqueous hydrogen chloride solution (1 M; 68.4 mL, 68.4 mmol) was added to (R)-N-(l-(3- ((diphenylmethylene)amino)-2-methylphenyl)propan-2-yl)-2,2-difluoropropan-l-amine (6.95 g, 17.1 mmol) in DCM (75 mL) and the biphasic mixture stirred rapidly for 1 hour. The layers were separated and the aqueous layer was extracted with DCM (2 x 40 mL). The aqueous layer was then basified by addition of saturated aqueous sodium carbonate and extracted with DCM (3 x 50 mL). The combined organic extracts were dried over magnesium sulfate, filtered and concentrated to afford (R)-3-(2-((2,2- difluoropropyl)amino)propyl)-2-methylaniline (3.72 g, 90%) as a yellow oil. XH NMR (500
MHz, CDCls, 27 °C) 1.06 (3H, d), 1.58 (3H, t), 2.11 (3H, s), 2.59 (1H, dd), 2.77 (1H, dd), 2.82 - 3.00 (3H, m), 3.59 (2H, s), 6.54 - 6.65 (2H, m), 6.95 (1H, t). mJz: ES+ [M+H]+
243.
Preparation of methyl (E)-3-(4-((l^,3R)-6-amino-2-(2,2-difluoropropyl)-3,5-dimethyl- -tetrahvdroiso uinolin-l-yl)-3,5-difluorophenyl)acrylate
Figure imgf000081_0002
Methyl (E)-3-(3,5-difluoro-4-formylphenyl)acrylate (6.91 g, 30.5 mmol) was added to a solution of (R)-3-(2-((2,2-difluoropropyl)amino)propyl)-2-methylaniline (3.70 g, 15.3 mmol) in acetic acid (80 mL) and water (1.38 mL). The reaction mixture was heated at 60
°C for 3 hours and then evaporated to a dark brown oil. The oil was dissolved in EtOAc (80 mL) and washed with saturated aqeuous sodium bicarbonate (3 x 75 mL). The combined aqueous layers were extracted with EtOAc (2 x 50 mL) and the combined organic layers were dried over magnesium sulfate, filtered and then concentrated to dryness. The resulting residue was dissolved in DCM (75 mL). Aqueous HC1 (2 M; 100 mL) was added and the biphasic mixture was stirred vigorously for 1 hour. The layers were separated and the aqueous layer was extracted with DCM (2 x 50 mL). The aqueous layer was then basified by addition of sodium carbonate and extracted with EtOAc (3 x 75 mL). The combined basic extracts were washed with saturated aqueous sodium chloride (50 mL), dried over magnesium sulfate, filtered and concentrated. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 50% EtOAc in heptane. Product fractions were evaporated to dryness to afford methyl (E)-3-(4-((15,,3R)-6-amino- 2-(2,2-difluoropropyl)-3,5-dimethyl- 1,2,3,4- tetrahydroisoquinolin-l-yl)-3,5- difluorophenyl)acrylate (3.91 g, 57%) as a white foam. XH NMR (500 MHz, CDC13, 27 °C) 1.04 (3H, d), 1.40 (3H, t), 2.07 (3H, s), 2.48 - 2.66 (2H, m), 2.95 - 3.08 (2H, m), 3.49 - 3.58 (3H, m), 3.80 (3H, s), 5.22 (IH, s), 6.37 (IH, d), 6.44 (2H, s), 6.96 (2H, d), 7.53 (IH, d). m/z ES+ [M+H]+ 451.
Preparation of methyl (E)-3-(4-((6^,8R)-7-(2,2-difluoropropyl)-8-methyl-6J,8,9- tetrahvdro-3H-pyrazolor4,3-f1iso uinolin-6-yl)-3,5-difluorophenyl)acrylate
Figure imgf000082_0001
Methyl (E)-3-(4-((15,3R)-6-amino-2-(2,2-difluoropropyl)-3,5-dimethyl-l,2,3,4- tetrahydroisoquinolin-l-yl)-3,5-difluorophenyl)acrylate (2.73 g, 6.06 mmol) in propionic acid (25 mL) was cooled to - 17 °C using a dry ice/acetone bath in a 3-necked flask.
Sodium nitrite (0.418 g, 6.06 mmol) in water (2.5 mL) was added dropwise over 2-3 minutes with no change in internal temperature. The reaction mixture was stirred at - 17 °C for 30 minutes and then diluted with ice-cold EtOAc (150 mL) with a maximum
temperature rise to -4 °C. The reaction mixture was stirred vigorously and neutralised by slow addition via dropping funnel of saturated aqueous sodium bicarbonate (200 mL) keeping the internal temperature at ~5 °C. The layers were separated and the organic layer was washed further with saturated aqueous sodium bicarbonate (200 mL), followed by saturated aqueous sodium chloride (50 mL). The combined aqueous phases were extracted with EtOAc (2 x 75 mL) and the combined organic layers were dried over magnesium sulfate, filtered and concentrated to a brown oil. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 45% EtOAc in heptane. Product fractions were evaporated to dryness to afford methyl (E)-3-(4-((6S,8R)-7-(2,2-difluoropropyl)-8-methyl- 6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3,5-difluorophenyl)acrylate (1.9 g, 69%) as a beige coloured solid. lH NMR (500 MHz, CDC13, 27 °C) 1.09 (3H, d), 1.42 (3H, t), 2.62 (1H, q), 2.95 (1H, dd), 3.10 (1H, dt), 3.46 (1H, dd), 3.64 - 3.73 (1H, m), 3.81 (3H, s), 5.37 (1H, s), 6.39 (1H, d), 6.78 (1H, d), 6.99 (2H, d), 7.19 (1H, d), 7.54 (1H, d), 8.08 (1H, d), 10.02 (1H, s). m/z: ES+ [M+H]+ 462.
Example 2: (E)-3-(3,5-difluoro-4-((6^,8R)-7- -fluoro-2-methylpropyl)-6,8-dimethyl- -tetrahvdro-3H-pyrazolor4,3-f1iso uinolin-6-yl)phenyl)acrylic acid
Figure imgf000083_0001
Cerium (IV) ammonium nitrate (0.932 g, 1.70 mmol) was added in portions to a solution of
Figure imgf000083_0002
7-(4-((E)-3-(iert-butoxy)-3-oxoprop- l-en-l-yl)-2,6-difluorophenyl)-6- (2-fluoro-2-methylpropyl)-5-methyl-4,5,6,7-tetrahydro- lH-pyrazolo[4,3-f]isoquinoline- l- carboxylate (0.510 g, 0.85 mmol) in acetonitrile (6.8 mL) and water (1.7 mL). The reaction was stirred at room temperature for 2 hours and then diluted with EtOAc and aqueous HC1 (IN). The layers were separated and the aqueous layer was extracted with EtOAc. The organic layer was washed with saturated aqueous sodium chloride, dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM. Product fractions were evaporated to dryness to afford a yellow solid (541 mg).
Methylmagnesium bromide (3 M in diethyl ether; 0.167 mL, 0.50 mmol) was added to a solution of the yellow solid (200 mg, 0.33 mmol) in THF (3.2 mL) at 0 °C. The reaction was stirred for 30 min and then warmed to room temperature over 30 min before being quenched by addition of methanol (1 mL). The volatiles were evaporated and the resulting residue was stirred in HC1 in dioxane (4N; 4 mL) at room temperature for 30 mins. The solution was passed through an SCX-2 column, eluting with methanol and then with NH3 in MeOH (1M). The basic filtrate was concentrated under reduced pressure and the resulting residue was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM. Product fractions were concentrated to dryness to afford (E)-3-(3,5- difluoro-4-((65',8R)-7-(2-fluoro-2-methylpropyl)-6,8-dimethyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)phenyl)acrylic acid (26.9 mg, 18%) as a yellow-brown solid. XH NMR (400 MHz, CDC13, 27 °C) 0.97 (3H, d), 1.15 (3H, d), 1.23 (3H, d), 1.91 (3H, s), 2.47 (1H, dd), 2.99 (1H, d), 3.18 (1H, t), 3.48 - 3.59 (1H, m), 3.94 (1H, s), 6.37 (1H, d), 6.89 (2H, d), 6.94 (1H, d), 7.21 (1H, d), 7.55 (1H, d), 8.12 (1H, s). m/z: ES+
[M+H]+ 458.
The
Figure imgf000084_0001
7-(4-((E)-3-(iert-butoxy)-3-oxoprop- 1-en- l-yl)-2,6-difluorophenyl)- 6-(2-fluoro-2-methylpropyl)-5-methyl-4,5,6,7-tetrahydro-lH-pyrazolo[4,3-f]isoquinoline- 1-carboxylate used as starting material was prepared as follows;
Preparation of (5R,7S)-tert-butyl 7-(4-((E)-3-(fert-butoxy)-3-oxoprop-l-en-l-yl)-2,6- difluorophenyl)-6-(2-fluoro-2-methylpropyl)-5-methyl-4,5,6,7-tetrahvdro-lH- pyrazolor4,3-flisoquinoline-l-carboxylate
Figure imgf000084_0002
Di-ie/t-butyl dicarbonate (517 mg, 2.37 mmol) was added to a solution of (E)-3-(3,5- difluoro-4-((65,,8R)-7-(2-fluoro-2-methylpropyl)-8-methyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)phenyl)acrylic acid (350 mg, 0.79 mmol) and 4- dimethylaminopyridine (148 mg, 1.18 mmol) in iert-BuOH (5.3 mL). The reaction was heated to 80 °C and maintained under these conditions for 4 hours. After cooling, the volatiles were evaporated and the crude product was purified by flash silica
chromatography, elution gradient 0 to 40% EtOAc in heptane. Product fractions were evaporated to dryness to afford
Figure imgf000085_0001
7-(4-((E)-3-(iert-butoxy)-3-oxoprop- l- en- l-yl)-2,6-difluorophenyl)-6-(2-fluoro-2-methylpropyl)-5-methyl-4,5,6,7-tetrahydro- lH- pyrazolo[4,3-f]isoquinoline- l-carboxylate (455 mg, 96%) as a pale yellow solid, which existed as a mixture of isomers (-4: 1 ratio) XH NMR (400 MHz, CDC13, 30 °C) Major
Isomer: 1.04 (3H, d), 1.14 (3H, d), 1.24 (3H, d), 1.52 (9H, s), 1.69 (9H, s), 2.30 (1H, dd), 2.81 - 3.07 (2H, m), 3.50 (1H, d), 3.79 (1H, s), 5.33 (1H, s), 6.32 (1H, d), 6.88 - 7.01 (3H, m), 7.42 (1H, d), 7.83 (1H, d), 8.19 (1H, s). m/z: ES+ [M+H]+ 600.
Example 3: (E)-3-(4-((6^,8R)-7-(tf)-3-fluoro-2-methylpropyl)-8-methyl-6,7,8,9- tetrahvdro-3H-pyrazolor4,3-f1iso uinolin-6-yl)-3-methoxyphenyl)acrylic acid
Figure imgf000085_0002
Aqueous lithium hydroxide (2 M; 1.13 mL, 2.26 mmol) was added to a stirred solution of methyl (E)-3-(4-((65,8R)-7-((5)-3-fluoro-2-methylpropyl)-8-methyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylate (102 mg, 0.23 mmol) in a mixture of THF:MeOH (2: 1 ; 1.5 mL). The mixture was stirred for 2.5 hours at room temperature and then concentrated under reduced pressure. Water (2 mL) was added and the mixture was treated with aqueous HC1 (6N) to adjust the pH to ~5. The mixture was extracted with EtOAc (3 x 5 mL) and the combined organic layers were washed with saturated aqueous sodium chloride, dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 20 to 100% EtOAc in hexanes. Fractions containing the desired product were combined and concentrated to dryness to afford
(E)-3-(4-((65,8R)-7-((5)-3-fluoro-2-methylpropyl)-8-methyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylic acid (81 mg, 82%) as a white solid. lH NMR (500 MHz, DMSO-J6, 27 °C) 0.81 (3H, d), 0.97 (3H, d), 2.06 (1H, dt), 2.10 - 2.25 (1H, m), 2.52 - 2.58 (1H, m), 2.78 (1H, dd), 3.04 (1H, dd), 3.26 - 3.30 (1H, m), 3.91 (3H, s), 4.32 - 4.45 (1H, m), 4.45 - 4.55 (1H, m), 5.28 (1H, s), 6.52 (1H, d), 6.63 (1H, d), 6.70 (1H, d), 7.02 (1H, d), 7.21 (1H, d), 7.34 (1H, d), 7.52 (1H, d), 8.06 (1H, s), 12.33 (1H, br s), 12.97 (1H, br s). m/z: ES+ [M+H]+ 438.
The methyl (E)-3-(4-((65,8R)-7-((5)-3-fluoro-2-methylpropyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylate used as starting material was prepared as follows;
Preparation of methyl (E)-3-(4-formyl-3-methoxyphenyl)acrylate
Figure imgf000086_0001
[l, -Bis(di-iert-butylphosphino)ferrocene]dichloropalladium(II) (0.061 g, 0.09 mmol) was added to a degassed solution of 4-bromo-2-methoxybenzaldehyde (1.00 g, 4.67 mmol), triethylamine (0.955 mL, 7.00 mmol) and methyl acrylate (0.631 mL, 7.00 mmol) in dimethylacetamide (13 mL). The stirred reaction was heated at 100 °C for 1 hour and then allowed to cool to ambient temperature and left to stand overnight. The reaction mixture was diluted with EtOAc and washed with water and saturated aqueous sodium chloride. The organic layer was dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 40 to 100% DCM in hexanes. Fractions containing the desired product were combined and concentrated to dryness to afford methyl (E)-3-(4-formyl-3- methoxyphenyl)acrylate (0.84 g, 82%) as a pale yellow solid. XH NMR (300 MHz, CDC13, 27 °C) 3.84 (3H, s), 3.98 (3H, s), 6.53 (1H, d), 7.10 (1H, d), 7.21 (1H, d), 7.69 (1H, d), 7.86 (1H, d), 10.47 (1H, d). m/z: ES+ [M+H]+ 221. Preparation of (S)-3-fluoro-2-methylpropan-l-ol
Figure imgf000087_0001
N,N-Diethyl- 1, 1, 2,3,3, 3-hexafluoropropan-l -amine (9.45 mL, 52.1 mmol) was added dropwise to a solution of methyl (R)-3-hydroxy-2-methylpropanoate (4.81 mL, 43.4 mmol) in DCM (40 mL) and a slight exotherm was noted. The reaction was stirred for 1 hour at room temperature and then stirred at 40 °C for 5 hours before cooling to room temperature overnight. The reaction mixture was poured onto ice and the layers were separated. The aqueous layer was extracted with DCM (2 x 75 mL) and the combined organic layers were dried over sodium sulfate, filtered and carefully concentrated at reduced pressure. The residue obtained was dissolved in THF (100 mL), cooled in an ice bath and treated with LAH (3.30 g, 86.9 mmol) portionwise over 10 min. The reaction was stirred at 0 °C for 1 hour and was then warmed to room temperature and stirred for 1 hour. The reaction mixture was then cooled to 0 °C and quenched sequentially with 3.3 mL of water, 3.3 mL of 15% NaOH and 9.9 mL of water. Magnesium sulfate was added until a granular solid was formed. The solid was filtered through celite and the solids were washed with diethyl ether (50 mL). The filtrate was washed with aqueous HC1 (IN; 2 x 100 mL) and the organic phase was dried over sodium sulfate, filtered and concentrated. The crude product was purified by flash silica chromatography, elution gradient 0 to 30% EtOAc in DCM. Fractions containing desired product were combined and concentrated under reduced pressure to give (S)-3-fluoro-2-methylpropan- l-ol (2.7 g, 66%). The ΧΗ NMR of the final product showed the isolated material to contain 1 mole equivalent of ethyl acetate. ΧΗ
NMR (300 MHz, DMSO-de, 27 °C) 0.86 (3H, dd) 1.72 - 1.95 (1H, m) 3.32 (2H, ddd) 4.17 - 4.32 (1H, m) 4.34 - 4.49 (1H, m) 4.55 (1H, t).
Preparation of (S)-3-fluoro-2-methylpropyl trifluoromethanesulfonate
Figure imgf000087_0002
(S)-3-Fluoro-2-methylpropan- l-ol (600 mg, 3.58 mmol) was dissolved in DCM (8 mL) and cooled to - 10 °C. The solution was then treated with triflic anhydride (0.666 mL, 3.94 mmol) followed by 2,6-lutidine (0.542 mL, 4.66 mmol). The reaction was stirred at - 10 °C for 1.5 hours and then washed with aqueous HC1 (IN) and saturated aqueous sodium bicarbonate. The organic phase was dried over sodium sulfate, filtered and concentrated carefully at reduced pressure to give crude (S)-3-fluoro-2-methylpropyl
trifluoromethanesulfonate as a yellow oil (1.2 g containing EtOAc and DCM, >100% yield). This material was used in the next step without further purification. XH NMR (300 MHz, DMSO- d6, 27 °C) 0.93 (3H, dd) 2.08 - 2.39 (1H, m) 4.15 - 4.38 (3H, m) 4.38 - 4.53 (1H, m)
Preparation of (^)-N-((R)-l-(3-bromo-2-methylphenyl)propan-2-yl)-3-fluoro-2- methylpropan- 1 -amine
Figure imgf000088_0001
(R)-l-(3-Bromo-2-methylphenyl)propan-2-amine (0.65 g, 2.9 mmol) was dissolved in 1,4- dioxane (10 mL) and treated with (S)-3-fluoro-2-methylpropyl trifluoromethanesulfonate (0.805 g, 3.59 mmol) dropwise followed by the addition of Hunig's Base (0.647 mL, 3.70 mmol). The reaction was heated at 85 °C overnight and then diluted with water and extracted with EtOAc (3 x 20 mL). The combined EtOAc extracts were dried over sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 16% MeOH in DCM.
Fractions containing product were concentrated to dryness to give (S)-N-((R)- l-(3-bromo- 2-methylphenyl)propan-2-yl)-3-fluoro-2-methylpropan- l-amine (0.68 g, 79%) as a brown oil. lH NMR (300 MHz, DMSO-J6, 27 °C) 0.77 - 0.97 (6H, m) 1.68 - 1.96 (1H, m) 2.34 (3H, s) 2.37 - 2.45 (1H, m) 2.65 - 2.97 (2H, m) 4.1 1 - 4.30 (1H, m) 4.30 - 4.48 (1H, m) 6.96 - 7.07 (1H, m) 7.12 (1H, d) 7.38 - 7.48 (1H, m) . Amine NH not observed and a 2H multiplet is obscured by DMSO. m/z: ES+ [M+H]+ 302. Preparation of 3-((R)-2-(((^)-3-fluoro-2-methylpropyl)amino)propyl)-2-methylaniline
Figure imgf000088_0002
Bis(dibenzylideneacetone)palladium (0) (0.038 g, 0.07 mmol), racemic-2,2'- bis(diphenylphosphino)- l,l'binaphthyl (0.041 g, 0.07 mmol), sodium ie/t-butoxide (0.319 g, 3.32 mmol) and diphenylmethanimine (0.371 mL, 2.21 mmol) were added to a degassed solution of (S)-N-((R)- l-(3-bromo-2-methylphenyl)propan-2-yl)-3-fluoro-2-methylpropan- 1-amine (0.669 g, 2.21 mmol) in toluene (10 mL). The reaction mixture was then heated at 90 °C for 2.5 hours before being cooled and filtered through celite. The filter cake was washed with DCM and the filtrate was concentrated under reduced pressure. The resulting residue was dissolved in DCM (10 mL) and aqueous HC1 (IN; 10 mL) was added. The biphasic mixture was stirred vigorously for 60 min. DCM (20 mL) and water (20 mL) were added and layers were separated. The aqueous layer was then basified by addition of aqueous NaOH (IN) and extracted with DCM (3 x 50 mL). The combined organic extracts were dried over sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 20% MeOH in DCM. Fractions containing desired product were evaporated to dryness to give 3-((R)-2-(((lS')-3-fluoro-2-methylpropyl)amino)propyl)-2-methylaniline (0.40 g, 75%) as a yellow-orange oil. lH NMR (300 MHz, DMSO-J6, 27 °C) 0.88 (6H, m) 1.63 - 1.93 (1H, m) 1.97 (3H, s) 2.28 - 2.42 (1H, m) 2.64 - 2.81 (2H, m) 4.14 - 4.31(1H, m) 4.31 - 4.47 (1H, m) 4.61 - 4.73 (2H, m) 6.28 - 6.38 (1H, m) 6.43 - 6.52 (1H, m) 6.77 (1H, t). Amine NH not observed and 2H obscured by DMSO peak, m/z: ES+ [M+H]+ 239.
Preparation of methyl (E)-3-(4-((l^,3R)-6-amino-2-(tf)-3-fluoro-2-methylpropyl)-3,5- dimethyl-l^^^-tetrahydroiso uinolin-l-vD-S-methoxyphenvDacrylate
Figure imgf000089_0001
3-((R)-2-(((lS')-3-fluoro-2-methylpropyl)amino)propyl)-2-methylaniline (456 mg, 1.91 mmol) and methyl (E)-3-(4-formyl-3-methoxyphenyl)acrylate (842 mg, 3.82 mmol) were suspended in AcOH (20 mL) and water (0.172 mL, 9.56 mmol). The reaction was stirred at 60 °C for 1 hour and then the temperature was increased to 80 °C. After 3.5 hours, the reaction was concentrated under reduced pressure and the resulting residue was dissolved in EtOAc (60 mL) and washed with saturated aqueous sodium bicarbonate (3 x 25 mL). The combined aqueous layers were extracted with EtOAc (2 x 20 mL) and then the combined organic layers were washed with saturated aqueous sodium chloride and concentrated under reduced pressure. The residue was dissolved in EtOAc (30 mL), treated with aqueous HCl (IN, 20 mL) and the biphasic mixture was stirred vigorously for 1 hour. The layers were separated and the organic layer was extracted with aqueous HCl (IN, 15 mL). The combined acidic aqueous layers were washed with DCM (10 mL) and then basified with solid sodium carbonate and extracted with DCM (3 x 20 mL). The combined organic extracts were dried over sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 5 to 60% EtOAc in hexanes. Fractions containing the desired product were combined and evaporated to dryness to afford methyl (E)-3-(4-((15,,3R)-6-amino-2-((lS,)-3- fluoro-2-methylpropyl)-3,5-dimethyl-l,2,3,4-tetrahydroisoquinolin- l-yl)-3- methoxyphenyl)acrylate (606 mg, 72%) as a pale yellow foam. XH NMR (400 MHz, CDC , 27 °C) 0.90 (3H, d), 1.02 (3H, d), 2.09 (3H, s), 2.12 - 2.18 (1H, m), 2.20 - 2.34 (1H, m), 2.37 - 2.53 (2H, m), 2.61 (1H, dd), 3.17 - 3.27 (1H, m), 3.57 (2H, br s), 3.81 (3H, s), 3.91 (3H, s), 4.33 - 4.68 (2H, m), 5.25 (1H, s), 6.39 (1H, d), 6.48 - 6.57 (2H, m), 6.72 (1H, d), 6.92 (1H, d), 7.02 (1H, s), 7.65 (1H, d). m/z ES+ [M+H]+ 441.
Preparation of methyl (E)-3-(4-((6^,8R)-7-((S)-3-fluoro-2-methylpropyl)-8-methyl-
6,7,8,9-tetrahvdro-3H-pyrazolor4,3-f1iso uinolin-6-yl)-3-methoxyphenyl)acrylate
Figure imgf000090_0001
A solution of sodium nitrite (94 mg, 1.4 mmol) in water (0.60 mL) was added dropwise over 2 minutes to a stirred solution of methyl (E)-3-(4-((lS,3R)-6-amino-2-((S)-3-fluoro-2- methylpropyl)-3,5-dimethyl-l,2,3,4-tetrahydroisoquinolin-l-yl)-3-methoxyphenyl)acrylate (600 mg, 1.36 mmol) in propionic acid (6 mL) at -20 °C. After 40 minutes the reaction was diluted with ice-cold EtOAc (50 mL). The biphasic mixture was stirred vigorously and neutralized to pH 7 by the slow addition of cold saturated aqueous sodium bicarbonate. The phases were separated and the organic layer was washed with saturated aqueous sodium bicarbonate (2 x 25 mL) and saturated aqueous sodium chloride (25 mL). The organic layer was then dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 5 to 50% EtOAc in hexanes. Fractions containing the desired product were combined and evaporated to dryness to afford methyl (E)-3-(4-((65',8R)-7-((lS,)-3-fluoro-2- methylpropyl)-8-methyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3- methoxyphenyl)acrylate (555 mg, 90%) as a pale orange solid. ΧΗ NMR (300 MHz, CDC , 27 °C) 0.90 (3H, d), 1.07 (3H, d), 2.13 - 2.39 (2H, m), 2.50 - 2.68 (1H, m), 2.77 - 2.94 (1H, m), 2.99 - 3.13 (1H, m), 3.39 (1H, ddd), 3.80 (3H, s), 3.94 (3H, s), 4.31 - 4.69 (2H, m), 5.41 (1H, s), 6.41 (1H, d), 6.72 (1H, d), 6.84 (1H, d), 6.90 (1H, d), 7.06 (1H, d), 7.24 (1H, d), 7.66 (1H, d), 8.12 (1H, s), 9.58 - 11.62 (1H, br s). mJ . ES+ [M+H]+ 452.
Example 4: (E)-3-(4-((6^,8R)-7- -fluoro-2-methylpropyl)-6,8-dimethyl-6,7,8,9- tetrahvdro-3H-pyrazolor4,3-f1iso uinolin-6-yl)-3-methoxyphenyl)acrylic acid
Figure imgf000091_0001
Aqueous NaOH (2N; 0.68 mL, 1.35 mmol) was added to a solution of methyl (E)-3-(4- ((65,,8R)-7-(2-fluoro-2-methylpropyl)-6,8-dimethyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3- f]isoquinolin-6-yl)-3-methoxyphenyl)acrylate (90 mg, 0.14 mmol) in THF (1.0 mL) and methanol (1.0 mL). The reaction was stirred at room temperature for 2 hours and then EtOAc and water were added. The pH was adjusted to ~6 by addition of aqueous HCl (2N) and the layers were separated. The aqueous layer was extracted with EtOAc and the organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 15% MeOH in DCM. Product fractions were evaporated to dryness to afford (E)-3-(4-((65,8R)-7-(2-fluoro-2-methylpropyl)-6,8-dimethyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylic acid (31 mg, 51%) as a colourless solid. lH NMR (500 MHz, CDC13, 27 °C) 0.92 (3H, d), 1.09 (3H, d), 1.14 (3H, d), 1.80 (3H, s), 2.37 (IH, dd), 2.88 - 3.05 (2H, m), 3.28 (3H, s), 3.46 (IH, d), 3.94 (IH, d), 6.40 (IH, d), 6.77 (IH, d), 6.84 (IH, s), 7.10 (2H, dd), 7.43 (IH, d), 7.69 (IH, d), 8.11 (IH, s). m/z: ES+ [M+H]+ 452.
The methyl (E)-3-(4-((65,8R)-7-(2-fluoro-2-methylpropyl)-6,8-dimethyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylate used as starting material was prepared as follows;
Preparation of fe -butyl (6^,8R)-7-(2-fluoro-2-methylpropyl)-6-(2-methoxy-4-((E)-3- methoxy-3-oxoprop-l-en-l-yl)phenyl)-8-methyl-6,7,8,9-tetrahvdro-3H-pyrazolor4,3- fl isoq uinoline-3-carbox ylate and fert-butyl (6^,8R)-7-(2-fluoro-2-methylpropyl)-6-(2- methoxy-4-((E)-3-methoxy-3-oxoprop-l-en-l-yl)phenyl)-8-methyl-6,7,8,9-tetrahvdro- -pyrazolor4,3-flisoquinoline-2-carboxylate
Figure imgf000092_0001
Di-ie/t-butyl dicarbonate (0.508 g, 2.33 mmol) was added to a solution of methyl (E)-3-(4- ((65,,8R)-7-(2-fluoro-2-methylpropyl)-8-methyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3- f]isoquinolin-6-yl)-3-methoxyphenyl)acrylate (1.00 g, 2.21 mmol) in DCM (8.9 mL) and the reaction was stirred at room temperature for 1 hour. The reaction was concentrated under reduced pressure and the resulting residue was passed through a plug of silica gel, eluting with 40% EtOAc in heptane. The filtrate was concentrated to dryness to afford two isomers of the N-Boc indazole (1.25 g, >100%, -2: 1 ratio of isomers) as a pale orange solid, which was used without purification. The identity of the major isomer was not established. Major Isomer: lH NMR (500 MHz, CDC13, 27 °C) 1.05 (3H, d), 1.21 (3H, dd), 1.25 (3H, dd), 1.72 (9H, s), 2.22 - 2.39 (IH, m), 2.70 (IH, dd), 2.79 - 2.93 (IH, m), 3.22 (IH, d), 3.73 (IH, dt), 3.80 (3H, s), 3.94 (3H, s), 5.29 (IH, s), 6.40 (IH, dd), 6.66 (IH, d), 6.96 (IH, ddd), 7.01 - 7.09 (2H, m), 7.35 (IH, d), 7.64 (IH, dd), 8.54 (IH, d). Minor Isomer: 1.05 (3H, d), 1.22 (3H, d), 1.24 - 1.26 (3H, m), 1.69 (9H, s), 2.21 - 2.35 (IH, m), 2.89 (IH, dd), 3.36 (IH, dd), 3.76 - 3.79 (2H, m), 3.80 (3H, s), 3.95 (3H, s), 5.44 (IH, s), 6.40 (IH, dd), 6.90 (IH, d), 6.96 (2H, dd), 7.07 (IH, d), 7.64 (IH, dd), 7.79 (IH, d), 8.18 (IH, d). m/z: ES+ [M+H]+ 552.
Preparation of methyl (E)-3-(4-((6^,8R)-7-(2-fluoro-2-methylpropyl)-6,8-dimethyl- -tetrahvdro-3H-pyrazolor4,3-f1iso uinolin-6-yl)-3-methoxyphenyl)acrylate
Figure imgf000093_0001
Ceric (IV) ammonium nitrate (2.24 g, 4.08 mmol) was added to a solution of tert-b tyl (65,,8R)-7-(2-fluoro-2-methylpropyl)-6-(2-methoxy-4-((E)-3-methoxy-3-oxoprop-l-en-l- yl)phenyl)-8-methyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline-3-carboxylate (1.00 g, 1.81 mmol, mixture of 2 isomers, -2: 1 ratio,) in acetonitrile (9.28 mL) and water (2.32 mL) and the reaction was stirred at room temperature for 2 hours. EtOAc and saturated aqueous sodium chloride were added and the layers were separated. The aqueous layer was extracted with EtOAc (x2) and the combined organic layers were dried over magnesium sulfate, filtered and concentrated under reduced pressure. The crude iminium ion was purified by flash silica chromatography, elution gradient 0 to 20% MeOH in EtOAc. Product fractions were evaporated to dryness to afford the iminium ion as an orange solid. The solid was dissolved in DCM:THF (1 : 1, 20 mL) and cooled to -78 °C. Methylmagnesium bromide in diethyl ether (2.5 M; 2.90 mL, 7.25 mmol) was added and the reaction was maintained under these conditions for 30 mins before being quenched by addition of water and saturated aqueous NH4C1. The reaction was extracted with DCM (x3) and the crude mixture was dried over sodium sulfate and concentrated to dryness. The resulting residue was stirred in HC1 in dioxane (4N; 5 mL) at room temperature for 1 hour. The mixture was then passed through an SCX-2 column, eluting with MeOH and then NH3 in MeOH (IN) to liberate the product. The basic filtrate was concentrated to dryness and the crude product was purified by flash silica chromatography, elution gradient 0 to 60% EtOAc in heptane. Product fractions were evaporated to dryness to afford methyl (E)-3-(4-
((65,,8R)-7-(2-fluoro-2-methylpropyl)-6,8-dimethyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3- f]isoquinolin-6-yl)-3-methoxyphenyl)acrylate (0.10 g, 12%) as a beige gum. XH NMR (500 MHz, CDCI3, 27 °C) 1.00 - 1.12 (6H, m), 1.13 (3H, d), 1.80 (3H, s), 2.30 - 2.46 (1H, m), 2.95 - 3.08 (2H, m), 3.27 (3H, s), 3.46 (1H, s), 3.79 (3H, s), 3.92 (1H, dd), 6.37 (1H, d), 6.76 (1H, d), 6.82 (1H, d), 7.06 (1H, dd), 7.10 (1H, d), 7.41 (1H, d), 7.62 (1H, d), 8.12 (1H, s). m/z: ES+ [M+H]+ 466.
Example 5: (E)-3-(4-((6^,8R)-7- -fluoro-3-methoxy-2-methylpropyl)-8-methyl- 6,7,8,9-tetrahvdro-3H-pyrazolor4,3-f1iso uinolin-6-yl)-3-methoxyphenyl)acrylic acid (ISOMER 1)
Figure imgf000094_0001
Aqueous sodium hydroxide (2N; 0.98 mL, 1.95 mmol) was added dropwise to a solution of methyl (E)-3-(4-((65,8R)-7-(2-fluoro-3-methoxy-2-methylpropyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylate (188 mg, 0.39 mmol) in a mixture of THF (2.0 mL) and MeOH (0.98 mL) at room temperature. The reaction was stirred at room temperature for 3 hours. The reaction was then diluted with water (50 mL) and the pH was adjusted was to pH 4.6 by addition of aqueous HC1 (2N). The mixture was then extracted with EtOAc (x2). The combined organic layers were washed with saturated aqueous sodium chloride, dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 8% MeOH in DCM. Product fractions were evaporated to dryness to afford (E)-3-(4-((65,,8R)-7-(2-fluoro-3-methoxy-2-methylpropyl)- 8-methyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylic acid (135 mg, 74%) as a beige solid. lH NMR (500 MHz, DMSO, 27 °C) 0.97 (3H, d), 1.19 (3H, d), 2.32 - 2.42 (1H, m), 2.79 - 2.91 (2H, m), 3.12 - 3.22 (5H, m), 3.46 (1H, dd), 3.49 - 3.58 (1H, m), 3.92 (3H, s), 5.33 (1H, s), 6.52 (1H, d), 6.62 (1H, d), 6.76 (1H, d), 7.06 (1H, dd), 7.18 (1H, d), 7.35 (1H, d), 7.52 (1H, d), 8.05 (1H, d), 12.36 (1H, s), 12.93 (1H, s). m/z: ES+ [M+H]+ 468.
The methyl (E)-3-(4-((65,8R)-7-(2-fluoro-3-methoxy-2-methylpropyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylate used as starting material was prepared as follows;
Preparation of diethyl 2-fluoro-2-methylmalonate
Figure imgf000095_0001
Sodium 2-methylpropan-2-olate (6.41 g, 66.7 mmol) was added portionwise over 5 minutes to a cooled solution (<10 °C) of diethyl 2-fluoro-2-methylmalonate (8.56 g, 48.1 mmol) in THF (150 mL). The mixture was then warmed to room temperature and stirred for 45 minutes. The solution was cooled and iodomethane (4.15 mL, 66.7 mmol) in tetrahydrofuran (15 mL) was added drop wise over 15 minutes with the temperature kept below 13 °C. The reaction was warmed to room temperature and stirred for 18 hours. The reaction was diluted with water (75 mL), extracted with EtOAc (2 x 40 mL) and the combined organic layers were washed with 10% aqueous sodium thiosulphate (50 mL) and saturated aqueous sodium chloride (50 mL) before being dried over sodium sulfate, filtered and concentrated under reduced pressure to afford crude diethyl 2-fluoro-2- methylmalonate (8.56 g, 80%) as an oil which was used without further purification.
NMR (500 MHz, CDCb, 27 °C) 1.31 (6H, t), 1.79 (3H, d), 4.30 (4H, qd).
Preparation of 2-fluoro-2-methylpropane-l,3-diol
Figure imgf000096_0001
Lithium aluminum hydride (4.40 g, 116 mmol) was added portionwise to a solution of diethyl 2-fluoro-2-methylmalonate (8.56 g, 44.5 mmol) in THF (225 mL) at 0 °C over a period of 40 minutes while maintaining an internal temperature of less than 5 °C. The reaction was then warmed to room temperature and stirred for 100 minutes. The reaction mixture was cooled to 0 °C and then quenched by dropwise addition of water (4.4 mL). The mixture was allowed to warm to room temperature and was then stirred under these conditions for 18 hours. Aqueous sodium hydroxide (15 wt%; 4.4 mL) and water (13.2 mL) were then added dropwise sequentially and the gelatinous suspension was stirred rapidly for 1 hour. The mixture was filtered through celite and the solids were washed with THF (2 x 15 mL) and EtOAc (2 x 20 mL). The solvent was evaporated to afford crude 2- fluoro-2-methylpropane- l,3-diol (3.09 g, 64%) as an oil, which was used without further purification. lH NMR (500 MHz, DMSO, 27 °C) 1.18 (3H, d), 3.42 (4H, dd), 4.81 (2H, t).
Preparation of 2-fluoro-2-methyl-3-(trityloxy)propan-l-ol
Figure imgf000096_0002
N,N-Dimethylpyridin-4-amine (0.349 g, 2.86 mmol) was added to a solution of 2-fluoro-2- methylpropane-l,3-diol (3.09 g, 28.6 mmol) and N-ethyl-N-isopropylpropan-2-amine (7.60 mL, 42.9 mmol) in DCM (111 mL) at room temperature under nitrogen.
(Chloromethanetriyl)tribenzene (7.17 g, 25.7 mmol) was then added in one portion. The reaction mixture was stirred for 18 hours at 40 °C and then cooled. The mixture was diluted with DCM (30 mL) and then washed with aqueous citric acid (1 M; 40 mL), saturated aqueous sodium bicarbonate (50 mL) and saturated aqueous sodium chloride (40 mL). The organic layer was dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting pale yellow gum was purified by flash silica
chromatography, eluting with 0-30% EtOAc/heptane to afford 2-fluoro-2-methyl-3- (trityloxy)propan-l-ol (4.62 g, 46%) as a colourless gum which solidified on standing to afford a white solid. lH NMR (500 MHz, DMSO, 27 °C) 1.26 (3H, d), 2.99 - 3.15 (2H, m), 3.48 (2H, dd), 4.91 - 4.96 (1H, m), 7.23 - 7.29 (3H, m), 7.30 - 7.41 (12H, m).
Preparation of ((2-fluoro-3-methoxy-2-methylpropoxy)methanetriyl)tribenzene
Figure imgf000097_0001
Sodium hydride (60 wt% in mineral oil; 0.319 g, 7.98 mmol) was added in one portion to a stirred solution of 2-fluoro-2-methyl-3-(trityloxy)propan-l-ol (2.33 g, 6.65 mmol) in THF (66 mL) at 0 °C under nitrogen while maintaining an internal temperature of less than 5 °C. The resulting mixture was stirred with the temperature kept below 5 °C for 5 minutes and was then allowed to warm to room temperature. After 25 minutes, iodomethane (0.455 mL, 7.31 mmol) was added drop wise over 2 minutes and the mixture was stirred at room temperature for 18 hours. The reaction mixture was quenched with saturated aqueous ammonium chloride (2 mL) and the mixture was partitioned between EtOAc and water. The layers were separated and the aqueous layer was extracted with EtOAc. The combined organic layers were washed with saturated aqueous sodium chloride and then dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 15% EtOAc in heptane. Product fractions were concentrated to dryness to afford ((2-fluoro-3-methoxy-2- methylpropoxy)methanetriyl)tribenzene (2.24 g, 92%) as a yellow gum. XH NMR (500
MHz, DMSO, 27 °C) 1.29 (3H, d), 3.07 (2H, d), 3.25 (3H, s), 3.49 (2H, dd), 7.27 (3H, ddt), 7.33 - 7.41 (12H, m). Preparation of N-((R)-l-(3-bromo-2-methylphenyl)propan-2-yl)-2-fluoro-3-methoxy- 2-methylpropan- 1 -amine
Figure imgf000098_0001
Trifluoromethanesulfonic anhydride (1.09 mL, 6.45 mmol) was added to a solution of ((2- fluoro-3-methoxy-2-methylpropoxy)methanetriyl)tribenzene (2.24 g, 6.15 mmol) in DCM (28.6 mL) and the reaction was stirred under these conditions for 30 minutes.
Triethylsilane (1.08 mL, 6.76 mmol) was added and the reaction was stirred for a further 30 minutes. Volatile components were then evaporated to afford crude 2-fluoro-3- methoxy-2-methylpropyl trifluoromethanesulfonate (1.52 g, 6.00 mmol) as a purple oil. This material was dissolved in 1,4-dioxane (15.6 mL) and (R)- l-(3-bromo-2- methylphenyl)propan-2-amine (1.37 g, 6.00 mmol) and diisopropylethylamine (1.56 mL, 9.00 mmol) were added. The mixture was heated at 85 °C for 16 hours and then cooled. The reaction was concentrated under reduced pressure and the resulting residue was dissolved in EtOAc and washed with saturated aqueous sodium chloride. The aqueous layer was extracted with EtOAc and the combined organic layers were dried over magnesium sulfate, filtered and evaporated. The crude material was purified by flash silica chromatography, elution gradient 0 to 100% EtOAc in heptane. Product fractions were evaporated to dryness to afford N-((R)- l-(3-bromo-2-methylphenyl)propan-2-yl)-2-fluoro- 3-methoxy-2-methylpropan-l -amine (1.53 g, 77%) as a yellow oil. XH NMR (500 MHz, CDC , 27°C) 1.04 (3H, d), 1.31 (3H, dd), 2.41 (3H, s), 2.53 - 2.69 (1H, m), 2.69 - 2.79 (1H, m), 2.78 - 2.93 (3H, m), 3.35 (3H, d), 3.37 - 3.49 (2H, m), 6.96 (1H, t), 7.07 (1H, d), 7.42 (1H, d). m/z: ES+ [M+H]+ 332.
Preparation of 3-((2R)-2-((2-fluoro-3-methoxy-2-methylpropyl)amino)propyl)-2- methylaniline
Figure imgf000098_0002
Pd2(dba)3 (0.089 g, 0.11 mmol) and sodium ie/t-butoxide (0.63 g, 6.6 mmol) were added to a degassed suspension of N-((R)- l-(3-bromo-2-methylphenyl)propan-2-yl)-2-fluoro-3- methoxy-2-methylpropan- 1 -amine (1.45 g, 4.36 mmol), diphenylmethanimine (0.81 mL, 4.8 mmol) and 2,2'-bis(diphenylphosphanyl)-l, l'-binaphthalene (0.136 g, 0.22 mmol) in toluene (16.6 mL). The reaction was heated at 90 °C for 3 hours. After cooling, the reaction mixture was diluted with EtOAc and washed with water. The aqueous layer was extracted with EtOAc and the combined organic layers were dried over magnesium sulfate and concentrated under reduced pressure. The resulting residue was dissolved in EtOAc (80 mL) and aqueous HC1 (2N; 80 mL) was added. The biphasic mixture was stirred vigourously for 18 hours. The layers were separated and the organic layer was extracted with aqueous HC1 (2N). The combined aqueous layers were extracted with EtOAc. The aqueous acid layer was purified by ion exchange chromatography, using an 50 g SCX-2 column. The desired product was eluted from the column using NH3 in MeOH (1 M) and product fractions were evaporated to dryness to afford 3-((2R)-2-((2-fluoro-3-methoxy-2- methylpropyl)amino)propyl)-2-methylaniline (0.96 g, 82%) as a solid. 1H NMR (500 MHz, DMSO, 27°C) 0.90 (3H, dd), 1.22 (3H, d), 1.42 (1H, s), 1.97 (3H, s), 2.32 - 2.40 (1H, m), 2.60 - 2.76 (4H, m), 3.23 - 3.26 (3H, m), 3.32 - 3.40 (2H, m), 4.67 (2H, s), 6.33 (1H, d), 6.47 (1H, d), 6.77 (1H, t). m/z: ES+ [M+H]+ 269.
Preparation of methyl (E)-3-(4-((l^,3R)-6-amino-2- -fluoro-3-methoxy-2- methylpropy -S^-dimethyl-l^^^-tetrahvdroisoquinolin-l-vD-S- methoxyphenvDacrylate
Figure imgf000099_0001
Methyl (E)-3-(4-formyl-3-methoxyphenyl)acrylate (1.64 g, 7.43 mmol) was added to a solution of 3-((2R)-2-((2-fluoro-3-methoxy-2-methylpropyl)amino)propyl)-2-methylaniline (0.95 g, 3.54 mmol) in acetic acid (15.4 mL). Water (0.32 mL, 18 mmol) was added and the reaction mixture was heated at 80 °C for 18 hours. After cooling, the volatiles were evaporated under vacuum and the resulting residue was dissolved in EtOAc (100 mL) and washed with saturated aqueous sodium bicarbonate (x2). Aqueous HC1 (2N; 100 mL) was added and the biphasic mixture was stirred vigorously for 30 minutes. The layers were separated and the organic layer was extracted with aqueous HC1 (2N). The combined aqueous layers were then extracted with EtOAc (x2). The aqueous acid layer was then purified by ion exchange chromatography, using an 50 g SCX-2 column that had been pre- treated with first methanol and then water. After introduction of the crude aqueous acid layer, the SCX column was washed with water and MeOH prior to elution of the desired product with NH3 in MeOH (1M). Product fractions were concentrated to dryness to afford crude product (1.2 g). Both water and methanol eluents from SCX purification were re-purified by SCX column as just described to afford additional crude material (0.6 g). The initial batch of crude product (1.2 g) was purified by flash silica chromatography, elution gradient 0 to 40% EtOAc in heptane. Fractions containing the 2nd eluting, more polar product were evaporated to dryness to afford ISOMER 2 as a yellow solid (0.4 g). Fractions containing the 1st eluting, less polar product, ISOMER 1, were combined with fractions containing both ISOMER 1 and ISOMER 2 and concentrated under reduced pressure. The resulting residue was then combined with the second batch of crude material (0.6 g) and purified by flash silica chromatography, elution gradient 0 to 30% EtOAc in heptane. Fractions were concentrated to dryness to afford 1st eluting, less polar product, methyl (E)-3-(4-((llS,,3R)-6-amino-2-(2-fluoro-3-methoxy-2-methylpropyl)-3,5-dimethyl- l,2,3,4-tetrahydroisoquinolin- l-yl)-3-methoxyphenyl)acrylate [ISOMER 1] (0.562 g, 34%) as a yellow solid and a 2nd eluting, more polar product (which was combined with the 0.4 g just described) to afford methyl (E)-3-(4-((llS',3R)-6-amino-2-(2-fluoro-3-methoxy-2- methylpropyl)-3,5-dimethyl-l,2,3,4-tetrahydroisoquinolin-l-yl)-3-methoxyphenyl)acrylate [ISOMER 2] (0.448 g, 27%) as a yellow solid.
[ISOMER 1] : lH NMR (500 MHz, DMSO, 27 °C) 0.91 (3H, d), 1.20 (3H, d), 1.94 (3H, s), 2.25 - 2.41 (2H, m), 2.66 - 2.79 (2H, m), 3.21 (4H, s), 3.31 - 3.38 (1H, m), 3.43 (1H, dd), 3.71 (3H, d), 3.88 (3H, s), 4.60 (2H, s), 5.14 (1H, s), 6.23 (1H, d), 6.35 (1H, d), 6.62 (1H, d), 6.71 (1H, d), 7.09 (1H, dd), 7.33 (1H, d), 7.59 (1H, d). m/z: ES+ [M+H]+ 471. [ISOMER 2] : XH NMR (500 MHz, DMSO, 27 °C) 0.91 (3H, d), 1.17 (3H, d), 1.94 (3H, s), 2.27 - 2.42 (2H, m), 2.71 (2H, dd), 3.22 (3H, s), 3.31 - 3.39 (2H, m), 3.43 - 3.51 (IH, m), 3.71 (3H, s), 3.89 (3H, s), 4.60 (2H, s), 5.13 (IH, s), 6.25 (IH, d), 6.36 (IH, d), 6.62 (IH, d), 6.72 (IH, d), 7.08 (IH, dd), 7.34 (IH, d), 7.59 (IH, d). m/z ES+ [M+H]+ 471.
Preparation of methyl (E)-3-(4-((6^,8R)-7-(2-fluoro-3-methoxy-2-methylpropyl)-8- methyl-6,7,8,9-tetrahvdro-3H-pyrazolor4,3-f1isoguinolin-6-yl)-3- methoxyphenvDacrylate (ISOMER 1)
Figure imgf000101_0001
Methyl (E)-3-(4-((15,3R)-6-amino-2-(2-fluoro-3-methoxy-2-methylpropyl)-3,5-dimethyl- l,2,3,4-tetrahydroisoquinolin- l-yl)-3-methoxyphenyl)acrylate [ISOMER 1] (446 mg, 0.95 mmol) in propionic acid (3.9 mL) and water (0.39 mL) was cooled to -17 °C using a dry ice/acetone bath. After stirring for, 15 minutes, a solution of sodium nitrite (65 mg, 0.95 mmol) in water (0.39 mL) was added dropwise over 5 minutes. The reaction mixture was stirred at -17 °C for a further 30 minutes and then diluted with ice-cold EtOAc (24 mL). The reaction mixture was stirred vigorously and neutralised by dropwise addition of ice cold saturated aqueous sodium bicarbonate (31 mL). After stirring for 1 hour, the layers were separated and the organic layer was washed with saturated aqueous sodium bicarbonate (2 x 50 mL) and saturated aqueous sodium chloride (50 mL). The combined aqueous layers were extracted with EtOAc (2 x 50 mL) and the combined organic layers were dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting brown oil was purified by silica column chromatography eluting with 5 to 50% EtOAc in heptane to afford methyl (E)-3-(4-((65,8R)-7-(2-fluoro-3-methoxy-2- methylpropyl)-8-methyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3- methoxyphenyl)acrylate (188 mg, 41%) as an orange-coloured solid. XH NMR (500 MHz, DMSO, 27 °C) 0.97 (3H, d), 1.19 (3H, d), 2.31 - 2.42 (1H, m), 2.79 - 2.91 (2H, m), 3.20 (5H, s), 3.45 (1H, dd), 3.49 - 3.56 (1H, m), 3.71 (3H, s), 3.92 (3H, s), 5.33 (1H, s), 6.66 (2H, s), 6.77 (1H, d), 7.10 (1H, dd), 7.18 (1H, d), 7.39 (1H, d), 7.60 (1H, d), 8.03 - 8.07 (1H, m), 12.95 (1H, s). m/z: ES+ [M+H]+ 482.
Example 6: (E)-3-(4-((6^,8R)-7- -fluoro-3-methoxy-2-methylpropyl)-8-methyl- 6,7,8,9-tetrahvdro-3H-pyrazolor4,3-f1iso uinolin-6-yl)-3-methoxyphenyl)acrylic acid (ISOMER 2)
Figure imgf000102_0001
Aqueous sodium hydroxide (2N; 1.89 mL, 3.76 mmol) was added dropwise to a solution of methyl (E)-3-(4-((65,8R)-7-(2-fluoro-3-methoxy-2-methylpropyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylate [ISOMER 2] (362 mg, 0.75 mmol) in a mixture of THF (3.8 mL) and MeOH (1.9 mL) at room temperature. The reaction was stirred at room temperature for 75 minutes and then diluted with water (50 mL). The pH was adjusted to pH 4.9 by addition of aqueous HCl (2N). The mixture was then extracted with EtOAc (x2). The combined organic layers were washed with saturated aqueous sodium chloride, dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 8% MeOH in DCM. Product fractions were evaporated to dryness to afford (E)-3-(4-((65,,8R)-7-(2-fluoro-3-methoxy-2-methylpropyl)- 8-methyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylic acid (263 mg, 75%) as a white solid. lH NMR (500 MHz, DMSO, 27 °C) 0.97 (3H, d), 1.17 (3H, d), 2.32 - 2.42 (1H, m), 2.78 - 2.89 (2H, m), 3.14 - 3.21 (1H, m), 3.22 (3H, s), 3.31 - 3.38 (1H, m), 3.43 (1H, dd), 3.48 - 3.55 (1H, m), 3.93 (3H, s), 5.33 (1H, s), 6.52 (1H, d), 6.64 (1H, d), 6.77 (1H, d), 7.04 (1H, dd), 7.18 (1H, d), 7.35 (1H, d), 7.52 (1H, d), 8.05 (1H, d), 12.34 (1H, s), 12.93 (1H, s). m/z: ES+ [M+H]+ 468.
The methyl (E)-3-(4-((65,8R)-7-(2-fluoro-3-methoxy-2-methylpropyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylate [ISOMER 2] used as starting material was prepared as follows;
Preparation of methyl (E)-3-(4-((6^,8R)-7-(2-fluoro-3-methoxy-2-methylpropyl)-8- methyl-6,7,8,9-tetrahvdro-3H-pyrazolor4,3-f1isoquinolin-6-yl)-3- methoxyphenvDacrylate (ISOMER 2)
Figure imgf000103_0001
Methyl (E)-3-(4-((llS',3R)-6-amino-2-(2-fluoro-3-methoxy-2-methylpropyl)-3,5-dimethyl- l,2,3,4-tetrahydroisoquinolin- l-yl)-3-methoxyphenyl)acrylate [ISOMER 2] (560 mg, 1.19 mmol) in propionic acid (4.9 mL) and water (0.49 mL) was cooled to -17 °C using a dry ice/acetone bath. After 15 minutes, a solution of sodium nitrite (82 mg, 1.2 mmol) in water (0.49 mL) was added dropwise over 5 minutes. The reaction mixture was stirred at -17 °C for a further 30 minutes and then diluted with ice-cold EtOAc (30 mL). The reaction mixture was stirred vigorously and neutralised by dropwise addition of ice cold saturated aqueous sodium bicarbonate (39 mL) and stirred for 1 hour. The layers were separated and the organic layer was washed with saturated aqueous sodium bicarbonate (2 x 50 mL) and saturated aqueous sodium chloride (50 mL). The combined aqueous layers were extracted with EtOAc (2 x 50 mL) and the combined organic layers were dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting brown oil was purified by flash silica chromatography, elution gradient 5 to 50% EtOAc in heptane to afford methyl (E)-3-(4-((65,8R)-7-(2-fluoro-3-methoxy-2-methylpropyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-methoxyphenyl)acrylate (364 mg, 64%) as a beige coloured solid. XH NMR (500 MHz, DMSO, 27 °C) 0.97 (3H, d), 1.16 (3H, d), 2.32 - 2.43 (IH, m), 2.79 - 2.88 (2H, m), 3.14 - 3.20 (IH, m), 3.22 (3H, s), 3.31 - 3.55 (3H, m), 3.71 (3H, s), 3.93 (3H, s), 5.33 (IH, s), 6.61 - 6.67 (2H, m), 6.77 (IH, d), 7.08 (IH, dd), 7.18 (IH, d), 7.40 (IH, d), 7.60 (IH, d), 8.05 (IH, s), 12.95 (IH, s). m/z ES+ [M+H]+ 482.
Example 7: (E)-3-(3-fluoro-4-((6^,8R)-7-(tf)-3-fluoro-2-methylpropyl)-8-methyl-
6,7,8,9-tetrahvdro-3H-pyrazolor4,3-f1iso uinolin-6-yl)-5-methoxyphenyl)acrylic acid
Figure imgf000104_0001
Methyl (E)-3-(3-fluoro-4-((8R)-7-((5)-3-fluoro-2-methylpropyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-5-methoxyphenyl)acrylate (260 mg, 0.55 mmol) was dissolved in MeOH (1.5 mL), water (4.5 mL) and THF (4.5 mL). Lithium hydroxide (265 mg, 11.1 mmol) was then added. The reaction was stirred at room temperature for 1.5 hours and then the solvent was removed under reduced pressure. The resulting residue was dissolved in water and the pH was adjusted to 6 using aqueous HC1 (6N). The resulting suspension was extracted with EtOAc and the organic layer was washed with saturated aqueous sodium chloride, dried over sodium sulfate and filtered. The filtrate was concentrated under reduced pressure and the resulting yellow residue was purified by flash silica chromatography, elution gradient 0 to 20% MeOH in DCM.
Product fractions were combined and concentrated under reduced pressure. The resulting residue was resolved by SFC (Chiralpak AD column, 5 μιη silica, 21.2 mm diameter, 250 mm length, 75 mL/min, 100 bar outlet pressure), eluting with isocratic 15% (methanol containing 0.2% NH4OH) in carbon dioxide. Product fractions were concentrated under reduced pressure to afford the 2nd eluting isomer, (E)-3-(3-fluoro-4-((65',8R)-7-((lS,)-3- fluoro-2-methylpropyl)-8-methyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)- 5-methoxyphenyl)acrylic acid (140 mg, 56%), as an off white solid H NMR (300 MHz, DMSO-Je, 27 °C) 0.70 (3H, d) 0.97 (3H, d) 1.83 (1H, br d) 2.12 (1H, br dd) 2.54 - 2.67 (1H, m) 2.90 (1H, br dd) 3.13 - 3.25 (1H, m) 3.42 (1H, m) 3.88 (3H, br s) 4.12 - 4.49 (2H, m) 5.32 (1H, br s) 6.49 - 6.72 (2H, m) 6.94 (1H, br d) 7.18 (2H, d) 7.40 (1H, d) 8.05 (1H, s) 12.76 - 13.03 (1H, m). Amine NH not observed, m/z: ES+ [M+H]+ 456.
The methyl (E)-3-(3-fluoro-4-((8R)-7-((5)-3-fluoro-2-methylpropyl)-8-methyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-5-methoxyphenyl)acrylate used as starting material was prepared as follows;
Preparation of methyl (E)-3-(4-((3R)-6-amino-2-((^)-3-fluoro-2-methylpropyl)-3,5- dimethyl-l^^^-tetrahydroiso uinolin-l-vD-S-fluoro-S-methoxyphenvDacrylate
Figure imgf000105_0001
3-((R)-2-(((lS')-3-Fluoro-2-methylpropyl)amino)propyl)-2-methylaniline (395 mg, 1.66 mmol) and methyl (E)-3-(3-fluoro-4-formyl-5-methoxyphenyl)acrylate (790 mg, 3.31 mmol) were suspended in acetic acid (7 mL) and water (0.14 mL). The reaction was heated at 60 °C under nitrogen for 6 hours and then concentrated under reduced pressure. The resulting residue was dissolved in EtOAc (30 mL) and neutralized with saturated aqueous sodium bicarbonate (15 mL). The aqueous layer was extracted with EtOAc (3 x 15 mL) and the combined organic layers were dried over sodium sulfate, filtered and concentrated under reduced pressure to an orange residue. This residue was stored in the freezer for 18 hours and then dissolved in EtOAc (20 mL), treated with aqueous HC1 (IN; 12 mL) and the biphasic mixture was stirred vigorously for 20 minutes. The layers were separated and the organic layer was extracted with aqueous HC1 (IN; 2 x 7 mL). The combined aqueous layers were extracted with DCM (10 mL). The aqueous layer was then basified with sodium carbonate and extracted with DCM (3 x 15 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 50% EtOAc in hexanes. Product fractions were combined and concentrated at reduced pressure to give an inseparable mixture of diastereoisomers of methyl (E)-3-(4-((3R)-6-amino-2- ((5,)-3-fluoro-2-methylpropyl)-3,5-dimethyl-l,2,3,4-tetrahydroisoquinolin- l-yl)-3-fluoro-5- methoxyphenyl)acrylate (0.58 g, 77%) as a yellow foam, m/z: ES+ [M+H]+ 459.
Preparation of methyl (E)-3-(3-fluoro-4-((8R)-7-((^)-3-fluoro-2-methylpropyl)-8- methyl-6,7,8,9-tetrahvdro-3H-pyrazolor4,3-f1isoquinolin-6-yl)-5- methoxyphenvDacrylate
Figure imgf000106_0001
A solution of sodium nitrite (88 mg, 1.3 mmol) in water (0.50 mL) was added dropwise over 1 minute to a stirred solution of methyl (E)-3-(4-((3R)-6-amino-2-((5,)-3-fluoro-2- methylpropyl)-3,5-dimethyl-l,2,3,4-tetrahydroisoquinolin-l-yl)-3-fluoro-5- methoxyphenyl)acrylate (582 mg, 1.27 mmol) in propionic acid (5 mL) at -20 °C. After 20 minutes the reaction was diluted with ice-cold EtOAc (25 mL). The biphasic mixture was stirred vigorously and neutralised by slow addition of cold saturated aqueous sodium bicarbonate (50 mL) resulting in gas evolution. The phases were separated and the organic layers were washed with saturated aqueous sodium bicarbonate (25 mL) and saturated aqueous sodium chloride (20 mL). The organic layer was dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting brown residue was purified by flash silica chromatography, elution gradient 0 to 70% EtOAc in to give an inseparable mixture of diastereoisomers of methyl (E)-3-(3-fluoro-4-((8R)-7-((lS')-3-fluoro-2- methylpropyl)-8-methyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-5- methoxyphenyl)acrylate (0.26 g, 44%) as a yellow-orange film, m/z: ES+ [M+H]+ 470. Example 8: (E)-3-(4-((6^,8R)-7- ,2-difluoropropyl)-6,8-dimethyl-6,7,8,9-tetrahvdro- 3H-pyrazolor4,3-f1iso uinolin-6-yl)-3-fluoro-5-methoxyphenyl)acrylic acid
Figure imgf000107_0001
HCl in dioxane (4N; 0.24 mL, 0.98 mmol) was added to a solution of tert-butyl (E)-3-(4- ((65,8R)-7-(2,2-difluoropropyl)-6,8-dimethyl-3-(tetrahydro-2H-pyran-2-yl)-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-fluoro-5-methoxyphenyl)acrylate (30 mg, 0.05 mmol) in DCM (0.5 mL). The solution was stirred for 3 hours and then concentrated under reduced pressure. The resulting solid was treated with HCl in dioxane (4N; 0.24 mL, 0.98 mmol) and the mixture was stirred for another 1.5 hours before being concentrated under reduced pressure. The resulting solid was partitioned between EtOAc and water and the pH was adjusted to ~pH 5 with saturated aqueous sodium bicarbonate. The phases were separated and the aqueous phase was extracted with EtOAc (2 x 3 mL). The combined organic layers were washed with saturated aqueous sodium chloride, dried over magnesium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by flash silica chromatography, elution gradient 1 to 10% MeOH in DCM. Fractions containing the desired product were evaporated to dryness. The product was then purified by Chiral SFC (Chiralpak AD column, 5 μιη silica, 21.2 mm diameter, 250 mm length, 40 °C column temperature; Outlet pressure 100 bar) eluting with isocratic 15% MeOH (buffered with 0.2% NH4OH) in C02 at 75 mL/min. Product fractions were combined and concentrated under reduced pressure to afford the (E)-3-(4-((65',8R)-7-(2,2- difluoropropyl)-6,8-dimethyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3- fluoro-5-methoxyphenyl)acrylic acid (9.0 mg, 39%) as a white solid. XH NMR (500 MHz, CD2C12, 27 °C) 1.10 - 1.18 (3H, m), 1.19 (3H, d), 1.92 (3H, d), 2.84 - 2.98 (IH, m), 3.06 (IH, d), 3.25 (IH, ddd), 3.44 - 3.52 (4H, m), 3.74 - 3.86 (IH, m), 6.44 (IH, d), 6.81 (IH, s), 6.86 (IH, br d), 6.93 (IH, d), 7.21 (IH, d), 7.64 (IH, d), 8.12 (IH, s). Indazole NH and acid OH protons not observed, m/z: ES+ [M+H]+ 474. Chiral analysis using analytical SFC (Chiralpak AD column, 5 μιη silica, 4.6 mm diameter, 100 mm length, 40 °C column temperature) eluting with isocratic 15% MeOH (buffered with 0.2% NH4OH) in C02 at 2.8 mL/min showed the product was isolated as a 96 : 4 ratio of trans : cis isomers.
The tert-butyl (E)-3-(4-((65,8R)-7-(2,2-difluoropropyl)-6,8-dimethyl-3-(tetrahydro-2H- pyran-2-yl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3-fluoro-5- methoxyphenyl)acrylate used as starting material was prepared as follows;
Preparation of 4-bromo-2-fluoro-6-methoxybenzaldehyde
Figure imgf000108_0001
Sodium methoxide in methanol (25 wt%; 7.9 mL, 34 mmol) was added to a stirred solution of 4-bromo-2,6-difluorobenzaldehyde (5.0 g, 23 mmol) in MeOH (35 mL). The reaction was heated under reflux conditions for 25 hours and then concentrated under reduced pressure. DCM and aqueous HC1 (IN) were added to the resulting solid and the mixture was stirred until the solid dissolved. The phases were separated and the organic layer was washed with saturated aqueous sodium chloride, dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 3 to 25% EtOAc in hexanes. Fractions containing the desired product were combined and concentrated under reduced pressure to afford 4- bromo-2-fluoro-6-methoxybenzaldehyde (4.8 g, 91%) as a white solid. XH NMR (300
MHz, CDCls, 27 °C) 3.95 (3H, s), 6.92 - 7.02 (2H, m), 10.37 (1H, d). m/z: ES+ [M+H]+
233. Preparation of (l^,3R)-l-(4-bromo-2-fluoro-6-methoxyphenyl)-2-(2,2- difluoropropyD-S^-dimethyl-l^^^-tetrahvdroisoquinolin^-amine
Figure imgf000109_0001
4-Bromo-2-fluoro-6-methoxybenzaldehyde (1.35 g, 5.78 mmol) was added to a stirred solution of (R)-3-(2-((2,2-difluoropropyl)amino)propyl)-2-methylaniline (700 mg, 2.89 mmol) in acetic acid (14 mL). Water (0.26 mL, 14 mmol) was added and the solution was heated sequentially at 60 °C for 1.5 hours, 75 °C for 1.5 hours, 85 °C for 75 minutes and then at 95 °C for 10 minutes. The reaction was cooled, concentrated under reduced pressure and the resulting brown residue was stored in a freezer overnight under nitrogen. The residue was then allowed to warm to ambient temperature, dissolved in EtOAc (50 mL) and washed with saturated aqueous sodium bicarbonate (3 x 30 mL). The combined aqueous layers were extracted with DCM (3 x 30 mL) and the combined organic layers were concentrated under reduced pressure. The resulting residue was dissolved in DCM (25 mL), treated with aqueous HC1 (IN; 25 mL) and the biphasic mixture was stirred vigorously for 1 hour. The phases were separated and the aqueous phase was washed with DCM (2 x 20 mL). The aqueous phase was then basified by addition of solid sodium carbonate and extracted with EtOAc (3 x 20 mL). The combined organic extracts were dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 5 to 50% EtOAc in hexanes. Fractions containing the desired product were combined and concentrated to dryness to afford (llS,,3R)-l-(4-bromo-2-fluoro-6-methoxyphenyl)-2-(2,2- difluoropropyl)-3,5-dimethyl-l,2,3,4-tetrahydroisoquinolin-6-amine (243 mg, 18%) as a pale orange solid. lH NMR (300 MHz, CDC13, 27 °C) 1.02 (3H, d), 1.30 - 1.47 (3H, m), 1.53 (2H, br s), 2.17 (3H, s), 2.38 - 2.53 (1H, m), 2.60 (1H, dd), 2.88 - 3.13 (2H, m), 3.58 (1H, br s), 3.82 (3H, br s), 5.31 (1H, br s), 6.44 (1H, d), 6.64 (1H, d), 6.76 (1H, d), 6.82 - 6.89 (1H, m). m/z: ES+ [M+H]+ 457. Preparation of (6^,8R)-6-(4-bromo-2-fluoro-6-methoxyphenyl)-7-(2,2- difluoropropyl)-8-methyl-6,7,8,9-tetrahvdro-3H-pyrazolor4,3-f1isoguinoline
Figure imgf000110_0001
A solution of sodium nitrite (36 mg, 0.52 mmol) in water (0.30 mL) was added dropwise over 1 minute to a stirred solution of (lS,3R)-l-(4-bromo-2-fluoro-6-methoxyphenyl)-2- (2,2-difluoropropyl)-3,5-dimethyl-l,2,3,4-tetrahydroisoquinolin-6-amine (0.24 mg, 0.52 mmol) in propionic acid (3 mL) at -20 °C. After 20 minutes the reaction was diluted with ice-cold EtOAc (15 mL). The biphasic mixture was stirred vigorously and neutralised by slow addition of cold saturated aqueous sodium bicarbonate. The phases were separated and the organic layers were washed with saturated aqueous sodium bicarbonate (15 mL) and saturated aqueous sodium chloride (10 mL). The organic layer was dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 5 to 50% EtOAc in hexanes, Fractions containing the desired product were combined and concentrated to dryness to afford (65,,8R)-6-(4-bromo-2-fluoro-6-methoxyphenyl)-7-(2,2-difluoropropyl)-8-methyl- 6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline (150 mg, 61%) as an orange solid. ΧΗ NMR (300 MHz, CDC13, 27 °C) 1.07 (3H, d), 1.38 (3H, t), 2.48 - 2.66 (1H, m), 2.93 (1H, dd), 3.06 (1H, dt), 3.46 (1H, dd), 3.59 - 3.73 (1H, m), 3.81 (3H, s), 5.43 (1H, br s), 6.64 - 6.81 (2H, m), 6.87 (1H, br s), 7.15 (1H, d), 8.08 (1H, s), 8.78 - 11.13 (1H, m). m/z: ES+ [M+] 468. Preparation of (6^,8R)-6-(4-bromo-2-fluoro-6-methoxyphenyl)-7-(2,2- difluoropropyl)-8-methyl-3-(tetrahvdro-2H-pyran-2-yl)-6,7,8,9-tetrahvdro-3H-
Pyrazolor4,3-f1isoguinoline
Figure imgf000111_0001
3,4-Dihydro-2H-pyran (0.044 mL, 0.48 mmol) and tosic acid monohydrate (6 mg, 0.03 mmol) were added to a solution of (6S,8R)-6-(4-bromo-2-fluoro-6-methoxyphenyl)-7-(2,2- difluoropropyl)-8-methyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline (150 mg, 0.32 mmol) in DCM (1.5 mL). The reaction was stirred at ambient temperature for 30 minutes and then heated under reflux conditions for 3.5 hours. The reaction was allowed to cool to ambient temperature and diluted further with DCM. The solution was washed with saturated aqueous sodium bicarbonate, dried over magnesium sulfate, filtered and concentrated under reduced pressure to afford (65,,8R)-6-(4-bromo-2-fluoro-6- methoxyphenyl)-7-(2,2-difluoropropyl)-8-methyl-3-(tetrahydro-2H-pyran-2-yl)-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinoline (172 mg, 97%) as a reddish-orange solid. The material was used without further purification. XH NMR (300 MHz, CDC13, 27 °C) 1.05 (3H, dd), 1.36 (3H, t), 1.53 - 1.79 (4H, m), 1.96 - 2.13 (1H, m), 2.19 - 2.26 (1H, m), 2.46 - 2.65 (1H, m), 2.74 (1H, dd), 3.05 (1H, dt), 3.37 (1H, d), 3.65 (1H, td), 3.73 - 3.92 (4H, m), 4.10 - 4.22 (1H, m), 5.35 (1H, br s), 5.61 - 5.71 (1H, m), 6.60 (1H, d), 6.77 (1H, d), 6.86 (1H, s), 7.36 (1H, d), 8.11 (1H, s). m/z: ES+ [M+H]+ 552. Preparation of (6^,8R)-6-(4-bromo-2-fluoro-6-methoxyphenyl)-7-(2,2- difluoropropyl)-6,8-dimethyl-3-(tetrahvdro-2H-pyran-2-yl)-6,7,8,9-tetrahvdro-3H-
Pyrazolor4,3-f1isoguinoline
Figure imgf000112_0001
Ceric (IV) ammonium nitrate (354 mg, 0.65 mmol) was added to a solution of (65,8Λ)-6- (4-bromo-2-fluoro-6-methoxyphenyl)-7-(2,2-difluoropropyl)-8-methyl-3-(tetrahydro-2H- pyran-2-yl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline (170 mg, 0.31 mmol) in a mixture of acetonitrile: water (4: 1, 2.5 mL). The reaction was stirred at ambient temperature for 20 minutes and then diluted with EtOAc. The mixture was washed with saturated aqueous sodium chloride and the aqueous phase was extracted with EtOAc (3 x 3 mL). The combined organic layers were filtered through a PTFE membrane, concentrated under reduced pressure and the resulting residue was dried briefly in vacuo. The crude product was dissolved in THF (2.5 mL) and cooled to -78 °C. Methylmagnesium bromide in diethyl ether (3M; 0.31 mL, 0.92 mmol) was added rapidly dropwise and the reaction was stirred for 30 minutes. Additional methylmagnesium bromide in diethyl ether (3M; 0.31 mL, 0.92 mmol) was added. The cooling bath removed and the reaction was allowed to warm to ambient temperature. The reaction was stirred for 2 hours and then cooled to 0 °C. Saturated aqueous ammonium chloride and water were added and the mixture was diluted with EtOAc before being filtered through diatomaceous earth. The phases were separated and the aqueous phase was extracted with DCM (x2). The combined organic layers were dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 45% EtOAc in hexanes. Fractions containing the desired product were combined and evaporated to dryness to afford (65,,8R)-6-(4-bromo-2-fluoro-6- methoxyphenyl)-7-(2,2-difluoropropyl)-6,8-dimethyl-3-(tetrahydro-2H-pyran-2-yl)- 6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline (30 mg, 17%) as a pale yellow film. ΧΗ NMR (300 MHz, CDCb, 27 °C) 1.08 - 1.18 (6H, m), 1.65 (IH, br s), 1.73 - 1.79 (2H, m), 1.84 (3H, dd), 2.01 - 2.12 (IH, m), 2.12 - 2.21 (IH, m), 2.53 - 2.63 (IH, m), 2.75 - 2.88 (IH, m), 2.98 (IH, d), 3.15 - 3.24 (IH, m), 3.32 - 3.42 (4H, m), 3.70 - 3.79 (2H, m), 4.01 - 4.08 (IH, m), 5.65 (IH, td), 6.69 (IH, d), 6.79 (IH, dt), 6.86 (IH, dd), 7.23 (IH, dd), 8.05 (IH, d). mJ . ES+ [M+H]+ 566.
Preparation of fe -butyl (E)-3-(4-((6^,8R)-7-(2,2-difluoropropyl)-6,8-dimethyl-3-
(tetrahvdro-2H-pyran-2-yl)-6,7,8,9-tetrahvdro-3H-pyrazolor4,3-f1isoguinolin-6-yl)-3- fluoro-5-methoxyphenyl)acrylate
Figure imgf000113_0001
A vial was charged with a stir bar, (65,,8R)-6-(4-bromo-2-fluoro-6-methoxyphenyl)-7-(2,2- difluoropropyl)-6,8-dimethyl-3-(tetrahydro-2H-pyran-2-yl)-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinoline (30 mg, 0.05 mmol), tert-butyl acrylate (0.012 mL, 0.08 mmol), [l, -bis(di-iert-butylphosphino)ferrocene]dichloropalladium(II) (1.7 mg, 2.7 μιηοΐ) and triethylamine (0.014 mL, 0.11 mmol) under argon. The vial was sealed and dioxane (0.5 mL) was added via syringe. The mixture was stirred at ambient temperature for 2 minutes and then heated under microwave conditions at 100 °C for 30 minutes. The reaction was heated in the microwave for a further 30 minutes at 125 °C before the reaction was cooled and dimethylacetamide (0.1 mL) plus further portions of dichloro [Ι,Γ- bis(di- tert- butylphosphino)ferrocene]palladium(II) (1.7 mg, 2.6 μιηοΐ) and tert-butyl acrylate (0.012 mL, 0.08 mmol) were added. The mixture was sparged with argon for 5 minutes and again heated under microwave conditions at 125 °C for 30 minutes. The cooled mixture was diluted with EtOAc and washed with saturated aqueous sodium chloride (x2). The organic layer was dried over magnesium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by flash silica chromatography, elution gradient 0 to 30% EtOAc in hexanes. Fractions containing the desired product were combined and concentrated to dryness to afford tert-butyl (E)-3-(4-((65',8R)-7-(2,2- difluoropropyl)-6,8-dimethyl-3-(tetrahydro-2H-pyran-2-yl)-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3-fluoro-5-methoxyphenyl)acrylate (30 mg, 92%) as a pale yellow film. lH NMR (300 MHz, CDC13, 27 °C) 1.00 - 1.09 (3H, m), 1.14 (3H, d), 1.43 - 1.49 (3H, m), 1.51 - 1.54 (9H, m), 1.74 - 1.77 (1H, m), 1.86 (3H, dd), 2.05 - 2.22 (2H, m), 2.49 - 2.66 (1H, m), 2.74 - 2.93 (1H, m), 2.99 (1H, d), 3.12 - 3.27 (1H, m), 3.40 (3H, d), 3.70 - 3.80 (2H, m), 4.04 (1H, br d), 5.65 (1H, ddd), 6.28 (1H, d), 6.67 (1H, d), 6.75 (1H, d), 6.88 (1H, dd), 7.23 (1H, dd), 7.42 (1H, d), 8.05 (1H, d). m/z: ES+ [M+H]+ 614.
Example 9: (E)-3-(3-fluoro-4-(7-(tf)-3-fluoro-2-methylpropyl)-8,8-dimethyl-6J,8,9- tetrahvdro-3H-pyrazolor4,3-f1iso uinolin-6-yl)-5-methoxyphenyl)acrylic acid
Figure imgf000114_0001
(E)-tert-Butyl 3-(3-fluoro-4-(7-((lS')-3-fluoro-2-methylpropyl)-8,8-dimethyl-3-(tetrahydro- 2H-pyran-2-yl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-5- methoxyphenyl)acrylate (1.24 g, 2.03 mmol) was stirred in dichloromethane (10 mL) and trifluoroacetic acid (5 mL) was added. The mixture was stirred at ambient temperature for 2 hours. Reverse phase C18 silica gel (3 g) was added and the solvent was removed in vacuo. The resulting powder was purified by flash reverse phase silica chromatography, elution gradient 10 to 70% MeCN in water containing 0.1% formic acid as modifier. Product fractions were evaporated to dryness to afford the 2nd eluting isomer (E)-3-(3- fluoro-4-(7-((5,)-3-fluoro-2-methylpropyl)-8,8-dimethyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-5-methoxyphenyl)acrylic acid (0.156 g, 16%) as a white solid lH NMR (500 MHz, DMSO, 100 °C) 0.55 (3H, dd), 0.97 (3H, s), 1.30 (1H, m), 1.34 (3H, s), 2.24 (IH, s), 2.92 (IH, d), 2.99 (IH, m), 3.20 (IH, d), 3.87 (3H, s), 4.14 (IH, td), 4.23 (IH, td), 5.39 (IH, s), 6.51 (IH, d), 6.62 (IH, d), 6.93 (IH, d), 7.14 (2H, d), 7.50 (IH, d), 8.00 (IH, d), 8.11 (IH, s). m/z: ES+ [M+H]+ 470. Chiral Analysis: The product was analysed by analytical HPLC (Chiral Technologies AY column, 5 μιη silica, 4.6 mm diameter, 250 mm length), using a 80/20 mixture of heptane/EtOH as eluent at a flowrate of 2 mL/min. Example 9 contains about 1.0% of ISOMER 2.
Also obtained from the flash reverse phase silica chromatography was the 1st eluting isomer (E)-3-(3-fluoro-4-(7-((lS')-3-fluoro-2-methylpropyl)-8,8-dimethyl-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-5-methoxyphenyl)acrylic acid (ISOMER 2) (0.193 g, 20%) as a white solid. lH NMR (500 MHz, DMSO, 100 °C) 0.74 (3H, dd), 0.96 (3H, s), 1.36 (3H, s), 1.43 (IH, d), 2.30 - 2.37 (IH, m), 2.81 (IH, ddd), 2.91 (IH, d), 3.18 (IH, d), 3.67 (IH, ddd), 3.86 (3H, s), 4.11 (IH, dd), 5.34 (IH, s), 6.51 (IH, d), 6.61 (IH, d), 6.92 (IH, dd), 7.13 (2H, d), 7.50 (IH, d), 7.99 (IH, d), 8.12 (IH, s). m/z: ES+ [M+H]+ 470.
The (E)-tert-bvXy\ 3-(3-fluoro-4-(7-((5')-3-fluoro-2-methylpropyl)-8,8-dimethyl-3- (tetrahydro-2H-pyran-2-yl)-6,7,8,9-tetrahydro-JH-pyrazolo[4,3-f]isoquinolin-6-yl)-5- methoxyphenyl)acrylate used as starting material was prepared as follows;
Preparation of 4-bromo-l-(tetrahvdro-2H-pyran-2-yl)-lH-indazole
Figure imgf000115_0001
3,4-Dihydro-2H-pyran (5.36 mL, 58.57 mmol) was added to a solution of 4-bromo- lH- indazole (5.77 g, 29.3 mmol) and 4-methylbenzenesulfonic acid (0.176 g, 1.02 mmol) in ethyl acetate (60 mL) and the mixture was heated at 70 °C for 16 hours. The mixture was cooled, added to saturated aqueous sodium bicarbonate (50 mL) and the phases were separated. The aqueous phase was extracted with EtOAc (10 mL) and the combined organic layers were washed with saturated aqueous sodium bicarbonate (10 mL) and saturated aqueous sodium chloride (10 mL) before being dried over magnesium sulfate, filtered and concentrated in vacuo. The resulting brown oil was purified by flash silica chromatography, elution gradient 10% EtOAc in heptane. Product fractions were evaporated to dryness to afford 4-bromo-l-(tetrahydro-2H-pyran-2-yl)-lH-indazole (7.78 g, 94%) as a white solid. lH NMR (400 MHz, CDC13, 30 °C) 1.60 - 1.88 (3H, m), 2.03 - 2.23 (2H, m), 2.55 (1H, dddd), 3.73 (1H, ddd), 4.00 (1H, ddd), 5.71 (1H, dd), 7.19 - 7.24 (1H, m), 7.32 (1H, dd), 7.55 (1H, d), 8.03 (1H, d).
Preparation of 2-methyl-l-(l-(tetrahvdro-2H-pyran-2-yl)-lH-indazol-4-yl)propan-2- ol
Figure imgf000116_0001
n-Butyllithium (1.6 M; 21.7 mL, 34.6 mmol) was added dropwise over 5 minutes to an oven-dried flask containing a solution of 4-bromo-l-(tetrahydro-2H-pyran-2-yl)-lH- indazole (9.28 g, 33.0 mmol) in THF (100 mL) at -78 °C. After stirring for 30 minutes, 2,2-dimethyloxirane (5.86 mL, 66.00 mmol) was added, followed by boron trifluoride diethyl etherate (4.07 mL, 33.0 mmol) dropwise. The reaction was stirred for a further 1 hour and then quenched by addition of saturated aqueous ammonium chloride. After warming, the solution was extracted with EtOAc (x2) and the combined organic layers were dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 50% EtOAc in heptane. Product fractions were evaporated to dryness to afford 2-methyl-l-(l- (tetrahydro-2H-pyran-2-yl)-lH-indazol-4-yl)propan-2-ol (6.81 g, 75%) as a light yellow gum. XH NMR (400 MHz, CDCI3, 27 °C) 1.27 (6H, d), 1.62 - 1.85 (3H, m), 2.04 - 2.24 (2H, m), 2.59 (1H, dddd), 3.08 (2H, s), 3.75 (1H, td), 4.01 - 4.09 (1H, m), 5.71 (1H, d), 6.99 (1H, d), 7.34 (1H, dd), 7.48 (1H, d), 8.05 - 8.12 (1H, m). m/z: ES+ [M+H]+ 275.
Preparation of N-(l-(lH-indazol-4-yl)-2-methylpropan-2-yl)-2-chloroacetamide
Figure imgf000116_0002
Concentrated sulfuric acid (15.4 mL) was added to a solution of 2-methyl-l-(l-(tetrahydro- 2H-pyran-2-yl)-lH-indazol-4-yl)propan-2-ol (9.00 g, 32.8 mmol) and 2-chloroacetonitrile (5.19 mL, 82.0 mmol) in acetic acid (61.5 mL) at 0 °C. The reaction was warmed to room temperature over 24 hours. The reaction mixture was poured onto ice-water (300 mL) and extracted with DCM (x3). The combined organic layers were dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by flash silica chromatography, elution gradient 25 to 75% EtOAc in heptane. Product fractions were evaporated to dryness to afford N-(l-(lH-indazol-4-yl)-2- methylpropan-2-yl)-2-chloroacetamide (4.55 g, 52%) as a pale brown solid. XH NMR (400 MHz, CDCls, 27 °C) 1.45 (6H, s), 3.40 (2H, s), 3.94 (2H, s), 6.32 (IH, s), 6.94 (IH, d), 7.31 - 7.38 (IH, m), 7.41 (IH, d), 8.15 (IH, s). m/z: ES+ [M+H]+ 266.
Preparation of l-(2H-indazol-4-yl)-2-methylpropan-2-amine
Figure imgf000117_0001
Thiourea (1.27 g, 16.7 mmol) was added to N-(l-(2H-indazol-4-yl)-2-methylpropan-2-yl)- 2-chloroacetamide (3.70 g, 13.9 mmol) in EtOH (50 mL) and acetic acid (10.0 mL) at room temperature. The resulting solution was maintained under reflux conditions with stirring for 16 hours. The reaction mixture was allowed to cool and was then concentrated to dryness. The resulting residue was partitioned between ethyl acetate and saturated aqueous sodium hydrogen carbonate. The organic layer was concentrated under reduced pressure. The resulting residue, together with the aqueous layer, was passed through SCX resin, washing with first methanol and then ammonia in methanol (7N) to elute the product. Product containing fractions were concentrated under reduced pressure and the resulting residue was purified by flash silica chromatography, elution gradient 5 to 10% MeOH (containing 1% aqueous NH3) in DCM. Product fractions were evaporated to dryness to afford l-(2H-indazol-4-yl)-2-methylpropan-2-amine (2.30 g, 87%). XH NMR (400 MHz, DMSO, 27 °C) 1.02 (6H, s), 2.91 (2H, s), 6.88 (IH, d), 7.25 (IH, dd), 7.36 (IH, d), 8.15 (IH, d), 12.92 (IH, s). m/z: ES+ [M+H]+ 190. Preparation of 6-(4-bromo-2-fluoro-6-methoxyphenyl)-8,8-dimethyl-6,7,8,9- tetrahvdro-2H-pyrazolor4,3-flisoquinoline
Figure imgf000118_0001
4-Bromo-2-fluoro-6-methoxybenzaldehyde (0.945 g, 4.06 mmol), l-(2H-indazol-4-yl)-2- methylpropan-2-amine (0.867 g, 4.58 mmol) and trifluoroacetic acid (8 mL) were heated in the microwave at 130 °C for 12 hours. The reaction was allowed to cool to room temperature then concentrated in vacuo. The resulting oil was dissolved in methanol (5 mL) and purified by ion exchange chromatography using an SCX column. The desired product was eluted from the column using N¾ in MeOH (1M) in MeOH and product fractions were concentrated to dryness to afford crude product which was further purified by flash silica chromatography, elution gradient 20 to 50% (3: 1 EtOAc:EtOH) in heptane. Product fractions were evaporated to dryness to afford 6-(4-bromo-2-fluoro-6- methoxyphenyl)-8,8-dimethyl-6,7,8,9-tetrahydro-2H-pyrazolo[4,3-f]isoquinoline (1.45 g, 88%) as a white solid. lH NMR (500 MHz, DMSO, 87 °C) 1.28 (3H, s), 1.47 (3H, s), 3.13 (2H, s), 3.69 (3H, s), 5.80 (1H, s), 6.64 (1H, d), 7.05 - 7.13 (2H, m), 7.26 (1H, d), 8.03 (1H, d). m/z: ES+ [M+H]+ 404, 406.
Preparation of 6-(4-bromo-2-fluoro-6-methoxyphenyl)-8,8-dimethyl-3-(tetrahvdro-
2H-pyran-2-yl)-6,7,8,9-tetrahvdro-3H-pyrazolor4,3-flisoquinoline
Figure imgf000118_0002
4-Methylbenzenesulfonic acid hydrate (0.819 g, 4.30 mmol) was added to 3,4-dihydro-2H- pyran (0.656 mL, 7.17 mmol) and 6-(4-bromo-2-fluoro-6-methoxyphenyl)-8,8-dimethyl- 6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline (1.45 g, 3.59 mmol) in DCM (40 niL) and the mixture was heated at 40 °C with stirring for 18 hours. Further portions of 3,4- dihydro-2H-pyran (0.066 mL, 0.72 mmol) and 4-methylbenzenesulfonic acid hydrate (0.082 g, 0.43 mmol) were added and the solution was stirred at 40 °C for a further 6 hours. The reaction mixture was quenched with saturated aqueous sodium bicarbonate (10 mL), extracted with DCM (2 x 20 mL) and the combined organic layers were dried over magnesium sulfate, filtered and concentrated under reduced pressure. The resulting residue was dissolved in DCM (3 mL) and then purified by flash silica chromatography, elution gradient 0 to 50% (3: 1 EtOAc:EtOH) in heptane. Product fractions were evaporated to dryness to afford 6-(4-bromo-2-fluoro-6-methoxyphenyl)-8,8-dimethyl-3-(tetrahydro-2H- pyran-2-yl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline (1.61 g, 92%) as a white solid. XH NMR (500 MHz, DMSO, 100 °C) 1.13 (3H, d), 1.29 (3H, s), 1.44 - 1.50 (IH, m), 1.59 - 1.63 (2H, m), 1.73 - 1.80 (IH, m), 1.94 - 2.00 (IH, m), 2.04 - 2.12 (IH, m), 2.42 (IH, dddd), 2.86 (IH, d), 2.95 - 3.02 (IH, m), 3.69 (IH, ddd), 3.75 (3H, d), 3.83 - 3.92 (IH, m), 5.63 (IH, s), 5.73 (IH, dd), 6.67 (IH, d), 7.01 (IH, d), 7.09 (IH, s), 7.32 (IH, d), 8.03 (IH, d). m/z: ES+ [M+H]+ 488, 490.
Preparation of 6-(4-bromo-2-fluoro-6-methoxyphenyl)-7-((^)-3-fluoro-2- methylpropyl)-8,8-dimethyl-3-(tetrahvdro-2H-pyran-2-yl)-6,7,8,9-tetrahvdro-3H- pyrazolor4,3-f1iso uinoline
Figure imgf000119_0001
(S)-3-Fluoro-2-methylpropyl trifluoromethanesulfonate (1.47 g, 6.55 mmol) was added to a solution of 6-(4-bromo-2-fluoro-6-methoxyphenyl)-8,8-dimethyl-3-(tetrahydro-2H- pyran-2-yl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline (1.60 g, 3.28 mmol) and diisopropylethylamine (1.98 mL, 11.5 mmol) in 1,4-dioxane (15 mL). The reaction was stirred at room temperature for 20 hours. Silica gel (5 g) was added and the reaction mixture was concentrated in vacuo. The resulting powder was purified by flash silica chromatography, elution gradient 0 to 25% (3: 1 EtOAc:EtOH) in heptane. Product fractions were evaporated to dryness to afford 6-(4-bromo-2-fluoro-6-methoxyphenyl)-7- ((S)-3-fluoro-2-methylpropyl)-8,8-dimethyl-3-(tetrahydro-2H-pyran-2-yl)-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinoline (1.80 g, 98%) as a brown gum. lH NMR (500 MHz, DMSO, 100 °C) 0.56 (IH, d), 0.75 (IH, d), 0.94 (3H, s), 1.13 (2H, s), 1.34 (3H, d), 1.59 (2H, d), 1.74 (IH, s), 1.90 - 2.00 (IH, m), 2.01 - 2.11 (IH, m), 2.18 (IH, s), 3.14 (IH, s), 3.30 - 3.49 (2H, m), 3.59 - 3.72 (IH, m), 3.97 - 4.28 (2H, m), 5.32 (IH, s), 5.70 (IH, dt), 6.61 - 6.68 (IH, m), 6.88 (IH, d), 7.06 (IH, s), 7.29 (IH, d), 8.02 (IH, s). m/z: ES+ [M+H]+ 562, 564.
Preparation of (E ert-butyl 3-(3-fluoro-4-(7-((^)-3-fluoro-2-methylpropyl)-8,8- dimethyl-3-(tetrahvdro-2H-pyran-2-yl)-6,7,8,9-tetrahvdro-3H-pyrazolor4,3- isoquinolin-6-yl)-5-methoxyphenyl)acrylate
Figure imgf000120_0001
l,l'-Bis(di-ieri-butylphosphino)ferrocene palladium dichloride (0.209 g, 0.32 mmol) was added to 6-(4-bromo-2-fluoro-6-methoxyphenyl)-7-((S)-3-fluoro-2-methylpropyl)-8,8- dimethyl-3-(tetrahydro-2H-pyran-2-yl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline (1.80 g, 3.20 mmol), ie/t-butyl acrylate (0.71 mL, 4.8 mmol), tetrabutylammonium chloride (0.089 g, 0.32 mmol) and N-methyldicyclohexylamine (1.03 mL, 4.80 mmol) in degassed acetonitrile (30 mL) under nitrogen. The resulting solution was heated under reflux conditions with stirring for 3 hours. Further portions of iert-butyl acrylate (0.71 mL, 4.8 mmol) and l,l'-bis(di-tert-butylphosphino)ferrocene palladium dichloride (0.209 g, 0.32 mmol) were added and the reaction was stirred under reflux conditions for a further 1 hour. The reaction mixture was concentrated to dryness; the resulting residue was dissolved in EtOAc (150 mL) and aqueous citric acid (0.5 M; 100 mL). The mixture was filtered through a pad of celite and the organic layer was washed with water (100 mL). The organic layer was dried over magnesium sulfate, filtered and adsorbed onto silica gel (5 g). The resulting powder was purified by flash silica chromatography, elution gradient 10 to 25% (3: 1 EtOAc:EtOH) in heptane. Product fractions were concentrated under reduced pressure and the isolated material was found to contain a significant amount of unreacted starting bromide and so l ,l'-bis(di-tert-butylphosphino)ferrocene palladium dichloride (0.209 g, 0.32 mmol) was added together with ie/t-butyl acrylate (0.71 mL, 4.8 mmol), N- methyldicyclohexylamine (1.03 mL, 4.80 mmol) and tetrabutylammonium chloride (0.089 g, 0.32 mmol) in degassed acetonitrile (30 mL) under nitrogen. The resulting reaction was heated under reflux conditions for 30 minutes. The reaction mixture was then diluted with EtOAc (150 mL) and water (50 mL) and the mixture was filtered through a pad of diatomaceous earth. The organic layer was dried over magnesium sulfate, filtered and adsorbed onto silica gel (3 g). The resulting powder was purified by flash silica chromatography, elution gradient 10 to 25% (3: 1 EtOAc:EtOH) in heptane. Product fractions were concentrated to dryness to afford (E)-iert-butyl 3-(3-fluoro-4-(7-((5,)-3- fluoro-2-methylpropyl)-8,8-dimethyl-3-(tetrahydro-2H-pyran-2-yl)-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-5-methoxyphenyl)acrylate (1.24 g, 64%) as a brown gum. m/z: ES+ [M+H]+ 610.
Example 10: (E)-3-(4-((6^,8^)-8-(difluoromethyl)-7-(2-fluoro-2-methylpropyl)-6,7,8,9- tetrahvdro-3H-pyrazolor4,3-f1iso uinolin-6-yl)-3,5-difluorophenyl)acrylic acid
Figure imgf000121_0001
HC1 in dioxane (4N; 1 mL) was added to a solution of tert-butyl (E)-3-(4-((65,,85,)-8- (difluoromethyl)-7-(2-fluoro-2-methylpropyl)-3-(tetrahydro-2H-pyran-2-yl)-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3,5-difluorophenyl)acrylate (120 mg, 0.19 mmol) in DCM (0.5 mL) and the reaction was stirred at room temperature for 2 hours. A white emulsion formed during this period. Water (0.25 mL) was added to make the reaction homogeneous and the reaction was stirred for a further 1 hour. The reaction was diluted with EtOAc (10 mL) and water (10 mL) and then the pH of the mixture was adjusted to ~5 by addition of saturated aqueous NaHC03. The layers were separated and the aqueous layer was extracted with EtOAc. The combined organic layers were dried over sodium sulfate and concentrated to dryness. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 100% EtOAc in heptane. Product fractions were concentrated to dryness to afford (E)-3-(4-((65,,85,)-8-(difluoromethyl)-7-(2-fluoro-2- methylpropyl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3,5- difluorophenyl)acrylic acid (81 mg, 87%) as a beige solid. XH NMR (500 MHz, CDC13, 27 °C) 1.15 (3H, d), 1.26 (3H, d), 2.53 (1H, dd), 3.29 (1H, dd), 3.38 - 3.48 (1H, m), 3.48 - 3.60 (1H, m), 3.90 - 4.00 (1H, m), 5.66 (1H, s), 5.87 (1H, td), 6.42 (1H, d), 6.83 (1H, d), 7.01 (2H, d), 7.25 (1H, d), 7.60 (1H, d), 8.12 (1H, d). m/z: ES+ [M+H]+ 480.
The tert-butyl (E)-3-(4-((65,85)-8-(difluoromethyl)-7-(2-fluoro-2-methylpropyl)-3- (tetrahydro-2H-pyran-2-yl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3,5- difluorophenyl)acrylate used as starting material was prepared as follows;
Preparation of 3-bromo-2-methylbenzaldehvde
Figure imgf000122_0001
w-BuLi in hexanes (1.6M; 28.5 mL, 45.6 mmol) was added dropwise to a solution of 1,3- dibromo-2-methylbenzene (9.50 g, 38.0 mmol) in THF (119 mL) at -78 °C and the reaction was maintained at this temperature for 30 minutes. N,N-Dimethylformamide (4.41 mL, 57.0 mmol) was added and the reaction was stirred for a further 30 minutes before being allowed to warm to 0 °C over 1 hour. The reaction was quenched by addition of water and then was extracted with EtOAc. The organic phase was dried over magnesium sulfate, filtered and concentrated to dryness to afford 3-bromo-2-methylbenzaldehyde (7.19 g, 95%) as a light yellow oil. lH NMR (500 MHz, CDC13, 27 °C) 2.75 (3H, s), 7.18 - 7.37 (1H, m), 7.78 (2H, ddd), 10.26 (1H, s). Preparation of (3-bromo-2-methylphenyl)methanol
Figure imgf000123_0001
Sodium borohydride (1.78 g, 47.1 mmol) was added to a solution of 3-bromo-2- methylbenzaldehyde (7.50 g, 37.7 mmol) in THF (151 mL) and the reaction was stirred at room temperature for 2 hours. The reaction was cooled in an ice-bath and quenched by addition of aqueous HC1 (2N), then extracted with EtOAc (x2). The combined organic layers were washed with saturated aqueous sodium chloride, dried over magnesium sulfate, filtered and concentrated to dryness to afford (3-bromo-2-methylphenyl)methanol (7.73 g,
>100%) as a pale yellow solid. XH NMR (500 MHz, CDC13, 27 °C) 1.99 (1H, br s), 2.41 (3H, s), 4.70 (2H, s), 7.05 (1H, t), 7.30 (1H, d), 7.49 - 7.53 (1H, m).
Preparation of l-bromo-3-(bromomethyl)-2-methylbenzene
Figure imgf000123_0002
Carbon tetrabromide (14.4 g, 43.3 mmol) was added portion-wise to a solution of (3- bromo-2-methylphenyl)methanol (7.25 g, 36.1 mmol) and triphenylphosphine (11.35 g, 43.27 mmol) in DCM (120 mL), resulting in an increase in internal reaction temperature to 40 °C, and the reaction was stirred at room temperature for 2 hours. The residue was passed through a pad of silica, eluting with DCM. The filtrate was concentrated to dryness and the crude product was purified by flash silica chromatography, elution gradient 0 to 25% EtOAc in heptane. Product fractions were concentrated to dryness to afford 1-bromo- 3-(bromomethyl)-2-methylbenzene (8.52 g, 90%) as a colourless oil. XH NMR (500 MHz,
CDC , 27 °C) 2.48 (3H, s), 4.52 (2H, s), 6.99 - 7.05 (1H, m), 7.22 - 7.27 (1H, m), 7.52
(1H, dd). Preparation of fert-butyl (S)-3-(3-bromo-2-methylphenyl)-2-((diphenylmethylene) amino)propanoate
Figure imgf000124_0001
l-Bromo-3-(bromomethyl)-2-methylbenzene (6.86 g, 26.0 mmol) was added to a solution of iert-butyl 2-((diphenylmethylene)amino)acetate (7.68 g, 26 mmol) and (llS,,2lS,,45,,5R)-2- ((R)-(allyloxy)(quinolin-4-yl)methyl)- l-(anthracen-9-ylmethyl)-5-vinylquinuclidin- l-ium bromide (1.58 g, 2.60 mmol) in toluene (130 mL) and aqueous KOH solution (50 wt%; 15.08 g, 130.0 mmol). The biphasic mixture was stirred vigorously at 0 °C for 4 hours. The reaction was diluted by addition of water (100 mL) and then extracted with EtOAc (x2). The combined organic layers were washed with saturated aqueous sodium chloride, dried over magnesium sulfate, filtered and concentrated to dryness. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 50% EtOAc in heptane. Product fractions were concentrated to dryness to afford tert-butyl (5,)-3-(3-bromo-2- methylphenyl)-2-((diphenylmethylene)amino)propanoate (9.96 g, 80%) as a pale yellow liquid. XH NMR (500 MHz, CDC13, 27 °C) 1.46 (9H, s), 1.99 (3H, s), 3.17 (1H, dd), 3.34 (1H, dd), 4.12 (1H, dd), 6.48 (2H, s), 6.82 - 6.89 (1H, m), 7.01 (1H, dd), 7.25 - 7.41 (7H, m), 7.53 - 7.59 (2H, m). m/z: ES+ [M+H]+ 478. Chiral analysis using analytical HPLC (Regis (R,R)Whelk-01 column, 5 μιη silica, 4.6 mm diameter, 250 mm length), using a 95/05 mixture of Heptane/EtOH as eluents at 2 niL/min showed the product existed in a 98.3 : 1.7 ratio of (1st eluting : 2nd eluting) isomers.
Preparation of fert-butyl (S)-2-amino-3-(3-bromo-2-methylphenyl)propanoate
Figure imgf000124_0002
Tert-butyl (lS,)-3-(3-bromo-2-methylphenyl)-2-((diphenylmethylene)amino)propanoate (9.0 g, 19 mmol) was stirred in EtOAc (63 mL) and aqueous HC1 (2N; 31.5 mL) at room temperature for 1 hour. The aqueous layer was basified by addition of aqueous NaOH (2N), then extracted with EtOAc (x2). The combined organic layers were dried over magnesium sulfate, filtered and concentrated to dryness. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 100% EtOAc in heptane. Product fractions were concentrated to dryness to afford iert-butyl (S)-2-amino-3-(3-bromo-2- methylphenyl)propanoate (4.98 g, 84%) as a pale yellow oil. XH NMR (500 MHz, CDC13,
27 °C) 1.40 (9H, s), 2.44 (3H, s), 2.80 (1H, dd), 3.12 (1H, dd), 3.55 (1H, dd), 6.94 - 7.00 (1H, m), 7.07 - 7.12 (1H, m), 7.45 (1H, dd). m/z: ES+ [M+H]+ 314.
Figure imgf000125_0001
Preparation of (S)-2-amino-3-(3-bromo-2-methylphenyl)propan-l-ol
Figure imgf000125_0002
Lithium borohydride in THF (2 M; 14.38 mL, 28.75 mmol) was added to a solution of tert- butyl (S)-2-amino-3-(3-bromo-2-methylphenyl)propanoate (7.23 g, 23.0 mmol) in THF (78 mL). The reaction was heated to 50 °C and maintained under these conditions for 1 hour. After cooling in an ice-bath, the reaction was quenched by addition of aqueous HC1 (IN). The aqueous layer was then basified by addition of aqueous NaOH (2N) and extracted with EtOAc (x3). The combined organic layers were dried over magnesium sulfate, filtered and concentrated to dryness to afford (S)-2-amino-3-(3-bromo-2-methylphenyl)propan-l-ol (5.93 g, >100%) as a straw coloured oil. lH NMR (500 MHz, CDC , 27 °C) 1.91 - 2.04 (2H, m), 2.41 (3H, s), 2.60 (1H, dd), 2.88 (1H, dd), 3.01 - 3.14 (1H, m), 3.40 (1H, dd), 3.63 (1H, dd), 6.94 - 7.01 (1H, m), 7.08 (1H, dd), 7.13 (1H, dd), 7.45 (1H, dd). m/z: ES+
[M+H]+ 244. Preparation of (S)-3-(3-bromo-2-methylphenyl)-2-((2-fluoro-2- methylprop yl)amino)propan- 1 -ol
Figure imgf000126_0001
2-Fluoro-2-methylpropyl trifluoromethanesulfonate (6.08 g, 27.1 mmol) was added to a solution of (lS')-2-amino-3-(3-bromo-2-methylphenyl)propan-l-ol (5.30 g, 21.7 mmol) and DIPEA (5.63 mL, 32.6 mmol) in 1,4-dioxane (56.4 mL). The reaction was heated to 90 °C and maintained under these conditions for 18 hours. The reaction was concentrated to dryness and the resulting residue was dissolved in EtOAc and washed with water. The aqueous layer was extracted with EtOAc and the combined organic layers were dried over sodium sulfate, filtered and concentrated to dryness. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 100% EtOAc in heptane. Product fractions were concentrated to dryness to afford (S)-3-(3-bromo-2-methylphenyl)-2-((2- fluoro-2-methylpropyl)amino)propan-l-ol (5.65 g, 82%) as a light brown oil. XH NMR
(500 MHz, CDC , 27 °C) 1.34 (3H, d), 1.38 (3H, d), 2.42 (3H, s), 2.61 (1H, dd), 2.74 - 2.91 (4H, m), 3.30 (1H, dd), 3.54 - 3.59 (1H, m), 6.95 - 7.01 (1H, m), 7.08 (1H, dd), 7.44 (1H, dd). m/z: ES+ [M+H]+ 318.
Preparation of (S)-3-(3-amino-2-methylphenyl)-2-((2-fluoro-2- methylprop yl)amino)propan- 1 -ol
Figure imgf000126_0002
Sodium ie/t-butoxide (2.64 g, 27.4 mmol) and Pd2(dba)3 (0.373 g, 0.46 mmol) were added to a degassed solution of (S)-3-(3-bromo-2-methylphenyl)-2-((2-fluoro-2- methylpropyl)amino)propan-l-ol (5.82 g, 18.3 mmol), diphenylmethanimine (3.22 mL, 19.2 mmol) and rac-BINAP (0.569 g, 0.91 mmol) in toluene (70 mL). The reaction was heated to 90 °C and maintained under these conditions for 2 hours. After cooling, EtOAc and saturated aqueous NH4C1 were added and the layers were separated. The aqueous layer was extracted with EtOAc and the combined organic layers were concentrated to dryness. The resulting residue was dissolved in EtOAc (50 mL) and aqueous HCl (2N; 50 mL) was added. The biphasic mixture was stirred vigorously for 1 hour and the layers were separated. The aqueous layer was extracted with EtOAc and the combined organic layers were extracted with aqueous HCl (2N). The combined aqueous layers were basified by addition of solid K2CO3 and, then extracted with DCM (x3). The combined DCM extracts were dried over sodium sulfate, filtered and concentrated to dryness to afford (S)-3-(3- amino-2-methylphenyl)-2-((2-fluoro-2-methylpropyl)amino)propan-l-ol (4.27 g, 92%) as a light brown oil, which was used directly in the next step without further purification. XH NMR (500 MHz, CDCI3, 27 °C) 1.33 (3H, d), 1.37 (3H, d), 2.12 (3H, s), 2.58 (1H, dd), 2.66 - 2.76 (1H, m), 2.78 - 2.81 (1H, m), 2.82 (1H, d), 2.84 - 2.89 (1H, m), 3.31 (1H, dd), 3.58 (1H, dd), 3.60 (2H, s), 6.59 (2H, d), 6.90 - 7.01 (1H, m). m/z: ES+ [M+H]+ 255.
Preparation of ((l^,3^)-6-amino-l-(4-bromo-2,6-difluorophenyl)-2-(2-fluoro-2- methylpropyD-S-methyl-l^^^-tetrahvdroisoquinolin-S-vDmethanol
Figure imgf000127_0001
(5,)-3-(3-Amino-2-methylphenyl)-2-((2-fluoro-2-methylpropyl)amino)propan- l-ol (4.20 g, 16.5 mmol) and 4-bromo-2,6-difluorobenzaldehyde (7.85 g, 35.5 mmol) were heated in a mixture of acetic acid (72 mL) and water (1.5 mL) at 65 °C and the reaction was maintained under these conditions for 18 hours. After cooling, the solvent was evaporated and the resulting residue was dissolved in DCM (60 mL) and washed with saturated aqueous NaHC03. To the organic phase was then added aqueous HCl solution (2N; 60 mL) and the biphasic mixture was stirred vigorously for 30 min. The layers were separated and the organic layer was extracted with aqueous HCl (2N; x2). The aqueous layer was basified by addition of aqueous NaOH (2N), then extracted with DCM (x2). The combined organic layers were dried over magnesium sulfate, filtered and concentrated to dryness. The resulting residue was dissolved in THF (20 mL) and MeOH (10 mL) and then aqueous NaOH (2N; 10 mL) was added. The reaction was stirred at room temperature for 1 hour and then extracted with EtOAc (x2). The combined organic layers were dried over magnesium sulfate, filtered and concentrated to dryness. The crude product was purified by flash silica chromatography, elution gradient 0 to 100% EtOAc in heptane. Product fractions were concentrated to dryness to afford ((llS,,35')-6-amino-l-(4-bromo-2,6- difluorophenyl)-2-(2-fluoro-2-methylpropyl)-5-methyl-l,2,3,4-tetrahydroisoquinolin-3- yl)methanol (4.15 g, 55%) as a pale yellow solid. lH NMR (500 MHz, CDC13, 27 °C) 1.32 (3H, d), 1.37 (3H, d), 2.07 (3H, s), 2.46 (1H, dd), 2.57 (1H, dd), 2.65 (1H, dd), 2.80 (1H, dd), 3.38 - 3.52 (1H, m), 3.56 (1H, dd), 3.66 (1H, dd), 5.33 (1H, s), 6.50 (1H, d), 6.58 (1H, d), 6.94 - 7.02 (2H, m). m/z: ES+ [M+H]+ 457.
Preparation of ((6^,8^)-6-(4-bromo-2,6-difluorophenyl)-7-(2-fluoro-2-methylpropyl)-
6,7,8,9-tetrahvdro-3H-pyrazolor4,3-f1isoquinolin-8-yl)methanol
Figure imgf000128_0001
Sodium nitrite (145 mg, 2.10 mmol) was added to a cooled solution of ((l^^^-e-amino-l- (4-bromo-2,6-difluorophenyl)-2-(2-fluoro-2-methylpropyl)-5-methyl- 1,2,3,4- tetrahydroisoquinolin-3-yl)methanol (915 mg, 2.00 mmol) in propionic acid (5 mL) and water (1 mL) at -10 °C and the reaction was maintained under these conditions for 1 hour. Water (20 mL) and DCM (40 mL) were added and the layers were separated. The aqueous layer was extracted with DCM (x2) and the combined organic layers were washed with saturated aqueous NaHC03, dried over sodium sulfate, filtered and concentrated to dryness. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 100% EtOAc in heptane. Product fractions were concentrated to dryness to afford ((65,,85,)-6-(4-bromo-2,6-difluorophenyl)-7-(2-fluoro-2-methylpropyl)-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-8-yl)methanol (424 mg, 45%) as a beige solid. XH NMR (500 MHz, CDCI3, 27 °C) 1.31 (3H, d), 1.36 (3H, d), 2.52 (1H, dd), 2.95 (1H, dd), 3.05 (1H, dd), 3.12 (1H, dd), 3.62 (2H, dd), 3.70 - 3.90 (1H, m), 5.50 (1H, s), 6.87 (1H, d), 7.01 (2H, d), 7.19 - 7.25 (1H, m), 8.08 (1H, d). m/z: ES+ [M+H]+ 468. Preparation of (6^,8^)-6-(4-bromo-2,6-difluorophenyl)-7-(2-fluoro-2-methylpropyl)-
6,7,8,9-tetrahvdro-3H-pyrazolor4,3-f1isoguinoline-8-carbaldehvde
Figure imgf000129_0001
Sufur trioxide-pyridine complex (477 mg, 3.00 mmol) was added as a solution in DMSO (5.7 mL) dropwise to a cooled solution of ((6S,8S)-6-(4-bromo-2,6-difluorophenyl)-7-(2- fluoro-2-methylpropyl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-8-yl)methanol (702 mg, 1.50 mmol) and triethylamine (0.5 mL, 3.75 mmol) in a mixture of DCM (5.7 mL) and DMSO (5.7 mL) at 0 °C and the reaction was allowed to warm to room temperature over 2 hours. The reaction was diluted with DCM (50 mL) and water (50 mL) and the layers were separated. The aqueous layer was extracted with DCM (x2) and the combined organic layers were washed with saturated aqueous sodium chloride, dried over magnesium sulfate, filtered and concentrated to dryness. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 100% EtOAc in heptane. Product fractions were concentrated to dryness to afford (65,,85,)-6-(4-bromo-2,6-difluorophenyl)-7- (2-fluoro-2-methylpropyl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline-8- carbaldehyde (512 mg, 73%) as a beige solid. XH NMR (500 MHz, CDC13, 27 °C) 1.10 (3H, d), 1.23 (3H, d), 2.56 (1H, dd), 3.35 (1H, t), 3.50 (1H, dd), 3.58 (1H, dd), 4.41 (1H, dt), 5.90 (1H, s), 6.78 (1H, d), 6.92 - 7.1 (2H, m), 7.20 (1H, d), 8.11 (1H, d), 9.75 (1H, s), 10.12 (1H, s). m/z: ES+ [M+H]+ 466.
Preparation of (6^,8^)-6-(4-bromo-2,6-difluorophenyl)-7-(2-fluoro-2-methylpropyl)-
3-(tetrahvdro-2H-pyran-2-yl)-6,7,8,9-tetrahvdro-3H-pyrazolor4,3-f1isoguinoline-8- carbaldehyde
Figure imgf000130_0001
3,4-Dihydro-2H-pyran (0.24 mL, 2.57 mmol) and /?-toluenesulfonic acid hydrate (32.8 mg, 0.17 mmol) were added to a solution of (6S,8S)-6-(4-bromo-2,6-difluorophenyl)-7-(2- fluoro-2-methylpropyl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline-8-carbaldehyde (800 mg, 1.72 mmol) in DCM (8.3 mL) and the reaction was stirred at room temperature for 1 hour. The reaction was then maintained under reflux conditions for 3 hours. After cooling, the reaction was diluted with DCM (25 mL) and washed with saturated aqueous NaHCCb (25 mL) before being dried over sodium sulfate, filtered and concentrated to dryness. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 50% EtOAc in heptane. Product fractions were concentrated to dryness to afford (6lS,,85,)-6-(4-bromo-2,6-difluorophenyl)-7-(2-fluoro-2-methylpropyl)-3-(tetrahydro-
2H-pyran-2-yl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline-8-carbaldehyde (772 mg, 82%) as a pale yellow solid. lH NMR (500 MHz, CDC13, 27 °C) 1.09 (3H, d), 1.23 (3H, dd), 1.59 - 1.68 (1H, m), 1.69 - 1.81 (2H, m), 2.00 - 2.09 (1H, m), 2.09 - 2.20 (1H, m), 2.43 - 2.65 (2H, m), 3.27 - 3.44 (1H, m), 3.48 (1H, dt), 3.51 - 3.60 (1H, m), 3.66 - 3.74 (1H, m), 3.94 - 4.07 (1H, m), 4.38 (1H, ddt), 5.64 (1H, ddd), 5.89 (1H, s), 6.77 (1H, d), 7.01 (2H, d), 7.27 - 7.33 (1H, m), 8.05 (1H, d), 9.72 (1H, d). m/z: ES+ [M+H]+ 550. Preparation of (6^,8^)-6-(4-bromo-2,6-difluorophenyl)-8-(difluoromethyl)-7-(2- fluoro-2-methylpropyl)-3-(tetrahvdro-2H-pyran-2-yl)-6,7,8,9-tetrahydro-3H-
Pyrazolor4,3-f1isoguinoline
Figure imgf000131_0001
Diethylamino sulfur trifluoride (0.384 mL, 2.91 mmol) was added to a cooled solution of (65,,85,)-6-(4-bromo-2,6-difluorophenyl)-7-(2-fluoro-2-methylpropyl)-3-(tetrahydro-2H- pyran-2-yl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline-8-carbaldehyde (800 mg, 1.45 mmol) in DCM (13.3 mL) at 0 °C. The reaction was allowed to warm to room temperature and stirred for 5 hours. After dilution with DCM (20 mL), the reaction was quenched by addition of saturated aqueous NaHC03. The layers were separated and the aqueous layer was extracted with DCM. The combined organic layers were dried over sodium sulfate, filtered and concentrated to dryness. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 25% EtOAc in heptane. Product fractions were concentrated to dryness to afford (6lS,,85,)-6-(4-bromo-2,6-difluorophenyl)-8- (difluoromethyl)-7-(2-fluoro-2-methylpropyl)-3-(tetrahydro-2H-pyran-2-yl)-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinoline (753 mg, 91%) as a beige solid. XH NMR (500
MHz, CDC13, 27 °C) 1.15 (3H, d), 1.26 (3H, dd), 1.59 - 1.83 (3H, m), 2.04 - 2.11 (1H, m), 2.14 (1H, dd), 2.41 - 2.51 (1H, m), 2.51 - 2.64 (1H, m), 3.25 (1H, dt), 3.34 - 3.52 (2H, m), 3.72 (1H, ddd), 3.91 (1H, s), 3.97 - 4.05 (1H, m), 5.59 (1H, s), 5.66 (1H, dt), 5.83 (1H, tdd), 6.78 (1H, dd), 7.01 (2H, d), 7.31 (1H, dd), 8.04 (1H, d). m/z: ES+ [M+H]+ 572. Preparation of fe -butyl (E)-3-(4-((6^,8^)-8-(difluoromethyl)-7-(2-fluoro-2- methylpropyl)-3-(tetrahvdro-2H-pyran-2-yl)-6,7,8,9-tetrahvdro-3H-pyrazolor4,3- f1iso uinolin-6-yl)-3,5-difluorophenyl)acrylate
Figure imgf000132_0001
Tert-butyl acrylate (0.044 mL, 0.30 mmol) was injected into a sealed microwave vial contaning a degassed solution of (65,,85,)-6-(4-bromo-2,6-difluorophenyl)-8- (difluoromethyl)-7-(2-fluoro-2-methylpropyl)-3-(tetrahydro-2H-pyran-2-yl)-6,7,8,9- tetrahydro-3H-pyrazolo[4,3-f]isoquinoline (114 mg, 0.20 mmol), [l, l '-bis(di-tert- butylphosphino)ferrocene]dichloropalladium(II) (6.54 mg, 10.0 μιηοΐ) and N,N- diisopropylethylamine (0.069 mL, 0.40 mmol) in 1,4-dioxane (1.7 mL). The reaction was heated in the microwave to 125 °C and maintained under these conditions for 1 hour. After cooling, the crude mixture was concentrated to dryness. The crude product was purified by flash silica chromatography, elution gradient 0 to 25% EtOAc in heptane. Product fractions were concentrated to dryness to afford iert-butyl (E)-3-(4-((65,,85,)-8-(difluoromethyl)-7- (2-fluoro-2-methylpropyl)-3-(tetrahydro-2H-pyran-2-yl)-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3,5-difluorophenyl)acrylate (125 mg, 100%) as a beige gum. XH NMR (500 MHz, CDC13, 27 °C) 1.08 - 1.16 (3H, m), 1.23 (3H, dd), 1.52 (9H, s), 1.56 - 1.81 (2H, m), 2.04 - 2.10 (IH, m), 2.13 (IH, dd), 2.42 - 2.61 (2H, m), 3.26 (IH, dt), 3.45 (2H, qd), 3.65 - 3.77 (IH, m), 3.94 (IH, d), 3.98 - 4.07 (2H, m), 5.63 (IH, s), 5.66 (IH, dd), 5.84 (IH, tdd), 6.31 (IH, dd), 6.80 (IH, d), 6.95 (2H, d), 7.31 (IH, dd), 7.41 (IH, d), 8.05 (IH, s). m/z: ES+ [M+H]+ 620. Example 11: (E)-3-(4-((6^,8^)-8-(difluoromethyl)-7-(2-fluoro-2-methylpropyl)-6- methyl-6,7,8,9-tetrahvdro-3H-pyrazolor4,3-f1isoguinolin-6-yl)-3,5- difluorophenvDacrylic acid
Figure imgf000133_0001
Aqueous NaOH (2N; 0.73 mL, 1.54 mmol) was added to a solution of methyl (E)-3-(4- ((6lS,,85,)-8-(difluoromethyl)-7-(2-fluoro-2-methylpropyl)-6-methyl-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3,5-difluorophenyl)acrylate (92 mg, 0.18 mmol) in THF (0.55 mL) and methanol (0. 55 mL) and the reaction was stirred at room temperature for 2 hours. EtOAc (10 mL) and water (10 mL) were added and then the pH of the aqueous layer was adjusted to ~5 by addition of aqueous HC1 (2N). The layers were separated and the aqueous layer was extracted with EtOAc. The combined organic layers were dried over sodium sulfate, filtered and concentrated to dryness. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 100% EtOAc in heptane. Product fractions were concentrated to dryness to afford (E)-3-(4-((65,,85,)-8-(difluoromethyl)-7-(2- fluoro-2-methylpropyl)-6-methyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)- 3,5-difluorophenyl)acrylic acid (66 mg, 74%) as a colourless solid. XH NMR (500 MHz,
CDC , 27 °C) 0.98 (3H, d), 1.20 (3H, d), 1.97 (3H, s), 2.55 (1H, dd), 3.26 (1H, d), 3.52 (1H, d), 3.59 - 3.70 (1H, m), 4.11 (1H, d), 5.91 (1H, td), 6.39 (1H, d), 6.91 (2H, d), 6.96 (1H, d), 7.25 (1H, d), 7.55 (1H, d), 8.13 (1H, s). m/z: ES+ [M+H]+ 494.
The methyl (E)-3-(4-((65,,8lS,)-8-(difluoromethyl)-7-(2-fluoro-2-methylpropyl)-6-methyl- 6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3,5-difluorophenyl)acrylate used as starting material was prepared as follows; Preparation of (6^,8^)-6-(4-bromo-2,6-difluorophenyl)-8-(difluoromethyl)-7-(2- fluoro-2-methylpropyl)-6-methyl-3-(tetrahvdro-2H-pyran-2-yl)-6,7,8,9-tetrahydro-
3H-pyrazolor4,3-flisoquinoline
Figure imgf000134_0001
Ceric (IV) ammonium nitrate (1.0 g, 1.83 mmol) was added to a solution of (6S,SS)-6-(4- bromo-2,6-difluorophenyl)-8-(difluoromethyl)-7-(2-fluoro-2-methylpropyl)-3-(tetrahydro- 2H-pyran-2-yl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline (500 mg, 0.87 mmol) in a mixture of acetonitrile (4.8 mL) and water (1.2 mL) and the reaction was stirred at room temperature for 1 hour. The reaction was then diluted with EtOAc (30 mL) and washed with saturated aqueous sodium chloride (30 mL). The aqueous layer was extracted with EtOAc (x2) and the combined organic layers were dried over magnesium sulfate, filtered and concentrated to dryness to afford the intermediate iminium ion as a red solid. The residue was dissolved in THF (5 mL) and cooled to -78 °C. Methylmagnesium bromide in diethyl ether (2.7 M; 0.97 mL, 2.62 mmol) was added and the reaction was maintained under these conditions for 1 hour. The reaction was quenched by addition of water and saturated aqueous NH4C1, before being extracted with EtOAc (x2). The combined organic layers were dried over sodium sulfate, filtered and concentrated to dryness. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 40% EtOAc in heptane. Product fractions were concentrated to dryness to afford (65,,85,)-6-(4-bromo-2,6- difluorophenyl)-8-(difluoromethyl)-7-(2-fluoro-2-methylpropyl)-6-methyl-3-(tetrahydro- 2H-pyran-2-yl)-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinoline (301 mg, 59%) as a colourless solid. XH NMR (500 MHz, CDCI3, 27 °C) 0.99 (3H, dd), 1.21 (3H, d), 1.60 - 1.68 (1H, m), 1.68 - 1.82 (2H, m), 1.92 (3H, s), 2.04 - 2.11 (1H, m), 2.11 - 2.20 (1H, m), 2.42 - 2.64 (2H, m), 3.23 (1H, d), 3.47 (1H, dt), 3.63 (1H, t), 3.68 - 3.76 (1H, m), 3.97 - 4.11 (2H, m), 5.62 - 5.68 (1H, m), 5.86 (1H, tdd), 6.91 (3H, dd), 7.31 (1H, dd), 8.05 (1H, t). m/z: ES+ [M+H]+ 586. Preparation of methyl (E)-3-(4-((6^,8^)-8-(difluoromethyl)-7-(2-fluoro-2- methylpropyl)-6-methyl-3-(tetrahvdro-2H-pyran-2-yl)-6,7,8,9-tetrahvdro-3H-
Pyrazolor4,3-f1iso uinolin-6-yl)-3,5-difluorophenyl)acrylate
Figure imgf000135_0001
Methyl acrylate (34.6 μί, 0.38 mmol) was added to a sealed microwave vial containing a degassed solution of (65,,85,)-6-(4-bromo-2,6-difluorophenyl)-8-(difluoromethyl)-7-(2- fluoro-2-methylpropyl)-6-methyl-3-(tetrahydro-2H-pyran-2-yl)-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinoline (141 mg, 0.24 mmol), [l, l'-bis(di-tert- butylphosphino)ferrocene]dichloropalladium(II) (8 mg, 0.01 mmol) and N,N- diisopropylethylamine (83 μί, 0.48 mmol) in 1,4-dioxane (2.28 mL). The reaction was heated in the microwave to 125 °C and maintained under these conditions for 1 hour. After cooling, the crude mixture was concentrated to dryness. The crude product was purified by flash silica chromatography, elution gradient 0 to 50% EtOAc in heptane. Product fractions were concentrated to dryness to afford methyl (E)-3-(4-((65,,85,)-8-(difluoromethyl)-7-(2- fluoro-2-methylpropyl)-6-methyl-3-(tetrahydro-2H-pyran-2-yl)-6,7,8,9-tetrahydro-3H- pyrazolo[4,3-f]isoquinolin-6-yl)-3,5-difluorophenyl)acrylate (136 mg, 96%) as a beige gum. XH NMR (500 MHz, CDC13, 27 °C) 0.97 (3H, dd), 1.19 (3H, d), 1.44 - 1.57 (1H, m), 1.59 - 1.79 (2H, m), 1.95 (3H, s), 2.05 - 2.11 (1H, m), 2.14 (1H, dd), 2.44 - 2.63 (2H, m), 3.24 (1H, d), 3.44 - 3.56 (1H, m), 3.55 - 3.75 (2H, m), 3.79 (3H, s), 3.96 - 4.13 (2H, m), 5.65 (1H, ddd), 5.87 (1H, tdd), 6.35 (1H, dd), 6.87 (2H, d), 6.93 (1H, d), 7.31 (1H, dd), 7.48 (1H, d), 8.06 (1H, dd). m/z: ES+ [M+H]+ 592. Preparation of methyl (E)-3-(4-((6^,8^)-8-(difluoromethyl)-7-(2-fluoro-2- methylpropyl)-6-methyl-6J,8,9 etrahvdro-3H-pyrazolor4,3-flisoguinolin-6-yl)-3,5- difluorophenvDacrylate
Figure imgf000136_0001
HC1 in dioxane (4N; 0.58 mL) was added to a solution of methyl (E)-3-(4-((65,85)-8- (difluoromethyl)-7-(2-fluoro-2-methylpropyl)-6-methyl-3-(tetrahydro-2H-pyran-2-yl)- 6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3,5-difluorophenyl)acrylate (137 mg, 0.23 mmol) in MeOH (0.58 mL) and the reaction was stirred at room temperature for 3 hours. The solvents were evaporated and the resulting residue was dissolved in DCM and washed with saturated aqueous NaHCCb. The aqueous layer was extracted with DCM and the combined organic layers were dried over sodium sulfate, filtered and concentrated to dryness. The resulting residue was purified by flash silica chromatography, elution gradient 0 to 50% EtOAc in heptane. Product fractions were concentrated to dryness to afford methyl (E)-3-(4-((6lS,,85,)-8-(difluoromethyl)-7-(2-fluoro-2-methylpropyl)-6-methyl- 6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolin-6-yl)-3,5-difluorophenyl)acrylate as a beige solid. XH NMR (500 MHz, CDC13, 27 °C) 0.98 (3H, d), 1.19 (3H, d), 1.97 (3H, t), 2.55 (1H, dd), 3.27 (1H, d), 3.46 - 3.59 (1H, m), 3.59 - 3.73 (1H, m), 3.79 (3H, s), 4.12 (1H, q), 5.91 (1H, td), 6.35 (1H, d), 6.88 (2H, d), 6.94 (1H, d), 7.23 (1H, d), 7.48 (1H, d), 8.14 (1H, d), 10.87 (1H, s). m/z: ES+ [M+H]+ 508. Example 12: (E)-3-r4-r(6^,8R)-7- -fluoro-2-methyl-propyl)-8-methyl-3,6,8,9- tetrahvdropyrazolor4,3-niso uinolin-6-yl1-3-methoxy-phenyl1prop-2-enoic acid
Figure imgf000137_0001
2M sodium hydroxide solution (2.0 ml, 4.00 mmol) was added to a solution of methyl (£)- 3-[4-[(65,8R)-3-acetyl-7-(2-fluoro-2-methyl-propyl)-8-methyl-8,9-dihydro-6H- pyrazolo[4,3-/]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoate (197 mg, 0.40 mmol) in THF (1.0 ml) and methanol (1.0 ml) and the reaction was stirred at ambient temperature for 2h. EtOAc (10 mL) and water (10 mL) were added and the pH of the aqueous phase was adjusted to ~6 by addition of 2N HC1 solution. The layers were separated and the aqueous phase was extracted with EtOAc (2 x 10 mL). The combined organic phases were dried (MgS04) and concentrated. The crude product was purified by flash silica chromatography, elution gradient 25 to 100% EtOAc in heptane. Pure fractions were evaporated to dryness to afford (E)-3-[4-[(65,,8R)-7-(2-fluoro-2-methyl-propyl)-8-methyl- 3,6,8,9-tetrahydropyrazolo[4,3-/]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoic acid (115 mg, 65%) as a pale yellow solid. lH NMR (400 MHz, CDC13, 27 °C) 1.06 (3H, d), 1.22 (3H, d), 1.26 - 1.3 (3H, m), 2.31 (1H, dd), 2.75 - 3.01 (2H, m), 3.34 (1H, dd), 3.75 (2H, q), 3.95 (3H, s), 5.44 (1H, s), 6.43 (1H, d), 6.78 (1H, d), 6.97 (1H, d), 7.02 (1H, d), 7.08 (1H, s), 7.17 (1H, d), 7.72 (1H, d), 8.08 (1H, s). m/z (ES+), [M+H]+ = 438.
The methyl (E)-3-[4-[(65,8R)-3-acetyl-7-(2-fluoro-2-methyl-propyl)-8-methyl-8,9- dihydro-6H-pyrazolo[4,3-/]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoate used as starting material in this reaction was prepared as follows:
2-fluoro-2-methylpropan-l-ol
Figure imgf000137_0002
Lithium aluminium hydride (3.37 g, 88.6 mmol) was added portion wise over 15 min to a cooled solution of ethyl 2-fluoro-2-methylpropanoate (9.9 g, 73.8 mmol) in diethyl ether (184 ml) at 0 °C. The reaction was stirred for 1 hr, then water (3.3 ml), followed by 15% NaOH solution (3.3 ml) and water (6.7 ml) were added sequentially. The suspension was stirred for 15 min, then filtered and the solids washed with diethyl ether. The filtrate was evaporated to give 2-fluoro-2-methylpropan-l-ol (5.90 g, 87%) as a colourless oil. XH NMR (400 MHz, CDC13, 27 °C) 1.37 (6H, d), 3.56 (2H, d), OH not observed. -fluoro-2-methylpropyl trifluoromethanesulfonate
Figure imgf000138_0001
Method A
Trifluoromethanesulfonic anhydride (12.06 ml, 71.24 mmol), followed by 2,6-lutidine (11.42 ml, 81.42 mmol) were added to a solution of 2-fluoro-2-methylpropan-l-ol (6.25 g, 67.8 mmol) in DCM (146 ml) at -10 °C. The reaction was stirred for 1 hr, then washed with 2N HC1 (2 x 100 ml) and saturated NaHCOs solution (2 x 100 ml). The organic phase was then dried over Na2S04 and concentrated to give 2-fluoro-2-methylpropyl
trifluoromethanesulfonate (12.89 g, 85%) as a red oil.
Method B
Trifluoromethanesulfonic anhydride (19.29 ml, 113.99 mmol) was added to a solution of 2-fluoro-2-methylpropan-l-ol (10.00 g, 108.56 mmol) in DCM (183 ml) at -10 °C (ice- acetone). Following the addition, 2,6-dimethylpyridine (15.17 ml, 130.3 mmol) was added in DCM (80 mL) dropwise and the reaction was stirred for 1 hr. 2N HC1 (150 mL) was added and the layers were separated. The aqueous layer was extracted with DCM, then the combined organic phases were dried and concentrated. The residue was passed through a plug of silica gel, eluting with DCM. The filtrate was carefully evaporated (no lower than 100 mbar, 40 °C) to afford 2-fluoro-2-methylpropyl trifluoromethanesulfonate (21.91 g, 90%) as a brown/red oil.
XH NMR (500 MHz, CDCI3, 27°C) 1.46 (6H, d), 4.41 (2H, d). tert-Butyl (4R)-4-methyl-2,2-dioxo-oxathiazolidine-3-carboxylate
Figure imgf000139_0001
To a solution of lH-imidazole (38.9 g, 570.69 mmol) and triethylamine (45.6 ml, 328.15 mmol) in anhydrous dichloromethane (524 ml) at -50 °C was added thionyl chloride (11.97 ml, 164.07 mmol) dropwise (exothermic, keeping T < -40 °C). The mixture was stirred for 5 minutes while cooling to -60 °C and a solution of (R)-tert-butyl l-hydroxypropan-2- ylcarbamate (25 g, 142.6 mmol) in anhydrous dichloromethane (524 ml) was added dropwise over 3 hours. The reaction was stirred while warming to room temperature overnight. Water was added (-500 mL) and the phases separated. The aqueous phase was further extracted into dichloromethane (150 mL). The combined organic phases were washed with water (200 mL), saturated brine (150 mL), dried over MgS04, filtered and concentrated in vacuo to give the sulfamidite intermediate tert-butyl (4R)-4-methyl-2- oxido-oxathiazolidin-2-ium-3-carboxylate (31.32 g, 142 mmol, 99%) as a pale oil.
Ruthenium(III) chloride hydrate (0.022 g, 0.10 mmol) was added to a stirred mixture of tert-butyl (4R)-4-methyl-2-oxido-oxathiazolidin-2-ium-3-carboxylate (31.11 g, 140.59 mmol) in acetonitrile (277 ml) and water (149 ml) at room temperature, followed by portion wise addition of sodium periodate (33.1 g, 154.65 mmol). An initial endothermic reaction to 12 °C (dissolution of periodate) was followed by a significant exotherm to -40 °C (white solid precipitated). The biphasic mixture was stirred at room temperature for 2 hours. The mixture was diluted into water (500 mL) and extracted into 3 x 500 mL ethyl acetate . The combined organics were washed with water, brine, dried over MgS04, filtered and concentrated in vacuo to give tert-butyl (4R)-4-methyl-2,2-dioxo- oxathiazolidine-3-carboxylate (31.7 g, 134 mmol, 95%) as a colourless solid. XH NMR (400 MHz, CDC , 30 °C) 1.51 (3H, d), 1.55 (9H, s), 4.19 (1H, dd), 4.41 (1H, pd), 4.66 (1H, dd). No mass ion detected. (2R)-l-(3-bromo-2-methyl-phenyl)propan-2-amine
Figure imgf000140_0001
w-BuLi (1.6M in hexanes) (67.6 mL, 108.10 mmol) was added dropwise to a solution of l,3-dibromo-2-methylbenzene (25.73 g, 102.95 mmol) in THF (250 mL) at -78 °C at such a rate as to keep the internal reaction temperature below -60 °C. After stirring for 30 min, tert-butyl (4R)-4-methyl-2,2-dioxo-oxathiazolidine-3-carboxylate (26.9 g, 113.24 mmol) was added in portions and the reaction was stirred for a further 30 min before being allowed to warm to 0 °C over 2 hours. IN Citric acid (200 mL) was added (at 0 °C) and the mixture was stirred for 15 min before it was extracted with EtOAc (2 x 250 mL). The combined organic layers were evaporated. The residue was stirred in 4N HC1 in dioxane (100 mL) at room temperature for 1 hour and then concentrated. The residue was dissolved in water (200 mL) and extracted with diethyl ether (2 x 200 mL). The aqueous was then basified by addition of Na2C03 and extracted with DCM (3 x 250 mL). The combined DCM extracts were dried (MgS04) and concentrated to afford (2R)-l-(3-bromo-2-methyl- phenyl)propan-2-amine (14.36 g, 61%) as a yellow oil. lH NMR (400 MHz, CDC13, 30 °C) 1.13 (3H, d), 2.40 (3H, s), 2.62 (1H, dd), 2.77 (1H, dd), 3.16 (1H, ddd), 6.97 (1H, t), 7.09(1H, dd), 7.43 (1H, dd). NH2 was not observed, m/z: ES+ [M+H]+ 228/230.
N-r(lR)-2-(3-bromo-2-methyl-phenyl)-l-methyl-ethyl1-2-fluoro-2-methyl-propan-l- amine
Figure imgf000140_0002
Method A
2-Fluoro-2-methylpropyl trifluoromethanesulfonate (35.2 g, 157.2 mmol) was added to a solution of (2R)-l-(3-bromo-2-methyl-phenyl)propan-2-amine (32.6 g, 142.90 mmol) and DIPEA (30.9 mL, 178.63 mmol) in dioxane (400 mL). The reaction was stirred at 85 °C for 21 hours (during this time a further 5 g of 2-fluoro-2-methylpropyl
trifluoromethanesulfonate was added). After cooling, the reaction was diluted with DCM (500 mL) and washed with water (500 mL). The organic phase was dried (MgS04) and concentrated. The crude product was purified by flash silica chromatography, elution gradient 20 to 80% EtOAc in heptane. Pure fractions were evaporated to dryness to afford N-[(lR)-2-(3-bromo-2-methyl-phenyl)-l-methyl-ethyl]-2-fluoro-2-methyl-propan-l-amine (32.0 g, 74%) as an orange oil.
Method B
2-Fluoro-2-methylpropyl trifluoromethanesulfonate (8.77 g, 39.11 mmol) was added to a solution of (2R)-l-(3-bromo-2-methylphenyl)propan-2-amine (7.138 g, 31.29 mmol) and DIPEA (9.46 ml, 54.76 mmol) in 1,4-dioxane (68.8 mL). The reaction was heated to 90 °C for 2 hr. After cooling, the solution was diluted with DCM and washed with water. The aqueous phase was extracted with DCM, then the combined organic phases were dried over MgS04 and concentrated. The crude product was purified by flash silica
chromatography, elution gradient 50 to 100% EtOAc in heptane. Pure fractions were evaporated to dryness to afford N-[(lR)-2-(3-bro mo-2-methyl-phenyl)-l -methyl-ethyl] -2- fluoro-2-methyl-propan-l -amine (9.10 g, 96%) as a yellow gum.
XH NMR (500 MHz, CDC13, 27°C) 1.05 (3H, d), 1.35 (6H, d), 2.41 (3H, s), 2.57 - 2.84 (3H, m), 2.88 (2H, tdd), 6.93 - 7.00 (1H, m), 7.08 (1H, dd), 7.42 (1H, dd). m/z: ES+
[M+H]+ 302. -r(2R)-2-r(2-fluoro-2-methyl-propyl)amino1propyl1-2-methyl-aniline
Figure imgf000141_0001
Method A
Degassed toluene (90 mL) was added to N-[(lR)-2-(3-bromo-2-methyl-phenyl)-l-methyl- ethyl]-2-fluoro-2-methyl-propan-l -amine (7 g, 23.16 mmol) followed by the addition of diphenylmethanimine (3.89 mL, 23.16 mmol), sodium iert-butoxide (3.34 g, 34.74 mmol), 2,2'-bis(diphenylphosphino)-l,l'-binaphthalene (0.433 g, 0.69 mmol), and
tris(dibenzylideneacetone)dipalladium(0) (0.318 g, 0.35 mmol). The reaction was heated to 90 °C for 2.5 hours under nitrogen. After cooling, the volatiles were removed under vacuum and the residue was dissolved in DCM (75 mL). IN HC1 (75 mL) was added and the biphasic mixture was stirred vigorously for 1 hour. The layers were separated and the aqueous was extracted with DCM. The aqueous was then basified by addition of 2N NaOH solution (40 mL), and extracted with DCM (3 x 150 mL). The combined basic extracts were dried (MgS04) and concentrated (5.24 g). The crude product was purified by flash silica chromatography (column base deactivated), elution gradient 10-50% EtOAc in heptane. Pure fractions were evaporated to dryness to afford 3-[(2R)-2-[(2-fluoro-2- methyl-propyl)amino]propyl]-2-methyl-aniline (4.66 g, 84%) as a yellow oil.
Method B
(R)-N-(l-(3-bromo-2-methylphenyl)propan-2-yl)-2-fluoro-2-methylpropan- l-amine (9.10 g, 30.11 mmol), diphenylmethanimine (5.56 ml, 33.12 mmol), sodium tert-butoxide (4.34 g, 45.16 mmol) and BINAP (0.750 g, 1.20 mmol) were added to a round bottom flask and suspended in toluene (145 ml). The solvent was degassed (evacuated and back-filled with nitrogen x 3), then Pd2(dba)3 (0.551 g, 0.60 mmol) was added and the reaction was heated to 90 °C for 3 hr. After cooling, the toluene was evaporated by vacuum, and the residue was dissolved in DCM (75 mL). IN HCl (75 mL) was added and the biphasic mixture was stirred for 1 hr. The layers were separated and the aqueous layer was extracted with DCM. The aqueous layer was then basified by addition of Na2C03 solution and extracted with DCM (x3). The combined basic extracts were dried over MgS04 and concentrated to afford (R)-3-(2-((2-fluoro-2-methylpropyl)amino)propyl)-2-methylaniline (6.82 g, 95%) as a yellow/brown oil.
XH NMR (500 MHz, CDC13, 27°C) 1.05 (3H, d), 1.34 (6H, d), 2.12 (3H, s), 2.42 - 2.93 (5H, m), 3.58 (2H, s), 6.49 - 6.69 (2H, m), 6.95 (1H, t). m/z: ES+ [M+H]+ 239.
4-bromo-2-methoxybenzaldehyde
Figure imgf000142_0001
Sodium methanolate (5.06 g, 93.59 mmol) was added to a solution of 4-bromo-2- fluorobenzaldehyde (9.5 g, 46.80 mmol) in methanol (117 mL). The reaction was heated to 70 °C for 6 hrs. 3 extra portions of sodium methanolate (5.06 g, 93.59 mmol) were added at intervals. After cooling, the volatiles were concentrated to -10 mL, and the reaction was diluted with EtOAc (200 mL) and washed with brine (100 mL). The organic layer was dried over Na2S04 and concentrated to give 4-bromo-2-methoxybenzaldehyde (9.45 g, 94%) as a cream solid. lH NMR (400 MHz, CDC13, 27 °C) 3.94 (3H, s), 7.13 - 7.22 (2H, m), 7.69 (1H, d), 10.39 (1H, d). No mass ion observed in LCMS.
Methyl (E)-3-(4-formyl-3-methoxyphenyl)prop-2-enoate
Figure imgf000143_0001
Methyl acrylate (5.94 ml, 65.92 mmol) was added in one portion to a degassed solution of 4-bromo-2-methoxybenzaldehyde (9.45 g, 43.9 mmol), tri-o-tolylphosphine (1.338 g, 4.39 mmol) and palladium (II) acetate (0.493 g, 2.20 mmol) in DMA (95 ml). Triethylamine (9.19 ml, 65.92 mmol) was added and the reaction stirred at 100 °C for 1 hr. The reaction mixture was cooled and poured into water (300 mL) and extracted with EtOAc (3 x 150 mL). The combined organic phases were washed with IN citric acid (150 mL) and saturated brine (100 mL), dried (MgS04) and evaporated to dryness. The crude product was purified by flash silica chromatography, elution gradient 10 to 50% EtOAc in heptane. Fractions containing the desired product were evaporated and triturated with 20% EtOAc in heptane (50 ml) to afford a solid which was collected by filtration and dried under vacuum to give methyl (E)-3-(4-formyl-3-methoxyphenyl)prop-2-enoate (6.19 g, 64%) as a pale yellow solid. XH NMR (400 MHz, CDCI3, 27 °C) 3.83 (3H, s), 3.97 (3H, s), 6.52 (1H, d), 7.09 (1H, d), 7.16 - 7.23 (1H, m), 7.67 (1H, d), 7.84 (1H, d), 10.45 (1H, d). m/z: ES- [M-H]- 219. MethvnE)-3-r4-r(l^,3R)-6-amino-2-(2-fluoro-2-methyl-propyl)-3,5-dimethyl-3,4- dihydro- lH-isoquinolin- 1-vH -3-methoxy-phenyl1prop-2-enoate
Figure imgf000144_0001
Methyl (E)-3-(4-formyl-3-methoxyphenyl)prop-2-enoate (0.832 g, 3.78 mmol) was added to a solution of 3-[(2R)-2-[(2-fluoro-2-methyl-propyl)amino]propyl]-2-methyl-aniline (0.500 g, 2.10 mmol) in acetic acid (10.30 ml) and water (0.189 ml, 10.49 mmol). The reaction was heated to 100 °C for 4h. After cooling, the acetic acid was evaporated under vacuum and the residue was dissolved in DCM (10 mL). IN HCl (10 mL) was added and the biphasic mixture was stirred at ambient temperature for lh. The layers were separated and the aqueous layer was extracted with DCM. The aqueous layer was then basified by addition of solid Na2C03 and extracted with DCM (2 x 10 mL). The combined organic extracts were dried (MgS04) and concentrated. The crude product was purified by flash silica chromatography, elution gradient 0 to 60% EtOAc in heptane. Pure fractions were evaporated to dryness to afford methyl (E)-3-[4-[(lS,3R)-6-amino-2-(2-fluoro-2-methyl- propyl)-3,5-dimethyl-3,4-dihydro- lH-isoquinolin- l-yl]-3-methoxy-phenyl]prop-2-enoate
(0.513 g, 56%) as a colourless solid. XH NMR (400 MHz, DMSO, 27 °C) 1.00 (3H, d), 1.23 (3H, d), 1.28 (3H, d), 2.07 (3H, s), 2.27 (1H, dd), 2.50 (1H, dd), 2.74 (1H, dd), 2.88 (1H, dd), 3.48 (2H, s), 3.54 - 3.66 (1H, m), 3.79 (3H, s), 3.91 (3H, s), 5.29 (1H, s), 6.38 (1H, d), 6.44 (1H, d), 6.92 - 7 (2H, m), 6.99 - 7.04 (2H, m), 7.63 (1H, d). m/z (ES+), [M+H]+ = 441. MethvnE)-3 4-r(6^,8R)-3-acetyl-7- -fluoro-2-methyl-propyl)-8-methyl-8,9-dihvdro- 6H- razolor4,3-niso uinolin-6-yl1-3-methoxy-phenyl1prop-2-enoate
Figure imgf000145_0001
Acetic anhydride (142 μΐ, 1.50 mmol) was added to a solution of methyl (E)-3-[4-[(15',3R)- 6-amino-2-(2-fluoro-2-methyl-propyl)-3,5-dimethyl-3,4-dihydro- lH-isoquinolin- l-yl]-3- methoxy-phenyl]prop-2-enoate (441 mg, 1.00 mmol) and potassium acetate (123 mg, 1.25 mmol) in chloroform (9.5 mL) under an atmosphere of nitrogen and the reaction was heated to 70 °C for 30 min. 18-Crown-6 (66.1 mg, 0.25 mmol) and isopentyl nitrite (336 μΐ, 2.50 mmol) were added and the reaction was stirred at 70 °C for 18 h. The reaction was diluted with DCM (25 mL) and washed with water (25 mL). The aqueous phase was extracted with DCM (25 mL) then the combined organics were dried (MgS04) and concentrated. The crude product was purified by flash silica chromatography, elution gradient 0 to 50% EtOAc in heptane. Pure fractions were evaporated to dryness to afford methyl (E)-3-[4-[(65,8R)-3-acetyl-7-(2-fluoro-2-methyl-propyl)-8-methyl-8,9-dihydro-6H- pyrazolo[4,3-/]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoate (205 mg, 42%) as an off-white solid. ¾ NMR (400 MHz, CDC13, 27 °C) 1.04 - 1.07 (3H, d), 1.20 (3H, d), 1.24 - 1.27 (3H, d), 2.25 (1H, d), 2.32 (1H, d), 2.76 (3H, s), 2.8 - 2.87 (1H, m), 2.88 - 2.96 (1H, m), 3.38 (1H, dd), 3.79 (3H, s), 3.95 (3H, s), 5.44 (1H, s), 6.40 (1H, d), 6.93 (1H, d), 6.95 - 7.01 (2H, m), 7.05 (1H, d), 7.63 (1H, d), 8.05 (1H, d), 8.13 (1H, d). m/z (ES+), [M+H]+ = 494. Alternative Preparation of Example 12: (E)-3-r4-r(6^,8R)-7-(2-fluoro-2-methyl- propyl)-8-methyl-3,6,8,9-tetrahvdropyrazolor4,3-niso uinolin-6-yl1-3-methoxy- phenyllprop-2-enoic acid
Figure imgf000146_0001
2.0M Sodium hydroxide (23.35 mL, 46.71 mmol) was added to a solution of methyl (E)-3- (4-((65',8R)-7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoate (2.109 g, 4.67 mmol) in THF (20 mL) / methanol (20 mL). The reaction was stirred at room temperature for 2 hr. The organic solvents were removed in vacuo then water (100 mL) was added and the pH adjusted to ~6 by addition of 2M HC1. This was then extracted with EtOAc (3 x 100 mL) then the combined organics were washed with brine (100 mL), dried (Na2S04), filtered and evaporated to an orange gum. The crude product was purified by flash silica
chromatography, elution gradient 20 to 100% EtOAc in heptane. Pure fractions were evaporated to dryness to afford (E)-3-[4-[(65,,8R)-7-(2-fluoro-2-methyl-propyl)-8-methyl- 3,6,8,9-tetrahydropyrazolo[4,3-/]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoic acid
(1.964 g, 96%) as a yellow solid. XH NMR (500 MHz, CDC13, 27°C) 1.06 (3H, d), 1.2 - 1.29 (6H, m), 2.32 (IH, dd), 2.7 - 2.98 (2H, m), 3.35 (IH, dd), 3.62 - 3.83 (IH, m), 3.96 (3H, s), 5.44 (IH, s), 6.43 (IH, d), 6.78 (IH, d), 6.98 (IH, dd), 7.03 (IH, d), 7.08 (IH, d), 7.17 (IH, d), 7.73 (IH, d), 8.08 (IH, d). m/z (ES+), [M+H]+ = 438.
The methyl (E)-3-(4-((65,8R)-7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9- tetrahydropyrazolo[4,3-/]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoate used as starting material in the previous reaction was prepared as follows: MethvnE)-3-(4-((6^,8R)-7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9- tetrahvdropyrazolor4,3-niso uinolin-6-yl1-3-methoxy-phenyl1prop-2-enoate
Figure imgf000147_0001
Sodium nitrite (0.586 g, 8.50 mmol) in water (12 mL) was added dropwise to a stirred solution of methyl (E)-3-[4-[(15,,3R)-6-amino-2-(2-fluoro-2-methyl-propyl)-3,5-dimethyl- 3,4-dihydro-lH-isoquinolin-l-yl]-3-methoxy-phenyl]prop-2-enoate (3.743 g, 8.50 mmol) in propionic acid (50 mL) and the reaction was stirred for 60 minutes at -10 °C. Water was added (150 mL) and the mixture extracted with EtOAc (2 x 150 mL). The combined organics were washed with water (200 mL), saturated NaHC03 (3 x 200 mL), brine (200 mL), dried (Na2S04) then filtered and evaporated to give an orange gum. The crude product was purified by flash silica chromatography, elution gradient 0 to 50% EtOAc in heptane. Pure fractions were evaporated to dryness to afford methyl (E)-3-(4-((65',8R)-7- (2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3-/]isoquinolin-6-yl]-3- methoxy-phenyl]prop-2-enoate (2.83 g, 74%) as a pale orange solid. XH NMR (500 MHz,
CDC13, 27°C) 1.06 (3H, d), 1.25 (6H, dd), 2.32 (1H, dd), 2.6 - 2.99 (2H, m), 3.34 (1H, d), 3.67 - 3.79 (1H, m), 3.80 (3H, s), 3.95 (3H, s), 5.44 (1H, s), 6.39 (1H, d), 6.77 (1H, d), 6.95 (1H, d), 7.00 (1H, d), 7.05 (1H, s), 7.14 (1H, d), 7.64 (1H, d), 8.07 (1H, s). m/z (ES+), [M+H]+ = 452.
Example 13: (E)-3-r4-r(6^,8R)-7- ,2-difluoropropyl)-8-methyl-3,6,8,9- tetrahvdropyrazolor4,3-niso uinolin-6-yl1-3-methoxy-phenyl1prop-2-enoic acid
Figure imgf000148_0001
7.5M sodium hydroxide (0.456 mL, 3.42 mmol) was added to a solution of methyl (E)-3- [4-[(65',8R)-3-acetyl-7-(2,2-difluoropropyl)-8-methyl-8,9-dihydro-6H-pyrazolo[4,3- /]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoate (0.17 g, 0.34 mmol) in THF (2 mL) / methanol (2 mL). The reaction was stirred for room temperature for 2 hrs. The mixture was concentrated. Water (20 mL) was added, then the pH of the aqueous was adjusted to ~7 by addition of IN citric acid. The aqueous was extracted with EtOAc (3 x 100 mL). The combined organics were dried (MgS04) and concentrated. The crude product was purified by flash silica chromatography, elution gradient 0 to 5% MeOH in DCM. Pure fractions were evaporated to dryness to afford (E)-3-[4-[(65',8R)-7-(2,2-difluoropropyl)-8-methyl- 3,6,8,9-tetrahydropyrazolo[4,3-/]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoic acid
(0.108 g, 72%) as a cream solid. XH NMR (400 MHz, DMSO, 27 °C) 1.09 (3H, d), 1.60 (3H, t), 2.93 (IH, dd), 3.08 (IH, q), 3.25 (IH, dd), 3.52 (2H, dd), 3.99 (3H, s), 5.45 (IH, s), 6.60 (IH, d), 6.72 (IH, d), 6.81 (IH, d), 6.95 - 7.2 (IH, m), 7.28 (IH, d), 7.44 (IH, d), 7.59 (IH, d), 8.13 (IH, d), 12.50 (IH, s), 13.04 (IH, s). m/z: ES+ [M+H]+ 442.
The methyl (E)-3-[4-[(65,8R)-3-acetyl-7-(2,2-difluoropropyl)-8-methyl-8,9-dihydro-6H- pyrazolo[4,3-/]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoate used as starting material was prepared as follows: ,2-difluoropropyl trifluoromethanesulfonate
Figure imgf000149_0001
Trifluoromethanesulfonic anhydride (12.95 ml, 76.50 mmol), followed by 2,6-lutidine (9.33 ml, 80.1 mmol) were added to a solution of 2,2-difluoropropan-l-ol (7 g, 72.9 mmol) in DCM (140 ml) at 0 °C. The reaction mixture was stirred at 0 °C for 45 mins and at room temperature for 30 mins, then washed with 2N HC1 (3 x 150 ml) and saturated NaHC03 solution (2 x 100 ml). The organic phase was then dried over Na2S04 and concentrated to give 2,2-difluoropropyl trifluoromethanesulfonate (14.30 g, 86%) as a brown oil. XH NMR (400 MHz, CDC13, 22 °C) 1.76 (3H, t), 4.51 (2H, t).
(R)-N-(l-(3-bromo-2-methylphenyl)propan-2-yl)-2,2-difluoropropan-l-amine
Figure imgf000149_0002
Method A
2,2-difluoropropyl trifluoromethanesulfonate (8.64 g, 37.9 mmol) was added to a solution of (R)-l-(3-bromo-2-methylphenyl)propan-2-amine (7.2 g, 31.56 mmol) and DIPEA (10.91 ml, 63.12 mmol) in 1,4-dioxane (68.0 ml). The reaction was stirred at 85 °C for 21 hours (during this time a further 5 g of 2,2-difluoropropyl trifluoromethanesulfonate was added). After cooling, the reaction was diluted with DCM (200 mL) and washed with water (200 mL). The organic phase was dried (MgS04) and concentrated. The crude product was purified by flash silica chromatography, elution gradient 10 to 70% EtOAc in heptane. Pure fractions were evaporated to dryness to afford (R)-iV-(l-(3-bromo-2- methylphenyl)propan-2-yl)-2,2-difluoropropan- l -amine (5.64 g, 58%) as a yellow oil. Method B
(R)-l-(3-bromo-2-methylphenyl)propan-2-amine (7.00 g, 30.7 mmol) and 2,2- difluoropropyl trifluoromethanesulfonate (7.65 g, 33.53 mmol) were dissolved in 1,4- dioxane (50 mL). N-ethyl-iV-isopropylpropan-2-amine (9.35 mL, 53.7 mmol) was added and the reaction mixture was heated at 90 °C for 2.5 hours under nitrogen. After cooling, the solution was diluted with DCM (50 mL) and washed with water (2 x 75 mL). The aqueous phase was extracted with DCM (2 x 30 mL) and the combined organic phases were dried over MgS04 and concentrated. The crude product was purified by flash silica chromatography, elution gradient 0-40% EtOAc in heptane. Pure fractions were evaporated to dryness to afford (R)-N-(l-(3-bromo-2-methylphenyl)propan-2-yl)-2,2-difluoropropan- 1-amine (6.85 g, 73%) as a yellowish oil.
XH NMR (500 MHz, CDC13, 27 °C) 1.06 (3H, d), 1.58 (3H, t), 2.40 (3H, s), 2.62 (IH, dd), 2.79 - 3.03 (4H, m), 6.97 (IH, t), 7.07 (IH, dd), 7.43 (IH, dd). NH not observed.
(R)-3-(2-((2,2-difluoropropyl)amino)propyl)-2-methylaniline
Degassed toluene (70 mL) was added to (R)-N-(l-(3-bromo-2-methylphenyl)propan-2-yl)- 2,2-difluoropropan- 1 -amine (5.64 g, 18.42 mmol) followed by the addition of
diphenylmethanimine (3.09 ml, 18.42 mmol), sodium iert-butoxide (2.66 g, 27.63 mmol), 2,2'-bis(diphenylphosphino)- l,l'-binaphthalene (0.344 g, 0.55 mmol), and
tris(dibenzylideneacetone)dipalladium(0) (0.253 g, 0.28 mmol). The reaction was stirred at 90 °C for 3 hours under nitrogen. After cooling, the volatiles were removed under vacuum and the residue was dissolved in DCM (30 mL). IN HC1 (30 mL) was added and the biphasic mixture was stirred vigorously for 1 hour. The layers were separated and the aqueous layer was extracted with DCM. The aqueous layer was then basified by addition of 2N NaOH solution (20 mL), and extracted with EtOAc (3 x 150 mL). The combined basic extracts were dried (MgS04) and evaporated to dryness to afford (R)-3-(2-((2,2- difluoropropyl)-amino)propyl)-2-methylaniline (4.46 g, 100%) as a yellow oil. XH NMR
(500 MHz, CDC , 30 °C) 1.06 (3H, d), 1.58 (4H, t), 2.11 (3H, s), 2.59 (IH, dd), 2.77 (IH, dd), 2.82 - 2.96 (3H, m), 3.58 (2H, s), 6.59 (2H, t), 6.95 (IH, t). m/z: ES+ [M+H]+ 243. MethvnE)-3 4-r(1^ R)-6-amino-2-(2,2-difluoropropyl)-3,5-dimethyl-3,4-dihvdro- lH-iso uinolin-l-yl1-3-methoxy-phenyl1prop-2-enoate
Figure imgf000151_0001
Method A
Methyl (E)-3-(4-formyl-3-methoxy-phenyl)prop-2-enoate (8.23 g, 37.4 mmol) was added to a solution of (R)-3-(2-((2,2-difluoropropyl)amino)propyl)-2-methylaniline (4.53 g, 18.70 mmol) in acetic acid (92 ml) and water (1.68 ml, 93.5 mmol). The reaction was heated to 100 °C for 4 hrs under nitrogen. The mixture was evaporated. The residue was basified with NaHC03 and extracted into EtOAc (3 x 100 mL). The combined organics were washed with brine, dried over MgS04, filtered and evaporated. The organic layer was then vigorously stirred with DCM (75 mL) and 1 N HC1 (75 mL) for 1 hr. The layers were separated and the organic layer was extracted with DCM (3 x 100 mL). The aqueous layer was then basified by addition of Na2C03 and extracted with EtOAc (3 x 100 mL). The combined organic extracts were dried (MgS04) and concentrated. The residue was purified by flash silica chromatography, elution gradient 10-60% EtOAc in heptane. Pure fractions were evaporated to dryness to afford methyl (E)-3-[4-[(15,,3R)-6-amino-2-(2,2- difluoropropyl)-3,5-dimethyl-3,4-dihydro- lH-isoquinolin- l-yl]-3-methoxy-phenyl]prop-2- enoate (2.503 g, 30%) as a white solid.
Method B
Methyl (E)-3-(4-formyl-3-methoxyphenyl)prop-2-enoate (7.63 g, 34.7 mmol) was added to a solution of (R)-3-(2-((2,2-difluoropropyl)amino)propyl)-2-methylaniline (4.00 g, 16.5 mmol) in acetic acid (75 mL) and water (1.487 mL, 82.54 mmol). The reaction mixture was heated to 60 °C for 2 hours then the temperature was raised to 80 °C and heated for 1 hour. The reaction mixture was evaporated to a dark brown oil that was dissolved in EtOAc (75 mL) and washed with sat. NaHC03 solution (3 x 75 mL). The combined aqueous layers were extracted with EtOAc (3 x 30 mL) and the combined organic layers dried (MgS04), filtered and the filtrate evaporated to dryness. The residue was dissolved in DCM (75 mL). 1M HC1 (25 mL) was added and the biphasic mixture was stirred vigorously for 1 hr. The layers were separated and the aqueous layer was extracted with DCM (2 x 50 ml). The aqueous phase was then basified by addition of Na2C03 and extracted with EtOAc (3 x 50 mL). The combined basic extracts were dried over MgS04 and concentrated. The crude material was purified by silica column chromatography eluting with 0-40% EtOAc/Heptane to afford methyl (E)-3-[4-[(15,3R)-6-amino-2-(2,2- difluoropropyl)-3,5-dimethyl-3,4-dihydro- lH-isoquinolin- l-yl]-3-methoxy-phenyl]prop-2- enoate (5.50 g, 75%) as an off-white foam.
¾ NMR (500 MHz, CDCb, 27 °C) 1.04 (3H, d), 1.54 (3H, t), 2.07 (3H, s), 2.42 - 2.64 (2H, m), 2.76 - 2.94 (2H, m), 3.39 - 3.48 (1H, m), 3.51 (2H, s), 3.80 (3H, s), 3.91 (3H, s), 5.33 (1H, s), 6.39 (1H, d), 6.46 (2H, s), 6.83 - 6.97 (2H, m), 7.02 (1H, d), 7.64 (1H, d). m/z ES+ [M+H]+ 445.
MethvnE)-3-r4-r(6^,8R)-3-acetyl-7- ,2-difluoropropyl)-8-methyl-8,9-dihvdro-6H-
Pyrazolor4,3-niso uinolin-6-yl1-3-methoxy-phenyl1prop-2-enoate
Figure imgf000152_0001
Acetic anhydride (1.331 ml, 14.08 mmol) was added to a solution of methyl (E)-3-[4- [(15,,3R)-6-amino-2-(2,2-difluoropropyl)-3,5-dimethyl-3,4-dihydro-lH-isoquinolin-l-yl]-3- methoxy-phenyl]prop-2-enoate (2.503 g, 5.63 mmol) and potassium acetate (1.382 g, 14.08 mmol) in chloroform (52.0 ml) under an atmosphere of nitrogen and the reaction was heated to 70 °C for 30 min. Isopentyl nitrite (3.03 mL, 22.52 mmol) and 18-crown-6 (0.372 g, 1.41 mmol) were added and the reaction was stirred at 70 °C for 18 hours. The reaction was diluted with DCM (100 mL) and washed with water (100 mL). The aqueous phase was extracted with DCM (100 mL) and the combined organic phases were dried (MgS04) and concentrated. The crude product was purified by flash silica chromatography, elution gradient 0 to 30% EtOAc in heptane. Pure fractions were evaporated to dryness to afford methyl (E)-3-[4-[(65,8R)-3-acetyl-7-(2,2-difluoropropyl)-8-methyl-8,9-dihydro-6H- pyrazolo[4,3-/]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoate (1.42 g, 51%) as a yellow foam. ¾ NMR (400 MHz, CDC13, 30 °C) 1.09 (3H, d), 1.48 (3H, t), 2.60 (1H, q), 2.77 (3H, s), 2.81 - 3.08 (2H, m), 3.31 (1H, dd), 3.54 - 3.71 (1H, m), 3.80 (3H, s), 3.94 (3H, s), 5.49 (1H, s), 6.40 (1H, d), 6.86 (1H, d), 6.94 (2H, t), 7.06 (1H, d), 7.54 - 7.71 (1H, m), 8.09 (1H, d), 8.13 (1H, d). m/z: ES+ [M+H]+ 498.
The gradient was increased from 30-100% EtOAc in heptane to yield the N-acetylated starting material methyl (E)-3-(4-((llS,,3R)-6-acetamido-2-(2,2-difluoropropyl)-3,5- dimethyl-3,4-dihydro- lH-isoquinolin- l-yl]-3-methoxy-phenyl]prop-2-enoate (0.24 g, 9%) as a brown foam. This could be recycled by resubmitting to the reaction conditions.
Alternative Preparation of Example 13: (E)-3-r4-r(6^,8R)-7-(2,2-difluoropropyl)-8- methyl-3,6,8,9-tetrahvdropyrazolor4,3- iiso uinolin-6-yl1-3-inethoxy-phenyl1prop-2- enoic acid
Figure imgf000153_0001
Sodium hydroxide (11.94 mL, 23.87 mmol) was added slowly to a solution of methyl (E)- 3-(4-((65',8R)-7-(2,2-difluoropropyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoate (2.175 g, 4.77 mmol) in methanol (15 mL) and THF (5 mL). The reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was diluted with water (15 ml) and adjusted to ~pH6 with 2M aq HC1. The mixture was extracted with EtOAc (3 x 25 mL), The combined organic phases were washed with brine (1 x 30 mL), dried (MgS04) and filtered, and the filtrate was evaporated. The residue was purified by column chromatography eluting with 0-100% EtOAc/Heptane to afford (E)-3-[4-[(65,8R)-7-(2,2-difluoropropyl)-8-methyl-3,6,8,9- tetrahydropyrazolo[4,3-/]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoic acid (1.850 g, 88%) as a fawn coloured foam. lH NMR (500 MHz, CDC13, 27 °C) 1.10 (3H, d), 1.52 (3H, t), 2.55 - 2.72 (1H, m), 2.82 - 3.08 (2H, m), 3.27 (1H, dd), 3.55 - 3.66 (1H, m), 3.96 (3H, s), 5.49 (1H, s), 6.43 (1H, d), 6.79 (1H, d), 6.89 (1H, d), 6.98 (1H, dd), 7.09 (1H, d), 7.19 (1H, d), 7.73 (1H, d), 8.08 (1H, d). ES+ [M+H]+ 442.
The methyl (E)-3-(4-((65,8R)-7-(2,2-difluoropropyl)-8-methyl-3,6,8,9- tetrahydropyrazolo[4,3-/]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoate used as starting material in the previous reaction was prepared as follows:
MethvnE)-3-(4-((6^,8R)-7-(2,2-difluoropropyl)-8-methyl-3,6,8,9- tetrahvdropyrazolor4,3-niso uinolin-6-yl1-3-methoxy-phenyl1prop-2-enoate
Figure imgf000154_0001
Sodium nitrite (0.031 g, 0.45 mmol) in water (1 mL) was added dropwise to a stirred solution of methyl (E)-3-[4-[(llS',3R)-6-amino-2-(2,2-difluoropropyl)-3,5-dimethyl-3,4- dihydro- lH-isoquinolin- l-yl]-3-methoxy-phenyl]prop-2-enoate (0.2 g, 0.45 mmol) in propionic acid (3 mL) and the reaction stirred for 30 minutes at -15 °C (MeOH/Ice). The reaction mixture was diluted with brine/water (1 : 1, v/v; 15 mL) and extracted with EtOAc (2 x 15 mL). The combined organic phases were washed with saturated Na2C03 solution (1 x 20 mL), brine (1 x 15 mL), dried (MgS04), filtered and the filtrate evaporated to a brown oil. The crude material was purified by silica column chromatography eluting with 0-50% EtOAc/Heptane to afford methyl (E)-3-(4-((65,8R)-7-(2,2-difluoropropyl)-8-methyl- 3,6,8,9-tetrahydropyrazolo[4,3-/]isoquinolin-6-yl]-3-methoxy-phenyl]prop-2-enoate (0.118 g, 58%) as a fawn-coloured solid. lH NMR (500 MHz, CDCI3, 27 °C) 1.10 (3H, d), 1.50 (3H, d), 2.57 - 2.69 (1H, m), 2.84 - 3.04 (2H, m), 3.26 (1H, dd), 3.57 - 3.64 (1H, m), 3.80 (3H, s), 3.95 (3H, s), 5.49 (IH, s), 6.40 (IH, d), 6.77 (IH, d), 6.87 (IH, d), 6.94 (IH, dd), 7.06 (IH, d), 7.18 (IH, d), 7.64 (IH, d), 8.07 (IH, d), 10.03 (IH, s).m/z: ES+ [M+H]+ 456.
Example 14: (E)-3-r3-fluoro-4-r(6^,8R)-7- -fluoro-2-methyl-propyl)-8-methyl-3,6,8,9- tetrahvdropyrazolor4,3-niso uinolin-6-yl1-5-methoxy-phenyl1prop-2-enoic acid
Figure imgf000155_0001
Sodium hydroxide (2.0M) (1.47 ml, 2.94 mmol) was added to methyl (E)-3-[3-fluoro-4- [(65,,8R)-7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3- /]isoquinolin-6-yl]-5-methoxy-phenyl]prop-2-enoate (138 mg, 0.29 mmol) in THF (2 ml) / MeOH (2 ml). The resulting solution was stirred at 20 °C for 2 hours. The crude product was purified by ion exchange chromatography, using an SCX column. The desired product was eluted from the column using 7M NIHb/MeOH and pure fractions were evaporated to dryness to afford (E)-3-[3-fluoro-4-[(65,,8R)-7-(2-fluoro-2-methyl-propyl)-8-methyl- 3,6,8,9-tetrahydropyrazolo[4,3-/]isoquinolin-6-yl]-5-methoxy-phenyl]prop-2-enoic acid (123 mg, 92%) as a pale yellow solid. lH NMR (500 MHz, DMSO, 27 °C) 0.94 (3H, d), 1.07 (6H, dd), 2.21 (IH, dd), 2.77 - 3.01 (2H, m), 3.25 - 3.36 (IH, m), 3.57 - 3.72 (IH, m), 3.85 (3H, s), 5.32 (IH, s), 6.47 - 6.66 (2H, m), 6.91 (IH, d), 7.05 - 7.23 (2H, m), 7.35 (IH, d), 8.04 (IH, d), 12.93 (IH, s). m/z: ES+ [M+H]+ 456.
The methyl (E)-3-[3-fluoro-4-[(65,8R)-7-(2-fluoro-2-methyl-propyl)-8-methyl-3,6,8,9- tetrahydropyrazolo[4,3-/]isoquinolin-6-yl]-5-methoxy-phenyl]prop-2-enoate used as starting material was prepared as follows: 4-bromo-2-fluoro-6-methoxybenzaldehyde
Figure imgf000156_0001
Sodium methanolate (2.90 g, 53.76 mmol) was added to a solution of 4-bromo-2,6- dif uorobenzaldehyde (9.90 g, 44.8 mmol) in methanol (90 mL) and the reaction was heated to reflux for 5 hours. After cooling, the volatiles were evaporated. The residue was dissolved in DCM (300 mL) and washed with IN HC1 (300 mL) solution and brine. The organic phase was dried and evaporated, then the crude product was purified by flash silica chromatography, elution gradient 0 to 100% EtOAc in heptane. Pure fractions were evaporated to dryness to afford 4-bromo-2-f uoro-6-methoxybenzaldehyde (6.79 g, 65%) as colourless solid. lH NMR (500 MHz, CDC13, 27 °C) 3.94 (3H, s), 6.90 - 7.00 (2H, m), 10.38 (1H, s).
Methyl (E)-3-(3-fluoro-4-formyl-5-methoxy-phenyl)prop-2-enoate
Figure imgf000156_0002
Pd- 118 (0.369 g, 0.57 mmol) was added to a degassed solution of 4-bromo-2-fluoro-6- methoxybenzaldehyde (6.60 g, 28.3 mmol), triethylamine (5.79 mL, 42.48 mmol) and methyl acrylate (3.83 mL, 42.48 mmol) in DMA (76 mL) and the reaction was heated to 100 °C for 6 hours. After cooling, the reaction was diluted with EtOAc (250 mL) and washed with water (25 mL). The organic layer was washed with brine (250 mL), then dried and evaporated. The crude product was purified by flash silica chromatography, elution gradient 0 to 50% EtOAc in heptane. Pure fractions were evaporated to dryness to afford methyl (E)-3-(3-fluoro-4-formyl-5-methoxy-phenyl)prop-2-enoate (6.55 g, 97%) as a yellow solid. lH NMR (500 MHz, DMSO, 27 °C) 3.74 (3H, s), 3.95 (3H, s), 6.92 (1H, d), 7.33 (1H, d), 7.42 (1H, s), 7.64 (1H, d), 10.26 (1H, s). m/z (ES+), [M+H]+ = 239. MethvnE)-3-r4-r(l^,3R)-6-amino-2-(2-fluoro-2-methyl-propyl)-3,5-dimethyl-3,4- dihvdro-lH-iso uinolin-l-yl1-3-fluoro-5-methoxy-phenyl1prop-2-enoate
Figure imgf000157_0001
Methyl (E)-3-(3-fluoro-4-formyl-5-methoxy-phenyl)prop-2-enoate (0.610 g, 2.56 mmol) and (R)-3-(2-((2-fluoro-2-methylpropyl)amino)propyl)-2-methylaniline (0.305 g, 1.28 mmol) were dissolved in acetic acid (6.28 ml). Water (0.115 ml, 6.40 mmol) was added and the reaction mixture was stirred at 100 °C for 2 hours. The mixture was evaporated and the residue was neutralised by dissolving in EtOAc (100 mL) and washing with sat.
NaHCCb solution (3 x 100 mL). The combined aqueous layers were extracted with EtOAc (100 mL) and the combined organic layers were evaporated to dryness. The residue was dissolved in DCM (25 mL) then IN HCl (25 mL) was added and the biphasic mixture was stirred vigorously for 1 hr. The layers were separated and the aqueous was extracted with DCM (25 mL). The aqueous was then basified by addition of Na2C03 and extracted with EtOAc (2 x 25 mL). The combined basic extracts were dried over MgS04, concentrated and the crude product was purified by flash silica chromatography, elution gradient 0 to 60% EtOAc in heptane. Pure fractions were evaporated to dryness to afford methyl (E)-3-
[4-[(llS',3R)-6-amino-2-(2-fluoro-2-methyl-propyl)-3,5-dimethyl-3,4-dihydro-lH- isoquinolin-l-yl]-3-fluoro-5-methoxy-phenyl]prop-2-enoate (0.185 g, 32%) as a yellow solid. XH NMR (500 MHz, CDC13, 27 °C) 0.98 (3H, d), 1.11 (3H, s), 1.15 (3H, s), 2.08 (3H, s), 2.20 (1H, dd), 2.59 (1H, dd), 2.86 (1H, t), 3.09 (1H, dd), 3.38 - 3.51 (2H, m), 3.65 (1H, s), 3.80 (6H, s), 5.28 (1H, s), 6.28 - 6.47 (3H, m), 6.72 (1H, d), 6.80 (1H, d), 7.56 (1H, d). m/z: ES+ [M+H]+ 459. MethvnE)-3-r3-fluoro-4-r(6^,8R)-7- -fluoro-2-methyl-propyl)-8-methyl-3,6,8,9- tetrahvdropyrazolor4,3-niso uinolin-6-yl1-5-methoxy-phenyl1prop-2-enoate
Figure imgf000158_0001
Sodium nitrite (0.055 g, 0.79 mmol) in water (2 mL) was added dropwise to a stirred solution of methyl (E)-3-[4-[(llS,,3R)-6-amino-2-(2-fluoro-2-methyl-propyl)-3,5-dimethyl- 3,4-dihydro-lH-isoquinolin-l-yl]-3-fluoro-5-methoxy-phenyl]prop-2-enoate (0.363 g, 0.79 mmol) in acetic acid (8 mL) and the reaction stirred for 30 minutes at 20 °C. Water (20 mL) was added and the mixture extracted with EtOAc (2 x 20 ml), combined organics were washed with sat. NaHC03 (20 mL), brine (20 mL), dried (Na2S04), filtered and evaporated to give an orange gum. The crude product was purified by flash silica chromatography, elution gradient 0 to 50% EtOAc in heptane. Pure fractions were evaporated to dryness to afford methyl (E)-3-[3-fluoro-4-[(65',8R)-7-(2-fluoro-2-methyl- propyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3-/]isoquinolin-6-yl]-5-methoxy- phenyl]prop-2-enoate (0.136 g, 37%) as an off white solid. XH NMR (500 MHz, CDC13, 27 °C) 1.03 (3H, d), 1.12 (3H, d), 1.16 (3H, d), 2.26 (1H, dd), 2.82 - 3.04 (2H, m), 3.30 - 3.56 (1H, m), 3.68 - 4.03 (7H, m), 5.42 (1H, s), 6.38 (1H, d), 6.59 - 6.91 (3H, m), 7.11 (1H, d), 7.57 (1H, d), 8.07 (1H, d), 9.94 (1H, s). m/z: ES+ [M+H]+ 470.
Example 15: (E)-3-r4-r(6^,8R)-7- ,2-difluoropropyl)-8-methyl-3,6,8,9- tetrahvdropyrazolor4,3-niso uinolin-6-yl1-3-fluoro-5-methoxy-phenyl1prop-2-enoic acid
Figure imgf000158_0002
Sodium hydroxide (2.0M) (2.112 mL, 4.22 mmol) was added to methyl (E)-3-[4-[(65,8R)- 7-(2,2-dif uoropropyl)-8-methyl-3,6,8,9-tetrahydropyrazolo[4,3-/]isoquinolin-6-yl]-3- fluoro-5-methoxy-phenyl]prop-2-enoate (0.2 g, 0.42 mmol) in THF (3 mL) / MeOH (3 mL). The resulting solution was stirred at 20 °C for 2 hours. The crude product was purified by ion exchange chromatography, using an SCX column. The desired product was eluted from the column using 7M NH3/MeOH and pure fractions were evaporated to dryness to afford (E)-3-[4-[(65,8R)-7-(2,2-difluoropropyl)-8-methyl-3,6,8,9- tetrahydropyrazolo[4,3-/]isoquinolin-6-yl]-3-fluoro-5-methoxy-phenyl]prop-2-enoic acid (0.176 g, 91 ) as a pale yellow solid. lH NMR (500 MHz, DMSO, 27 °C) 0.99 (3H, d), 1.32 (3H, t), 2.92 (1H, dd), 3.04 (1H, dd), 3.16 (3H, s), 3.25 - 3.33 (1H, m), 3.53 (1H, d), 3.80 - 3.97 (2H, m), 5.38 (1H, s), 6.57 (1H, d), 6.59 (1H, d), 6.89 (1H, d), 7.15 (1H, s), 7.17 (1H, s), 7.28 (1H, d), 8.04 (1H, d), 12.99 (1H, s). m/z: ES+ [M+H]+ 460.
The methyl (E)-3-[4-[(65',8R)-7-(2,2-difluoropropyl)-8-methyl-3,6,8,9-tetrahydropyrazolo- [4,3-/]isoquinolin-6-yl]-3-fluoro-5-methoxy-phenyl]prop-2-enoate used as starting material was prepared as follows:
MethvnE)-3 4-r(1^ R)-6-amino-2-(2,2-difluoropropyl)-3,5-dimethyl-3,4-dihvdro- lH-iso uinolin-l-yl1-3-fluoro-5-methoxy-phenyl1prop-2-enoate
Figure imgf000159_0001
Methyl (E)-3-(3-fluoro-4-formyl-5-methoxy-phenyl)prop-2-enoate (0.983 g, 4.13 mmol) and (R)-3-(2-((2,2-difluoropropyl)amino)propyl)-2-methylaniline (0.50 g, 2.06 mmol) were dissolved in acetic acid (10.13 mL). Water (0.186 ml, 10.32 mmol) was added and the reaction mixture was stirred at 100 °C for 2 hours. The mixture was evaporated and the residue was neutralised by dissolving in EtOAc (100 mL) and washing with sat. NaHC03 solution (3 x 100 mL). The combined aqueous layers were extracted with EtOAc (100 mL) and the combined organic layers were evaporated to dryness. The residue was dissolved in DCM (25 mL) then IN HC1 (25 mL) was added and the biphasic mixture was stirred vigorously for 16 hours. The layers were separated and the aqueous layer was extracted with DCM. The aqueous layer was then basified by addition of Na2C03 and extracted with EtOAc (2 x 25 mL). The combined basic extracts were dried over MgS04 and
concentrated. The crude product was purified by flash silica chromatography, elution gradient 0 to 60% EtOAc in heptane. Pure fractions were evaporated to dryness to afford methyl (E)-3-[4-[(llS',3R)-6-amino-2-(2,2-difluoropropyl)-3,5-dimethyl-3,4-dihydro-lH- isoquinolin-l-yl]-3-fluoro-5-methoxy-phenyl]prop-2-enoate (0.080 g, 8%) as a yellow solid. XH NMR (500 MHz, CDC13, 27 °C) 1.01 (3H, d), 1.28 - 1.42 (3H, m), 2.06 (3H, d), 2.37 - 2.69 (2H, m), 2.86 - 3.14 (2H, m), 3.36 - 3.63 (3H, m), 3.74 - 3.95 (6H, m), 5.33 (1H, s), 6.26 - 6.45 (3H, m), 6.73 (1H, d), 6.77 - 6.88 (1H, m), 7.57 (1H, d). m/z: ES+ [M+H]+ 463.
MethvnE)-3-r4-r(6^,8R)-7-(2,2-difluoropropyl)-8-methyl-3,6,8,9-tetrahvdropyrazolo- r4,3-niso uinolin-6-yl1-3-fluoro-5-methoxy-phenyl1prop-2-enoate
Figure imgf000160_0001
Sodium nitrite (0.052 g, 0.76 mmol) in water (2 mL) was added dropwise to a stirred solution of methyl (E)-3-[4-[(llS',3R)-6-amino-2-(2,2-difluoropropyl)-3,5-dimethyl-3,4- dihydro-lH-isoquinolin-l-yl]-3-fluoro-5-methoxy-phenyl]prop-2-enoate (0.351 g, 0.76 mmol) in acetic acid (8 mL) and the reaction stirred for 30 minutes at 20 °C. Water (20 mL) was added and the mixture extracted with EtOAc (2 x 20 mL), combined organics washed with sat. NaHC03 (20 mL), brine (20 mL), dried (Na2S04), filtered and evaporated to give an orange gum. The crude product was purified by flash silica chromatography, elution gradient 0 to 50% EtOAc in heptane. Pure fractions were evaporated to dryness to afford methyl (E)-3-[4-[(65,8R)-7-(2,2-difluoropropyl)-8-methyl-3,6,8,9- tetrahydropyrazolo-[4,3-/]isoquinolin-6-yl]-3-fluoro-5-methoxy-phenyl]prop-2-enoate (0.164 g, 46%) as an off white solid. XH NMR (500 MHz, CDCb, 27 °C) 1.07 (3H, d), 1.35 (3H, t), 2.39 - 2.65 (IH, m), 2.93 (IH, dd), 3.06 (IH, dt), 3.32 - 3.56 (IH, m), 3.60 - 3.75 (IH, m), 3.77 - 3.98 (6H, m), 5.48 (IH, s), 6.39 (IH, d), 6.65 - 6.92 (3H, m), 7.05 - 7.17 (IH, m), 7.58 (IH, d), 8.07 (IH, d), 10.00 (IH, s). m/z: ES+ [M+H]+ 474.

Claims

1. A compound of Formula (I):
Figure imgf000162_0001
(I)
or a pharmaceutically acceptable salt thereof, wherein:
R1 is fluoro or methoxy;
R2, R3 and R4 are each independently hydrogen or fluoro;
R5 is hydrogen or methyl;
R6 is hydrogen or methyl;
R7 is methyl, CHF2 or cyclopropyl;
R8 is hydrogen or methyl;
R9 is hydrogen, fluoro, CH2OH, CH2OMe, CH2F or CHF2;
R10 is hydrogen, methyl or fluoro; and
R11 is hydrogen, methyl, fluoro or CH2F; or
R10 and R11 taken together with the carbon atom to which they are attached form a cyclopropyl ring or an oxetane ring.
2. A compound of Formula (I) or a pharmaceutically acceptable salt thereof, as claimed in claim 1, wherein the group -CH(R8)-C(R9)(R10)(Rn) in the compound of Formula (I) is selected from the group consisting of:
Figure imgf000163_0001
3. A compound of Formula (I), as claimed in claim 1 or claim 2, or a pharmaceutically acceptable salt thereof, for use as a medicament.
4. A compound of Formula (I), as claimed in claim 1 or claim 2, or a pharmaceutically acceptable salt thereof, for use in the prevention or treatment of cancer in a warm-blooded animal such as man.
5. A compound of Formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 or claim 2, for use in the treatment of breast or gynaecological cancer.
6. A method for the prevention or treatment of cancer in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of the Formula (I) as claimed in claim 1 or claim 2, or a
pharmaceutically acceptable salt thereof.
7. A pharmaceutical composition comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof, as claimed in claim 1 or claim 2, and a
pharmaceutically acceptable diluent or carrier.
8. A combination suitable for use in the treatment of cancer comprising a compound of Formula (I) as claimed in claim 1 or claim 2, or a pharmaceutically acceptable salt thereof, and another anti-tumour agent.
PCT/EP2017/059240 2016-04-20 2017-04-19 Indazole derivatives that down-regulate the estrogen receptor and possess anti-cancer activity WO2017182495A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201662325024P 2016-04-20 2016-04-20
US62/325,024 2016-04-20
US201762445436P 2017-01-12 2017-01-12
US62/445,436 2017-01-12

Publications (1)

Publication Number Publication Date
WO2017182495A1 true WO2017182495A1 (en) 2017-10-26

Family

ID=58549150

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2017/059240 WO2017182495A1 (en) 2016-04-20 2017-04-19 Indazole derivatives that down-regulate the estrogen receptor and possess anti-cancer activity

Country Status (1)

Country Link
WO (1) WO2017182495A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020058725A3 (en) * 2018-09-21 2020-04-30 Mexichem Fluor S.A. De C.V. Methods of preparing partially fluorinated esters
JP2020186250A (en) * 2016-10-24 2020-11-19 アストラゼネカ・アクチエボラーグAstrazeneca Aktiebolag 6,7,8,9-tetrahydro-3h-pyrazolo[4,3-f]isoquinoline derivatives useful in treatment of cancer
WO2023081857A1 (en) 2021-11-04 2023-05-11 Skyhawk Therapeutics, Inc. Condensed pyridazine amine derivatives treating sca3

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
WO2008023161A1 (en) 2006-08-23 2008-02-28 Kudos Pharmaceuticals Limited 2-methylmorpholine pyrido-, pyrazo- and pyrimido-pyrimidine derivatives as mtor inhibitors
WO2015092634A1 (en) * 2013-12-16 2015-06-25 Novartis Ag 1,2,3,4-tetrahydroisoquinoline compounds and compositions as selective estrogen receptor antagonists and degraders
WO2016202161A1 (en) * 2015-06-16 2016-12-22 江苏恒瑞医药股份有限公司 Piperidine derivative and preparation method and pharmaceutical use thereof

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4683202B1 (en) 1985-03-28 1990-11-27 Cetus Corp
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4683195B1 (en) 1986-01-30 1990-11-27 Cetus Corp
WO2008023161A1 (en) 2006-08-23 2008-02-28 Kudos Pharmaceuticals Limited 2-methylmorpholine pyrido-, pyrazo- and pyrimido-pyrimidine derivatives as mtor inhibitors
WO2015092634A1 (en) * 2013-12-16 2015-06-25 Novartis Ag 1,2,3,4-tetrahydroisoquinoline compounds and compositions as selective estrogen receptor antagonists and degraders
WO2016202161A1 (en) * 2015-06-16 2016-12-22 江苏恒瑞医药股份有限公司 Piperidine derivative and preparation method and pharmaceutical use thereof

Non-Patent Citations (41)

* Cited by examiner, † Cited by third party
Title
"Bioreversible Carriers in Drug Design", 1987, PERGAMON PRESS
"Cancer Genome Atlas Network", NATURE, vol. 490, 2012, pages 61 - 70
"Comprehensive Medicinal Chemistry", vol. 5, 1990, PERGAMON PRESS
"Design of Pro-drugs", 1985, ELSEVIER
"Laboratory Protocols for Mutation Detection", 1996, OXFORD UNIVERSITY PRESS
"Methods in Enzymology,", vol. 42, 1985, ACADEMIC PRESS, pages: 309 - 396
"Molecular Cloning: A Laboratory Manual", vol. 1-3, 2012, COLD SPRING HARBOR LABORATORY PRESS
"PCR", 1997, BIOS SCIENTIFIC PUBLISHERS LIMITED
ANDERSON SM, EXPERT REV MOL DIAGN, vol. 11, 2011, pages 635 - 642
CHEN ET AL., ANAL BIOCHEM, vol. 239, 1996, pages 61 - 9
DAHLMAN-WRIGHT ET AL., PHARMACOL. REV., vol. 58, 2006, pages 773 - 781
GIBBS ET AL., NUCL. ACIDS RES., vol. 17, 1989, pages 2437 - 248
H. BUNDGAARD ET AL., JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 77, 1988, pages 285
H. BUNDGAARD, ADVANCED DRUG DELIVERY REVIEWS, vol. 8, 1992, pages 1 - 38
H. BUNDGAARD: "A Textbook of Drug Design and Development", 1991, article "Design and Application of Pro-drugs", pages: 113 - 191
J. MED. CHEM, vol. 47, 2004, pages 6658 - 6661
KATO ET AL., SCIENCE, vol. 270, 1995, pages 1491 - 1494
KUSHNER ET AL., PURE APPLIED CHEMISTRY, vol. 75, 2003, pages 1757 - 1769
LANDEGREN, U. ET AL., SCIENCE, vol. 242, 1988, pages 229 - 237
LEWIS, R., GENETIC ENGINEERING NEWS, vol. 10, 1990, pages 54 - 55
LI, S. ET AL., CELL REP., vol. 4, 2013, pages 1116 - 1130
LONARD ET AL., MOL. CELL, vol. 5, 2000, pages 939 - 948
M. J. MCPHERSON; S. G. MAILER; R. BEYNON; C. HOWE: "PCR (Basics: From Background to Bench)", 15 October 2000, SPRINGER VERLAG
MARCELINO ET AL., BIOTECHNIQUES, vol. 26, 1999, pages 1134 - 1148
MEYERSON M. ET AL., NAT REV GENET., vol. 11, 2010, pages 685 - 696
MOK ET AL., GYNAECOLOGIC ONCOLOG, vol. 52, 1994, pages 247 - 252
N. KAKEYA ET AL., CHEM. PHARM. BULL., vol. 32, 1984, pages 692
NEWTON ET AL., NUCLEIC ACIDS RES., vol. 17, 1989, pages 2503 - 2516
NOLLAU ET AL., CLIN. CHEM., vol. 43, 1997, pages 1114 - 1120
PIKE ET AL., STRUCTURE, vol. 9, 2001, pages 145 - 153
PROSSNER, TIBTECH, vol. 11, 1993, pages 238
ROBINSON ET AL., NAT. GENETICS, vol. 45, 2013, pages 144601451
STENOIEN ET AL., MOL. CELL BIOL., vol. 21, 2001, pages 4404 - 4412
STERN ET AL., CRITICAL REVIEWS IN ONCOLOGY/HAEMATOLOGY, vol. 54, 2005, pages 1 - 29
T. HIGUCHI; V. STELLA: "Pro-Drugs as Novel Delivery Systems", vol. 14, A.C.S. SYMPOSIUM SERIES
T.W. GREEN: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY AND SONS
TOY ET AL., NAT. GENETICS, vol. 45, 2013, pages 1439 - 1445
TZUKERMAN ET AL., MOL. ENDOCRINOLOGY, vol. 8, 1994, pages 21 - 30
WAKELING ET AL., CANCER RES., vol. 51, 1991, pages 3867 - 3873
WARDELL ET AL., BIOCHEM. PHARM, vol. 82, 2011, pages 122 - 130
WARNER ET AL., STEROIDS, vol. 71, 2006, pages 91 - 95

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2020186250A (en) * 2016-10-24 2020-11-19 アストラゼネカ・アクチエボラーグAstrazeneca Aktiebolag 6,7,8,9-tetrahydro-3h-pyrazolo[4,3-f]isoquinoline derivatives useful in treatment of cancer
JP6993477B2 (en) 2016-10-24 2022-01-13 アストラゼネカ・アクチエボラーグ 6,7,8,9-Tetrahydro-3H-pyrazolo [4,3-f] isoquinoline derivative useful in the treatment of cancer
WO2020058725A3 (en) * 2018-09-21 2020-04-30 Mexichem Fluor S.A. De C.V. Methods of preparing partially fluorinated esters
WO2023081857A1 (en) 2021-11-04 2023-05-11 Skyhawk Therapeutics, Inc. Condensed pyridazine amine derivatives treating sca3
WO2023081858A1 (en) 2021-11-04 2023-05-11 Skyhawk Therapeutics, Inc. Condensed-pyrazine amine derivatives for treating sca3

Similar Documents

Publication Publication Date Title
US10961241B2 (en) Chemical compounds
US10130617B2 (en) Chemical compounds
US10125135B2 (en) Chemical compounds
US10149839B2 (en) Chemical compounds
US11046689B2 (en) Selective estrogen receptor down-regulators
WO2017182495A1 (en) Indazole derivatives that down-regulate the estrogen receptor and possess anti-cancer activity

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

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

Ref document number: 17718079

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 17718079

Country of ref document: EP

Kind code of ref document: A1