WO2023105008A1 - Small-molecule inhibitors of the frs2-fgfr interaction - Google Patents

Small-molecule inhibitors of the frs2-fgfr interaction Download PDF

Info

Publication number
WO2023105008A1
WO2023105008A1 PCT/EP2022/085077 EP2022085077W WO2023105008A1 WO 2023105008 A1 WO2023105008 A1 WO 2023105008A1 EP 2022085077 W EP2022085077 W EP 2022085077W WO 2023105008 A1 WO2023105008 A1 WO 2023105008A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
halogen
independently selected
compound
compound according
Prior art date
Application number
PCT/EP2022/085077
Other languages
French (fr)
Inventor
Karthiga KUMAR
Sally Oxenford
Original Assignee
Universität Zürich
Invasight Ag
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 Universität Zürich, Invasight Ag filed Critical Universität Zürich
Publication of WO2023105008A1 publication Critical patent/WO2023105008A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D221/00Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00
    • C07D221/02Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00 condensed with carbocyclic rings or ring systems
    • C07D221/04Ortho- or peri-condensed ring systems
    • C07D221/06Ring systems of three rings
    • C07D221/16Ring systems of three rings containing carbocyclic rings other than six-membered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis

Definitions

  • the present invention relates to small-molecule inhibitors of the FRS2-FGFR interaction.
  • the present invention also relates to the small-molecule inhibitors for use as a medicament and for use in cancer or metastasis treatment or prevention.
  • Metastasis the dissemination and growth of neoplastic cells in an organ distant from that in which they originated, causes as much as 90% of cancer-associated mortality.
  • Effective cancer therapy is largely dependent on the capability to prevent metastasis specifically and less toxic, targeted anti-metastatic therapies are urgently needed.
  • An important and fundamental cause of metastasis in the majority of all solid tumours is the deregulated motile behaviour of the cancer cells.
  • the microenvironment shapes cell behaviour and determines metastatic outcomes of tumours.
  • Kumar et al. (Cell Reports, 2018, vol. 23, issue 13, P3798- 3812) addressed how microenvironmental cues control tumour cell invasion in paediatric brain tumour, medulloblastoma (MB).
  • TGF-p regulates pro-migratory bFGF function in a context- dependent manner.
  • the non-canonical TGF-p pathway causes ROCK activation and cortical translocation of ERK1/2, which antagonizes FGFR signalling by inactivating FGFR substrate 2 (FRS2), and promotes a contractile, non-motile phenotype.
  • FRS2 FGF receptor
  • TGF-p counters inactivation of FRS2 and restores pro-migratory signalling.
  • the objective of the present invention is to provide means and methods to provide small-molecule inhibitors of the FRS2-FGFR interaction. This objective is attained by the subject-matter of the independent claims of the present specification. Summary of the Invention
  • a first aspect of the invention relates to a compound of a general formula (I): wherein
  • each other R 1 is independently selected from OR°, CN, halogen, NR N1 R N2 , SO 2 R S , NHSO 2 R S , SR S , COOR A ,
  • each other R 2 is independently selected from OR°, CN, halogen, NR N1 R N2 , SO 2 R S , NHSO 2 R S , SR S , COOR A ,
  • - n 0, 1 , 2, or 3;
  • R N1 and R N2 being independently selected from H, or unsubstituted or CN- and/or hydroxyl-substituted methyl;
  • R s , R A and R° being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl.
  • a second aspect of the invention relates to a compound according to the first aspect for use as a medicament.
  • a further aspect of the invention relates to a compound according to the first aspect for use in treatment or prevention of cancer, metastasis, or an FGFR-driven disease or for use as an angiogenesis antagonist.
  • FGFR fibroblast growth factor receptor
  • FRS2 FGFR substrate 2
  • FRS2 FGFR substrate 2
  • PTB phospho-tyrosine binding domain
  • FRS2 interacts with the FGFRs via the c-terminal phospho-tyrosine binding (PTB) domain and serves as a molecular hub by assembling both positive and negative signalling proteins to mediate important FGF-induced cellular functions. It transmits the signal from the FGFRs (outside of the cell) to the inside of the cell.
  • PTB phospho-tyrosine binding
  • Binding site 1 is not involved in FGFR binding and located below the interaction site of FGFR’s N-terminus with FRS2.
  • Binding site 2 is the extended surface area interacting with FGFR’s C-terminal end.
  • the mechanism of compound-target interaction, conformational change in the target domain and transmission blockade is unique and does not depend on receptor tyrosine kinase inhibition.
  • FRS2 does not have any shared protein domains with other adapter proteins.
  • the compounds also interfere specifically with those FGFR functions that are particularly relevant for tumorigenesis and tumor progression.
  • the compounds In contrast to existing FGFR targeting strategies, the compounds also interfere specifically with those FGFR functions that are particularly relevant for tumorigenesis and tumor progression, such as proliferation, migration and invasion and angiogenesis.
  • FRS2-FGFR interaction is altered in many types of cancer, for example in prostate cancer (Yang, F. et al. Cancer Res 73, 3716-3724, 2013, Liu J et al. Oncogene. 2016 Apr 7;35(14):1750-9), esophageal cancer (Nemoto, T., Ohashi, K., Akashi, T., Johnson, J. D. & Hirokawa, K. Pathobiology 65, 195-203, 1997), thyroid cancer (St Bernard, R. et al.
  • Ci-Cs alkyl in the context of the present specification signifies a saturated linear or branched hydrocarbon having 1 , 2, 3, 4, 5 or 6 carbon atoms, wherein one carbon-carbon bond may be unsaturated and/or one CH2 moiety may be exchanged for oxygen (ether bridge) or nitrogen (NH, or NR with R being methyl, ethyl, or propyl; amino bridge).
  • Ci-Ce alkyl include the examples given for C1-C4 alkyl above, and additionally 3-methylbut-2-enyl, 2-methylbut-3-enyl, 3-methylbut-3-enyl, n-pentyl, 2- methylbutyl, 3-methylbutyl, 1 , 1-dimethylpropyl, 1 ,2-dimethylpropyl, 1 ,2-dimethylpropyl, pent- 4-inyl, 3-methyl-2-pentyl, and 4-methyl-2-pentyl.
  • a Cs alkyl is a pentyl or cyclopentyl moiety and a Ce alkyl is a hexyl or cyclohexyl moiety.
  • C3-C7 cycloalkyl in the context of the present specification relates to a saturated hydrocarbon ring having 3, 4, 5, 6 or 7 carbon atoms, wherein in certain embodiments, one carbon-carbon bond may be unsaturated.
  • Non-limiting examples of a C3-C7 cycloalkyl moiety include cyclopropanyl (-C3H5), cyclobutanyl (-C4H7), cyclopentenyl (C5H9), and cyclohexenyl (CeHn) moieties.
  • a cycloalkyl is substituted by one Ci to C4 unsubstituted alkyl moiety.
  • a cycloalkyl is substituted by more than one Ci to C4 unsubstituted alkyl moieties.
  • carbocycle in the context of the present specification relates to a cyclic moiety composed of carbon and hydrogen atoms only.
  • An aromatic carbocycle is also named aryl.
  • a non-aromatic carbocycle is also named cycloalkyl.
  • heterocycle in the context of the present specification relates to a cyclic moiety, wherein at least one ring atom is replaced or several ring atoms are replaced by a nitrogen, oxygen and/or sulphur atom.
  • An aromatic heterocycle is also named heteroaryl.
  • a non- aromatic heterocycle is a cycloalkyl, wherein at least one ring atom is replaced or several ring atoms are replaced by a nitrogen, oxygen and/or sulphur atom.
  • heterobicycle in the context of the present specification relates to two directly connected cycles, wherein at least one ring atom is replaced or several ring atoms are replaced by a nitrogen, oxygen and/or sulphur atom.
  • a heterobicycle is composed of two heterocycles or of one heterocycle and one carbocycle.
  • unsubstituted C n alkyl when used herein in the narrowest sense relates to the moiety -C n H2n- if used as a bridge between moieties of the molecule, or -C n H2n+i if used in the context of a terminal moiety.
  • unsubstituted C n alkyl and substituted C n alkyl include a linear alkyl comprising or being linked to a cyclical structure, for example a cyclopropane, cyclobutane, cyclopentane or cyclohexane moiety, unsubstituted or substituted depending on the annotation or the context of mention, having linear alkyl substitutions.
  • the total number of carbon and -where appropriate- N, O or other hetero atom in the linear chain or cyclical structure adds up to n.
  • Me is methyl CH3
  • Et is ethyl -CH2CH3
  • Prop is propyl -(CH 2 ) 2 CH 3 (n-propyl, n-pr) or -CH(CH 3 ) 2 (iso-propyl, i-pr), but is butyl -C 4 H 9 , -(CH 2 )3CH 3 , -CHCH3CH2CH3, -CH 2 CH(CH 3 )2 or -C(CH 3 )3.
  • substituted alkyl in its broadest sense refers to an alkyl as defined above in the broadest sense, which is covalently linked to an atom that is not carbon or hydrogen, particularly to an atom selected from N, O, F, B, Si, P, S, Cl, Br and I, which itself may be -if applicable- linked to one or several other atoms of this group, or to hydrogen, or to an unsaturated or saturated hydrocarbon (alkyl or aryl in their broadest sense).
  • substituted alkyl refers to an alkyl as defined above in the broadest sense that is substituted in one or several carbon atoms by groups selected from amine NH2, alkylamine NHR, imide NH, alkylimide NR, amino(carboxyalkyl) NHCOR or NRCOR, hydroxyl OH, oxyalkyl OR, oxy(carboxyalkyl) OCOR, carbonyl O and its ketal or acetal (OR)2, nitril ON, isonitril NO, cyanate ONO, isocyanate NCO, thiocyanate CNS, isothiocyanate NOS, fluoride F, choride Cl, bromide Br, iodide I, phosphonate PO3H2, PO3R2, phosphate OPO3H2 and OPO3R2, sulfhydryl SH, suflalkyl SR, sulfoxide SOR, sulfonyl SO2
  • hydroxyl substituted group refers to a group that is modified by one or several hydroxyl groups OH.
  • amino substituted group refers to a group that is modified by one or several amino groups NH2.
  • carboxyl substituted group refers to a group that is modified by one or several carboxyl groups COOH.
  • Non-limiting examples of amino-substituted alkyl include -CH2NH2, -CH2NHMe, -CH2NHEt, -CH2CH2NH2, -CH 2 CH 2 NHMe, -CH 2 CH 2 NHEt, -(CH 2 )3NH 2 , -(CH 2 )3NHMe, -(CH 2 )3NHEt, -CH 2 CH(NH 2 )CH 3 , -CH 2 CH(NHMe)CH 3 , -CH 2 CH(NHEt)CH 3 , -(CH 2 )3CH 2 NH 2 , -(CH 2 )3CH 2 NHMe, -(CH 2 )3CH 2 NHEt, -CH(CH 2 NH2)CH 2 CH3, -CH(CH 2 NHMe)CH 2 CH 3 , -CH(CH 2 NHEt)CH 2 CH 3 , -CH 2 CH(CH 2 NH2)CH3, -CH 2 CH(CH 2 N
  • Non-limiting examples of hydroxy-substituted alkyl include -CH2OH, -(CH2)2OH, -(CH2)sOH, -CH 2 CH(OH)CH 3 , -(CH 2 ) 4 OH, -CH(CH 2 OH)CH 2 CH 3 , -CH 2 CH(CH 2 OH)CH 3 , -CH(OH)(CH 2 ) 2 OH, -CH 2 CH(OH)CH 2 OH, -CH 2 CH(OH)(CH 2 )2OH and -CH 2 CH(CH 2 OH) 2 for terminal moieties and -CHOH-, -CH2CHOH-, -CH 2 CH(OH)CH 2 -, -(CH 2 ) 2 CHOHCH2-, - CH(CH 2 OH)CH 2 CH 2 -, -CH 2 CH(CH 2 OH)CH 2 -, -CH(OH)(CH 2 CHOH, -CH(OH)(CH 2 CHOH, -CH(
  • sulfoxyl substituted group refers to a group that is modified by one or several sulfoxyl groups -SO2R, or derivatives thereof, with R having the meaning as laid out in the preceding paragraph and different from other meanings assigned to R in the body of this specification.
  • amine substituted group refers to a group that is modified by one or several amine groups -NHR or -NR2, or derivatives thereof, with R having the meaning as laid out in the preceding paragraph and different from other meanings assigned to R in the body of this specification.
  • carbonyl substituted group refers to a group that is modified by one or several carbonyl groups -COR, or derivatives thereof, with R having the meaning as laid out in the preceding paragraph and different from other meanings assigned to R in the body of this specification.
  • An ester refers to a group that is modified by one or several ester groups -CO2R, with R being defined further in the description.
  • An amide refers to a group that is modified by one or several amide groups -CON HR, with R being defined further in the description.
  • halogen-substituted group refers to a group that is modified by one or several halogen atoms selected (independently) from F, Cl, Br, I.
  • fluoro substituted alkyl refers to an alkyl according to the above definition that is modified by one or several fluoride groups F.
  • fluoro-substituted alkyl include -CH 2 F, -CHF 2 , -CF 3 , -(CH 2 ) 2 F, -(CHF) 2 H, -(CHF) 2 F, -C 2 F 5 , -(CH 2 ) 3 F, -(CHF) 3 H, - (CHF) 3 F, -C 3 F 7 , -(CH 2 ) 4 F, -(CHF) 4 H, -(CHF) 4 F and -C 4 F 9 .
  • Non-limiting examples of hydroxyl- and fluoro-substituted alkyl include -CHFCH2OH, - CF2CH2OH, -(CHF) 2 CH 2 OH, -(CF 2 )2CH 2 OH, -(CHF) 3 CH 2 OH, -(CF 2 ) 3 CH 2 OH, -(CH 2 ) 3 OH, -CF 2 CH(OH)CH 3 , -CF 2 CH(OH)CF 3 , -CF(CH 2 OH)CHFCH 3 , and -CF(CH 2 OH)CHFCF 3 .
  • aryl in the context of the present specification signifies a cyclic aromatic C5-C10 hydrocarbon.
  • aryl include, without being restricted to, phenyl and naphthyl.
  • a heteroaryl is an aryl that comprises one or several nitrogen, oxygen and/or sulphur atoms.
  • heteroaryl include, without being restricted to, pyrrole, thiophene, furan, imidazole, pyrazole, thiazole, oxazole, pyridine, pyrimidine, thiazin, quinoline, benzofuran and indole.
  • An aryl or a heteroaryl in the context of the specification additionally may be substituted by one or more alkyl groups.
  • the term pharmaceutical composition refers to a compound of the invention, or a pharmaceutically acceptable salt thereof, together with at least one pharmaceutically acceptable carrier.
  • the pharmaceutical composition according to the invention is provided in a form suitable for topical, parenteral or injectable administration.
  • the term pharmaceutically acceptable earner includes any solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (for example, antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drugs, drug stabilizers, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, and the like and combinations thereof, as would be known to those skilled in the art (see, for example, Remington: the Science and Practice of Pharmacy, ISBN 0857110624).
  • treating or treatment of any disease or disorder refers in one embodiment, to ameliorating the disease or disorder (e.g. slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms thereof).
  • treating or treatment refers to alleviating or ameliorating at least one physical parameter including those which may not be discernible by the patient.
  • treating or treatment refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both.
  • metastasis in the context of the present specification relates to the dissemination and growth of neoplastic cells outside the original tumor bed in the same organ or in an organ distant from that in which they originated.
  • the treatment or prevention with the disclosed compounds is employed for metastasis associated with aberrant FGFR signalling.
  • the compounds of the invention specifically reduce the motile behaviour of metastatic cells and reduce dissemination.
  • the compounds of the invention are employed for prevention or treatment of motility and dissemination of cancerous cells.
  • FGFR1 is frequently amplified in 20 - 25% of squamous non-small cell lung cancer (Weiss, J. et al., Science Translational Medicine (2010) doi:10.1126/scitranslmed.3001451) and 15% breast cancer (Andre, F. et al., Clin Cancer Res., 15, 441-452 (2009)) and mutated in 18% of midline gliomas (Di Stefano, A. L.
  • FGFR2 is mainly activated by gene fusions in intrahepatic cholangiocarcinomas (iCCA, 15%) and mutations in 10% of endometrial tumors have also been described (Konecny, G. E. et al., The Lancet Oncology 16, 686-694 (2015); Verlingue, L. et al., European Journal of Cancer 87, 122-130 (2017).).
  • FGFR3 is affected by mutations in urothelial carcinomas (up to 20% in the metastatic setting 7 ); gene fusions (mainly FGFR3-TACC3) are present in glioblastomas and gliomas (3-6% (Di Stefano, A.
  • FGFR1-4 signal via Fibroblast Growth Factor Receptor Substrate 2 (FRS2)-dependent (RAS/MAPK and PI3K/AKT) and FRS2-independent (PLC-y, JAK-STAT) pathways (Turner, N.
  • FRS2 Fibroblast Growth Factor Receptor Substrate 2
  • FRS2 interacts with FGFRs via its phosphotyrosine binding domain (PTB) (Gotoh, N., Cancer Science 99, 1319-1325 (2008)) and increased expression or activation for FRS2 is involved in tumorigenesis of several tumor entities (Zhang, K. et al., Cancer Research 73, 1298-1307 (2013); Li, J.-L. & Luo, European review for medical and pharmacological sciences 24, 97-108 (2020); Wu, S. et al., Nature Communications 1-12 (2019) doi:10.1038/s41467-019-08576-5; Liu, J.
  • PTB phosphotyrosine binding domain
  • FGFR-driven invasiveness depends on FRS2 (Huynh, H. et al., Hepatology 69, 943-958 (2019)).
  • FGF ligands of FGFRs are highly expressed in skeletal muscle (Pedersen, B. K. & Febbraio, M. A., Nature Reviews Endocrinology vol. 8457- 465 (2012)), bone (Su, N., Du, X. L. & Chen, Frontiers in Bioscience vol. 132842-2865 (2008)) and in CSF-secreting choroid plexus (Greenwood, S.
  • Resistance to FGFR inhibitors can evolve similarly as to other RTK inhibitors, either by the formation of gatekeeper mutations in the catalytic domain or the activation of alternative RTKs, which enable bypass mechanism for downstream signaling activation (Yamaoka, T., et al., Int. J. Mol. Sci. 19, 1-35 (2016)).
  • Such mutations in FGFRs can occur in the ATP binding cleft and may create a steric conflict to limit drug-binding efficacy. Examples include FGFR3_V555M, FGFR1_V561 and FGFR2_V564, which induce resistance to FGFR inhibitors in vitro (Chell, V. et al., Oncogene 32, 3059-3070 (2013); Byron S. A. et a!., Neoplasia 15, 975-988 (2013)).
  • the inventors’ approach to target the non-enzymatically active FGFR adaptor protein FRS2 could prevent the evolution of FGFR gatekeeper mutations or help overcoming the resistance of gatekeeper FGFR-driven tumors by blocking signaling downstream of the RTK.
  • Targeting FRS2 is likely also effective against tumors driven by the FGFR3-TACC3 fusion, where FRS2 is phosphorylated and transmits signaling to the oncogenic MAP kinase pathway (Chew, N. J. et al., Cell Communication and Signaling 18, 1-17 (2020)).
  • a first aspect of the invention relates to a compound of a general formula (I) or (lb): wherein
  • each other R 1 is independently selected from OR°, CN, halogen, NR N1 R N2 , SO 2 R S , NHSO 2 R S , SR S , COOR A ,
  • each other R 2 is independently selected from OR°, CN, halogen, NR N1 R N2 , SO 2 R S , NHSO 2 R S , SR S , COOR A ,
  • - n 0, 1 , 2, or 3;
  • R N1 and R N2 being independently selected from H, or unsubstituted or CN- and/or hydroxyl-substituted methyl;
  • R s , R A and R° being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl;
  • - o is 0, 1 , or 2, particularly o is 0 or 1 , more particularly o is 0;
  • - X is N or CH, particularly X is CH.
  • the compound is of formula (I).
  • each R 1 is independently selected from OR°, CN, halogen, SO 2 R S , NHSO 2 R S , SR S , COOR A
  • n is 1, 2, or 3. In certain embodiments, n is 2.
  • n is 0 or 1. In certain embodiments, m is 1.
  • the compound is described by a general formula (II): wherein R 1 , R 2 , n and m have the same definition as above.
  • the compound is described by a general formula (lib): wherein X, o, R 1 , R 2 , n and m have the same definition as above.
  • the compound is described by a general formula (III): wherein R 1 , R 2 , and n have the same definition as above.
  • the compound is described by a general formula (lllb): wherein X, o, R 1 , R 2 and n have the same definition as above.
  • the compound is described by a general formula (IV): wherein R 1 , R 2 , and n have the same definition as above.
  • the compound is described by a general formula (IVb): wherein X, o, R 1 , R 2 and n have the same definition as above. In certain embodiments, the compound is described by a general formula (V): wherein R 1 and n have the same definition as above.
  • the compound is described by a general formula (Vb): wherein X, o, R 1 , R 2 and n have the same definition as above.
  • the compound is described by a general formula (VI):
  • the compound is described by a general formula (Vlb):
  • R 3 is selected from the group of H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl;
  • one R 1 is CN, and the other R 1 is/are independently selected from CN, halogen, OR°, COOR A , SR S , NHSO2R s , SC>2R S with R s , R A , and R° being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl.
  • the other R 1 is/are halogen.
  • the other R 1 is/are is Cl.
  • each R 1 is independently selected from halogen, CN, OR°, COOR A , SR S , NHSO2R s , SO2R s with R s , R A , and R° being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl.
  • R 1 is halogen.
  • R 1 is Cl.
  • one R 1 is CN, and the other R 1 is/are independently selected from SR S , NHSO2R s , SC>2R s with each R s being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl.
  • each R 1 is independently selected from CN, SR s , NHSC>2R S , SC>2R S with each R s being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl.
  • each R 1 is independently selected from halogen, CN, OR°, COOR A , SR S , NHSO 2 R s , SO 2 R S with R s , R A , and R° being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl.
  • R 1 is halogen.
  • R 1 is Cl.
  • one R 1 is CN, and the other R 1 is/are independently selected from SR s , NHSO2R s , SO2R s with each R s being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl.
  • each R 1 is independently selected from CN, SR S , NHSO2R s , SO2R s with each R s being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl.
  • R 1 is CN. In certain embodiments, R 1 is SR s . In certain embodiments, R 1 is NHSO2R S . In certain embodiments, R 1 is SO2R s . In certain embodiments, R 1 is NHSR S .
  • each R 2 is independently selected from halogen, CN, OR°, SR S , NHSO2R S , SO2R S with R s , R A , and R° being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl.
  • R 2 is halogen.
  • R 2 is Cl.
  • each R 2 is independently selected from CN, SR s , NHSO2R s , SO2R s with each R s being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl.
  • R 2 is CN. In certain embodiments, R 2 is SR S . In certain embodiments, R 2 is NHSO2R S . In certain embodiments, R 2 is SO2R s In certain embodiments, R 2 is NHSR s .
  • one R 1 is CN, and the other R 1 is/are independently selected from NHSR s , SR S , NHSO2R s , SO2R s with each R s being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl- substituted methyl.
  • one R 1 is CN and one other R 1 is selected from NHSR s , SR S , NHSO2R S , SO2R S
  • n is 2, and one R 1 is CN and the other one is NHSO2R s .
  • a second aspect of the invention relates to a compound according to the first aspect for use as a medicament.
  • a third aspect of the invention relates to a compound according to the first aspect for use in treatment or prevention of cancer.
  • a fourth aspect of the invention relates to a compound according to the first aspect for use in treatment or prevention of metastasis.
  • a fifth aspect of the invention relates to a compound according to the first aspect for use as an angiogenesis antagonist, particularly an angiogenesis antagonist in treatment or prevention of cancer.
  • said cancer or said metastasis arises from a cancer selected from ovarian cancer, urothelial carcinomas, breast cancer, endometrial adenocarcinnoma, cholagiocarcinoma, non-small cell lung cancer, head and cancer, sarcoma, melanoma, glioma, pancreatic cancer, renal cell carcinoma, neuroendocrine carcinoma, leiomyosarcoma, liposarcoma, bladder cancer, medulloblastoma, pediatric brain tumours, multiple myeloma, colorectal cancer and gastric cancer.
  • a cancer selected from ovarian cancer, urothelial carcinomas, breast cancer, endometrial adenocarcinnoma, cholagiocarcinoma, non-small cell lung cancer, head and cancer, sarcoma, melanoma, glioma, pancreatic cancer, renal cell carcinoma, neuroendocrine carcinoma, leiomyosarcoma, liposarcoma,
  • said cancer or said metastasis arises from a cancer selected from ovarian cancer, urothelial carcinomas, breast cancer, endometrial adenocarcinnoma, non-small cell lung cancer, head and cancer, sarcoma, melanoma, glioma, bladder cancer, medulloblastoma, pediatric brain tumours, multiple myeloma, colorectal cancer and gastric cancer.
  • a cancer selected from ovarian cancer, urothelial carcinomas, breast cancer, endometrial adenocarcinnoma, non-small cell lung cancer, head and cancer, sarcoma, melanoma, glioma, bladder cancer, medulloblastoma, pediatric brain tumours, multiple myeloma, colorectal cancer and gastric cancer.
  • said cancer is selected from bladder cancer, multiple myeloma, gastric cancer, pediatric brain tumour, medulloblastoma, glioblastoma, ependymoma, colorectal cancer and sarcoma. In certain embodiments, said cancer is selected from bladder cancer, pediatric brain tumour, medulloblastoma, multiple myeloma, colorectal cancer and gastric cancer.
  • a sixth aspect of the invention relates to a compound according to the first aspect for use in prevention or treatment of an FGFR-driven disease.
  • a method or treating cancer or metastasis in a patient in need thereof comprising administering to the patient a compound according to the above description.
  • a dosage form for the prevention or treatment of cancer comprising a non-agonist ligand or antisense molecule according to any of the above aspects or embodiments of the invention.
  • a pharmaceutically acceptable salt of said drug may be present as a pharmaceutically acceptable salt of said drug.
  • Pharmaceutically acceptable salts comprise the ionized drug and an oppositely charged counterion.
  • Non-limiting examples of pharmaceutically acceptable anionic salt forms include acetate, benzoate, besylate, bitatrate, bromide, carbonate, chloride, citrate, edetate, edisylate, embonate, estolate, fumarate, gluceptate, gluconate, hydrobromide, hydrochloride, iodide, lactate, lactobionate, malate, maleate, mandelate, mesylate, methyl bromide, methyl sulfate, mucate, napsylate, nitrate, pamoate, phosphate, diphosphate, salicylate, disalicylate, stearate, succinate, sulfate, tartrate, tosylate, triethiodide and valerate.
  • Non-limiting examples of pharmaceutically acceptable cationic salt forms include aluminium, benzathine, calcium, ethylene diamine, lysine, magnesium, meglumine, potassium, procaine, sodium, trometh
  • Dosage forms may be for enteral administration, such as nasal, buccal, rectal, transdermal or oral administration, or as an inhalation form or suppository.
  • parenteral administration may be used, such as subcutaneous, intravenous, intrahepatic or intramuscular injection forms.
  • a pharmaceutically acceptable carrier and/or excipient may be present.
  • compositions comprising a compound of the present invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the composition comprises at least two pharmaceutically acceptable carriers, such as those described herein.
  • the compound of the present invention is typically formulated into pharmaceutical dosage forms to provide an easily controllable dosage of the drug and to give the patient an elegant and easily handleable product.
  • the pharmaceutical composition is formulated in a way that is suitable for topical administration such as aqueous solutions, suspensions, ointments, creams, gels or sprayable formulations, e.g., for delivery by aerosol or the like, comprising the active ingredient together with one or more of solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives that are known to those skilled in the art.
  • the pharmaceutical composition can be formulated for oral administration, parenteral administration, or rectal administration.
  • the pharmaceutical compositions of the present invention can be made up in a solid form (including without limitation capsules, tablets, pills, granules, powders or suppositories), or in a liquid form (including without limitation solutions, suspensions or emulsions).
  • the dosage regimen for the compounds of the present invention will vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the species, age, sex, health, medical condition, and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; the route of administration, the renal and hepatic function of the patient, and the effect desired.
  • the compounds of the invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three, or four times daily.
  • the pharmaceutical composition or combination of the present invention can be in unit dosage of about 1-1000 mg of active ingredient(s) for a subject of about 50-70 kg.
  • the therapeutically effective dosage of a compound, the pharmaceutical composition, or the combinations thereof, is dependent on the species of the subject, the body weight, age and individual condition, the disorder or disease or the severity thereof being treated. A physician, clinician or veterinarian of ordinary skill can readily determine the effective amount of each of the active ingredients necessary to prevent, treat or inhibit the progress of the disorder or disease.
  • compositions of the present invention can be subjected to conventional pharmaceutical operations such as sterilization and/or can contain conventional inert diluents, lubricating agents, or buffering agents, as well as adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers and buffers, etc. They may be produced by standard processes, for instance by conventional mixing, granulating, dissolving or lyophilizing processes. Many such procedures and methods for preparing pharmaceutical compositions are known in the art, see for example L. Lachman et al. The Theory and Practice of Industrial Pharmacy, 4th Ed, 2013 (ISBN 8123922892).
  • the invention further encompasses, as an additional aspect, the use of a compound as identified herein, or its pharmaceutically acceptable salt, as specified in detail above, for use in a method of manufacture of a medicament for the treatment or prevention of cancer or metastasis.
  • the invention encompasses methods of treatment of a patient having been diagnosed with a disease associated with cancer or metastasis.
  • This method entails administering to the patient an effective amount of a compound as identified herein, or its pharmaceutically acceptable salt, as specified in detail herein.
  • Fig. 1 shows the efficacy of the efficacy of F3.18, F18.2, F18.7, F18.8, F18.9 at 3 different concentrations - 1 .M, 5 .M and 10 .M.
  • Fig. 2 shows the efficacy of F3.18, F18.2, F18.7, F18.8, F18.9 at 10 .M
  • Fig. 3 shows the binding affinities and dissociation constant (Kd) of F3.18, F18.2,
  • Nano diffraction scanning fluorimetry nanoDSF
  • Microscale thermophoresis MST are biophysical assays used to assess the binding of the compounds to the target protein. Any temperature shift above 1.5 degree Celsius is considered as indication for significant binding
  • Fig. 4 shows the effective inhibitory concentration of compound F3.18
  • Fig. 5 A and B show the Biochemical specificity of F3.18, F18.2, F18.7, F18.8, F18.9 determining the ability of the compounds to inhibit FGF signalling pathway without affecting other signalling pathways.
  • Fig 5a shows for 1) the Control - DAOY l-A-EGFP cells unstimulated, serum starved overnight and then lysed,
  • bFGF (100ng/ml) - Overnight serum starved DAOY l-A-EGFP cells stimulated with bFGF for 10 minutes and then lysed and 3) F3.18 (10 .M) - Overnight serum starved DAOY l-A-EGFP cells treated with F3.18 for four hours, cells stimulated with bFGF for 10 minutes and then lysed.
  • Fig 5B shows for 1) the Control - DAOY LA-EGFP cells unstimulated, serum starved overnight and then lysed, 2) bFGF (100ng/ml) - Overnight serum starved DAOY l_A-EGFP cells stimulated with bFGF for 10 minutes and then lysed and
  • F18.8 (10 .M) Overnight serum starved DAOY l-A-EGFP cells treated with F18.8 for four hours, cells stimulated with bFGF for 10 minutes and then lysed
  • F18.9 (10 ,M) Overnight serum starved DAOY LA-EGFP cells treated with F18.9 for four hours, cells stimulated with bFGF for 10 minutes and then lysed.
  • Fig. 6 shows the structure of the compounds F3.18, F18.2, F18.7, F18.8 and F18.9.
  • Binding site 1 is not involved in FGFR binding and located below the interaction site of FGFR’s N-terminus with FRS2.
  • Binding site 2 is the extended surface area interacting with FGFR’s C-terminal end.
  • Fig. 9 Spheroid invasion assay using DAOY cells stimulated with bFGF +/- BGJ398 or F18.7 to determine the EC50 of F18.7.
  • Fig. 10 Spheroid invasion assay using DAOY cells stimulated with bFGF +/- BGJ398 or F3.18 series compounds.
  • Fig. 11 Spheroid invasion assay using DAOY cells stimulated with bFGF +/- BGJ398 or F18.7 series compounds.
  • Fig. 12 Cell titer gio assay performed with DAOY cells treated with BGJ398 or FF18.7.
  • Fig. 13 Cell titer gio assay performed with AGS cells treated with BGJ398 or F3.118.
  • Fig. 14 Cell titer gio assay performed with DMS114 cells treated with BGJ398 or
  • Fig. 15 Cell titer gio assay performed with HCT 116 cells treated with BGJ398 or F3.18.
  • Fig. 16 Cell titer gio assay performed with M059K cells treated with BGJ398 or F3.18.
  • Fig. 17 Cell titer gio assay performed with RT112 cells treated with BGJ398 or F3.18.
  • Fig. 18 Cell titer gio assay performed with SNLI16 cells treated with BGJ398 or F3.18.
  • Fig. 19 Cell titer gio assay performed with SKOV3 cells treated with BGJ398 or F3.18.
  • Fig. 20 Table showing the in vitro absorption, distribution, metabolism, elimination and toxicity (ADMET) properties of F3.18.
  • Efflux ration represents the permeability of F3.18,
  • Semi-thermodynamic solubility shows the solubility of F3.18 in aqueous solutions.
  • Intrinsic clearance and t1/2 shows the metabolic stability of F3.18 MTT shows the toxicity of F3.18 and potency shows the efficacy of F3.18.
  • Fig. 21 In vivo pharmacokinetics, 3 mice/treatment, serum concentration of compounds in pM.
  • Fig. 22 Immunoblots using various FGFR-driven cell lines treated with BGJ398 or F3.18 showing the effect of the treatment on the downstream effectors of FGF signalling.
  • Fig. 23 Spheroid invasion assay using DAOY cells stimulated with bFGF +/- BGJ398 or F18.1 , F18.4 and F18.10 series compounds. F3.18 is used as positive control.
  • Fig. 24 shows chemical structure and properties of F18.7 (F2i) AND F1873 (Non-nitro analog of F2i).
  • Fig. 25 shows in-vitro properties of F18.7 and F1873.
  • Fig. 26 shows in-vitro ADME of F18.7 and F1873.
  • Fig. 27 shows tested compounds.
  • Fig. 28 shows functional assay - test for inhibition of cell invasion by the compounds using ACINDA via Spheroid invasion assay (SIA).
  • Fig. 29 shows functional assay - test for inhibition of cell invasion by the compounds using ACINDA via Spheroid invasion assay (SIA).
  • Control unstimulated cells in the form of spheroids.
  • bFGF Maximum invasion possible -s activated by the FGF signaling.
  • BGJ398 - FGFR kinase inhibitor for comparison.
  • F18.7 - F2i (lead) parent compound.
  • F1873 non-nitro analogue of F2i (lead).
  • Fig. 30 shows nanoDSF - assay.
  • a +ve or -ve temperature shift of 1.5°C is a significant binder to target protein.
  • Fig. 31 shows determination of lipophilicity - LogD.
  • LogD lipophilicity
  • LogD lipophilicity
  • Optimal logD range: 1 to 3. Most compounds showed acceptable logD values within the range of 2 to 4. Some compounds showed high logD values of >4. Few compounds were weakly ionizable and not clearly detected (ND) by LCMS.
  • Fig. 32 shows determination of aqueous solubility.
  • Fig. 33 shows determination of Kd (cell free IC50) and ADME properties.
  • Fig. 34 shows tested compounds.
  • Fig. 35 shows the percentage of inhibition in spheroid invasion assay.
  • Control is unstimulated cells in the form of spheroids.
  • BGJ398 is an FGFR inhibitor for comparison.
  • Fig. 36 shows a nanoDSF biophysical assay. A +ve or -ve temperature shift of 1.5°C is a significant binder to target protein.
  • Fig. 37 shows KD, IC50, and EC50 data for selected compounds. KD was determined using microscale thermophoresis. IC50 was determined using cell titer gio assay. EC50 was determined using spheroid invasion assay.
  • the inventors designed an inhibitor of FRS2-FGFR interaction by screening a large library of fragments of small molecules.
  • the inventors identified F3.18 as a putative small molecule inhibitor of FRS2-FGFR interaction.
  • the inventors confirmed the binding of F3.18 to FRS2 using biophysical assays - nanoDSF, MST and NMR analysis.
  • the inventors evaluated the efficacy of F3.18 in inhibiting cancer cell invasion and proliferation using FGFR-driven cancer cell models. Results from the spheroid invasion assay and cell titer gio assay show that F3.18 effectively inhibits cancer cell invasion and proliferation in all the FGFR-driven cancer cell lines tested. To test the effect of F3.18 on FGF signaling pathway, we used immunoblotting.
  • F3.18 inhibits the FGF signal transduction by inhibiting the phosphorylation of the downstream effectors of FGF signaling pathway.
  • the inventors used in vitro ADMET studies and in vivo PK studies to determine the ‘drug-like’ properties of F3.18. Results from these assays demonstrate that F3.18 has good permeability, moderate solubility and intrinsic clearance, low toxicities, and high potency.
  • the in vivo PK studies show that F3.18 is well- tolerated in mice and could be safely administered via oral and intravenous route to living organisms for the treatment of FGFR-driven diseases.
  • SIA Spheroid invasion assay
  • aCDc automated cell dissemination counter
  • Purified FRS2 protein tagged with 6X Histidine residues and Guanine nucleotide-binding protein subunit beta (GB1) was diluted in the protein buffer (100mM sodium phosphate, 50mM NaCI, 0.5mM EDTA, 50mM arginine, 1 mM TCEP, pH 7.0) to final concentration of 30 .M.
  • the compounds were dissolved in 100% at 50 or 100mM and further diluted to 1 mM with a final concentration of 100% DMSO.
  • Compound and protein were mixed at 1 :1 ration yielding final concentrations of 15 .M and 500 .M for the compounds.
  • the mixture was incubated at room temperature for 15 minutes before measurement. The measurement was performed on a Prometheus system in high sensitivity capillaries. Samples were subjected to a temperature gradient of 20 to 95°C with 1°C/min intervals.
  • MST Microscale thermophoresis
  • Purified FRS2 protein tagged with 6X Histidine residues and Guanine nucleotide-binding protein subunit beta (GB1) was labelled with 2 nd generation BLUE-NHS dye.
  • the protein was labelled at a final concentration of 20 .M with 60 .M dye.
  • the labelling was performed in the protein buffer without arginine supplementation.
  • Arginine was re-buffered to protein’s buffer post-labelling.
  • the compounds were dissolved in 100% at 50 or 100mM and further diluted to 1mM with a final concentration of 100% DMSO. The compounds were then diluted. In a 1 :1 serial dilution from 1 mM to 61.04nM in protein buffer supplemented with 10% DMSO.
  • Radioimmunoprecipitation assay (RIPA) buffer.
  • RIPA buffer lysates were resolved by SDS- PAGE and transferred to a nitrocellulose membrane using a transfer apparatus according to the manufacturer’s instructions (Bio-Rad).
  • Membranes were probed with primary antibodies against phospho-FRS2, FRS2, ERK1/2, phospho-ERK1/2, AKT, phosphor-AKT, phospho- PKC and tubulin.
  • HRP-linked secondary antibodies (1 :5000) were used to detect the primary antibodies. Chemiluminescence detection was performed using ChemiDoc Touch Gel and Western Blot imaging system (BioRad). Cell titer gio assay
  • the metabolic activity and the proliferation of the cells were determined using the Cell Titer gio assay from Promega according to the manufacturer’s instructions.
  • 250 cells/1 OO l/per well (for up to 72 h incubation) were seeded in Greiner Bio-One p-clear 384 well plates (655090, Greiner Bio-One) and incubated overnight at 37°C.
  • the old media was then replaced with fresh serum-free media and the cells were treated with BGJ398 or F3.18 till the desired time point.
  • 10 pl of the Cell titer gio reagent was added to each well (final concentration of cell titer gio reagent per well is 1:10) and incubated at 37° C for 30 minutes. The luminescence was then measured with a signal integration time of 0.5 to 1 second per well.
  • RIPA buffer FGFR-driven cell lysates were resolved by SDS-PAGE and transferred to a nitrocellulose membrane using a transfer apparatus according to the manufacturer’s instructions (Bio-Rad).
  • Membranes were probed with primary antibodies against phospho- FRS2, FRS2, ERK1/2, phospho-ERK1/2, AKT, phospho-AKT, phospho-PKC and tubulin.
  • HRP-linked secondary antibodies (1:5000) were used to detect the primary antibodies.
  • Chemiluminescence detection was performed using ChemiDoc Touch Gel and Western Blot imaging system (BioRad). Integrated density of Immuno-reactive bands was quantified using Adobe Photoshop CS5.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to small-molecule inhibitors of the FRS2-FGFR interaction. The present invention relates the small-molecule inhibitors for use as a medicament and for use in cancer or metastasis treatment or prevention.

Description

Small-molecule inhibitors of the FRS2-FGFR interaction
This application claims the right of priority of European Patent Application EP21213222.9 filed 08.12.2021 , incorporated by reference herein.
Field
The present invention relates to small-molecule inhibitors of the FRS2-FGFR interaction. The present invention also relates to the small-molecule inhibitors for use as a medicament and for use in cancer or metastasis treatment or prevention.
Background of the Invention
Metastasis, the dissemination and growth of neoplastic cells in an organ distant from that in which they originated, causes as much as 90% of cancer-associated mortality. Effective cancer therapy is largely dependent on the capability to prevent metastasis specifically and less toxic, targeted anti-metastatic therapies are urgently needed. An important and fundamental cause of metastasis in the majority of all solid tumours is the deregulated motile behaviour of the cancer cells. The microenvironment shapes cell behaviour and determines metastatic outcomes of tumours. Kumar et al. (Cell Reports, 2018, vol. 23, issue 13, P3798- 3812) addressed how microenvironmental cues control tumour cell invasion in paediatric brain tumour, medulloblastoma (MB). They show that bFGF promotes MB tumour cell invasion through FGF receptor (FGFR) in vitro and that blockade of FGFR represses brain tissue infiltration in vivo. TGF-p regulates pro-migratory bFGF function in a context- dependent manner. Under low bFGF, the non-canonical TGF-p pathway causes ROCK activation and cortical translocation of ERK1/2, which antagonizes FGFR signalling by inactivating FGFR substrate 2 (FRS2), and promotes a contractile, non-motile phenotype. Under high bFGF, negative-feedback regulation of FRS2 by bFGF-induced ERK1/2 causes repression of the FGFR pathway. Under these conditions, TGF-p counters inactivation of FRS2 and restores pro-migratory signalling. These findings pinpoint coincidence detection of bFGF and TGF-p signalling by FRS2 as a mechanism that controls tumour cell invasion. Thus, targeting FRS2 represents an emerging strategy to abrogate aberrant FGFR signalling.
Based on the above-mentioned state of the art, the objective of the present invention is to provide means and methods to provide small-molecule inhibitors of the FRS2-FGFR interaction. This objective is attained by the subject-matter of the independent claims of the present specification. Summary of the Invention
A first aspect of the invention relates to a compound of a general formula (I):
Figure imgf000003_0001
wherein
- one R1 or one R2 is CN;
- each other R1 is independently selected from OR°, CN, halogen, NRN1RN2, SO2RS, NHSO2RS, SRS, COORA,
- each other R2 is independently selected from OR°, CN, halogen, NRN1RN2, SO2RS, NHSO2RS, SRS, COORA,
- n is 0, 1 , 2, or 3;
- m is 0, 1, 2, 3, 4, or 5; with the proviso that n+m 0;
- with RN1 and RN2 being independently selected from H, or unsubstituted or CN- and/or hydroxyl-substituted methyl;
- with Rs, RA and R° being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl.
A second aspect of the invention relates to a compound according to the first aspect for use as a medicament.
A further aspect of the invention relates to a compound according to the first aspect for use in treatment or prevention of cancer, metastasis, or an FGFR-driven disease or for use as an angiogenesis antagonist.
The transmission of signals from activated fibroblast growth factor receptor (FGFR) tyrosine kinases promotes oncogenic functions in tumor cells, including proliferation, survival and cell migration and invasion. The interruption of signal transmission from activated FGFRs to downstream signal transduction cascades by kinase inhibitors designed against FGFRs is an established means of attenuating these oncogenic functions. In addition to aberrant activation of FGFRs in numerous malignancies, FGFR activation is also observed to act as an evasion mechanism in cancers of patients subjected to targeted therapies with kinase inhibitors, which results in tumor re-growth and progression. The small molecule compounds described in this application will prevent signal transmission from activated FGFRs to downstream effector molecules, specifically to the mitogen activated protein kinases (MAPKs), a key driver of tumorigenesis.
The compounds bind to FRS2. FGFR substrate 2 (FRS2) is a key adaptor protein that is largely specific to FGF signalling pathway. It is an exclusive downstream effector of FGFRs. FRS2 interacts with the FGFRs via the c-terminal phospho-tyrosine binding (PTB) domain and serves as a molecular hub by assembling both positive and negative signalling proteins to mediate important FGF-induced cellular functions. It transmits the signal from the FGFRs (outside of the cell) to the inside of the cell. Hence, targeting FRS2, which is very upstream of the FGF signalling pathway, effectively shuts down the downstream effectors, especially MAPKs of FGFR signaling.
The compounds specifically bind to the phosphotyrosine binding (PTB) domain of the FRS2 protein (Fig. 7). Compound binding induces a conformational shift in the PTB domain that prevents FGFR-induced signal transmission through FRS2. Two potential binding sites were initially selected: Binding site 1 is not involved in FGFR binding and located below the interaction site of FGFR’s N-terminus with FRS2. Binding site 2 is the extended surface area interacting with FGFR’s C-terminal end.
The mechanism of compound-target interaction, conformational change in the target domain and transmission blockade is unique and does not depend on receptor tyrosine kinase inhibition. In addition, unlike FGFRs, FRS2 does not have any shared protein domains with other adapter proteins. Thus, compared to the existing kinase inhibitors, much less off-target activity is expected. In contrast to existing FGFR targeting strategies, the compounds also interfere specifically with those FGFR functions that are particularly relevant for tumorigenesis and tumor progression.
In contrast to existing FGFR targeting strategies, the compounds also interfere specifically with those FGFR functions that are particularly relevant for tumorigenesis and tumor progression, such as proliferation, migration and invasion and angiogenesis. There is evidence of the FRS2-FGFR interaction being altered in many types of cancer, for example in prostate cancer (Yang, F. et al. Cancer Res 73, 3716-3724, 2013, Liu J et al. Oncogene. 2016 Apr 7;35(14):1750-9), esophageal cancer (Nemoto, T., Ohashi, K., Akashi, T., Johnson, J. D. & Hirokawa, K. Pathobiology 65, 195-203, 1997), thyroid cancer (St Bernard, R. et al. Endocrinology 146, 1145-1153, 2005), hepatocellular carcinoma (Zheng, N., Wei, W. Y. & Wang, Z. W. Transl Cancer Res 5, 1-6, 2016, Matsuki M et al. Cancer Med. 2018 Jun;7(6):2641-2653), testicular cancer (Jiang, X. et al. J Diabetes Res, 2013), medulloblastoma (Santhana Kumar, K. et al. Cell Rep 23, 3798-3812 e3798, 2018), rhabdomyosarcoma (Goldstein, M., Meller, I. & Orr-Urtreger, A. Gene Chromosome Cane 46, 1028-1038, 2007), gastric cancer (Kunii, K. et al. Cancer Res 68, 3549-3549, 2008), pulmonary pleomorphic carcinoma (Lee, S. et al. J Cancer Res Clin 137, 1203-1211 , 2011), breast cancer (Penaultllorca, F. et al. Int J Cancer 61 , 170-176, 1995), non-small cell lung cancer (Dutt, A. et al. Pios One 6, 2011), Liposarcoma (Zhang, K. Q. et al. Cancer Res 73, 1298-1307, 2013), cervical cancer (Jang, J. H., Shin, K. H. & Park, J. G. Cancer Res 61 , 3541-3543, 2001), colorectal cancer (Sato, T. et al. Oncol Rep 21 , 211-216, 2009), melanoma (Becker, D., Lee, P. L., Rodeck, U. & Herlyn, M. Oncogene 7, 2303-2313, 1992), multiple myeloma (Kalff, A. & Spencer, A. Blood Cancer J, 2, 2012), endometrial cancer (Konecny, G. E. et al. Mol Cancer Ther 12, 632-642, 2013), bladder cancer (Cappellen, D. et al. Nat Genet 23, 18-20, 1999, Wu S et al. Nat Commun. 2019 Feb 12;10(1):720), glioblastoma (Morrison, R. S. et al. Cancer Res 54, 2794-2799, 1994), squamous cell carcinoma of the lung (Weiss, J. et al. Sci Transl Med 4, 2012), ovarian cancer (Cole, C. et al. Cancer Biol Ther 10, 2010), head and neck cancer (Koole, K. et al. Virchows Arch 469, S31-S31 , 2016), and pancreatic cancer (Ishiwata, T. et al. Am J Pathol 180, 1928-1941 , 2012).
Detailed Description of the Invention
Terms and definitions
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art (e.g., in cell culture, molecular genetics, nucleic acid chemistry, hybridization techniques and biochemistry). Standard techniques are used for molecular, genetic and biochemical methods (see generally, Sambrook et al., Molecular Cloning: A Laboratory Manual, 2d ed. (1989) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. and Ausubel et al., Short Protocols in Molecular Biology (1999) 4th Ed, John Wiley & Sons, Inc.) and chemical methods.
A Ci-Cs alkyl in the context of the present specification signifies a saturated linear or branched hydrocarbon having 1 , 2, 3, 4, 5 or 6 carbon atoms, wherein one carbon-carbon bond may be unsaturated and/or one CH2 moiety may be exchanged for oxygen (ether bridge) or nitrogen (NH, or NR with R being methyl, ethyl, or propyl; amino bridge). Nonlimiting examples for a Ci-Ce alkyl include the examples given for C1-C4 alkyl above, and additionally 3-methylbut-2-enyl, 2-methylbut-3-enyl, 3-methylbut-3-enyl, n-pentyl, 2- methylbutyl, 3-methylbutyl, 1 , 1-dimethylpropyl, 1 ,2-dimethylpropyl, 1 ,2-dimethylpropyl, pent- 4-inyl, 3-methyl-2-pentyl, and 4-methyl-2-pentyl. In certain embodiments, a Cs alkyl is a pentyl or cyclopentyl moiety and a Ce alkyl is a hexyl or cyclohexyl moiety.
The term C3-C7 cycloalkyl in the context of the present specification relates to a saturated hydrocarbon ring having 3, 4, 5, 6 or 7 carbon atoms, wherein in certain embodiments, one carbon-carbon bond may be unsaturated. Non-limiting examples of a C3-C7 cycloalkyl moiety include cyclopropanyl (-C3H5), cyclobutanyl (-C4H7), cyclopentenyl (C5H9), and cyclohexenyl (CeHn) moieties. In certain embodiments, a cycloalkyl is substituted by one Ci to C4 unsubstituted alkyl moiety. In certain embodiments, a cycloalkyl is substituted by more than one Ci to C4 unsubstituted alkyl moieties.
The term carbocycle in the context of the present specification relates to a cyclic moiety composed of carbon and hydrogen atoms only. An aromatic carbocycle is also named aryl. A non-aromatic carbocycle is also named cycloalkyl.
The term heterocycle in the context of the present specification relates to a cyclic moiety, wherein at least one ring atom is replaced or several ring atoms are replaced by a nitrogen, oxygen and/or sulphur atom. An aromatic heterocycle is also named heteroaryl. A non- aromatic heterocycle is a cycloalkyl, wherein at least one ring atom is replaced or several ring atoms are replaced by a nitrogen, oxygen and/or sulphur atom.
The term heterobicycle in the context of the present specification relates to two directly connected cycles, wherein at least one ring atom is replaced or several ring atoms are replaced by a nitrogen, oxygen and/or sulphur atom. A heterobicycle is composed of two heterocycles or of one heterocycle and one carbocycle.
The term unsubstituted Cn alkyl when used herein in the narrowest sense relates to the moiety -CnH2n- if used as a bridge between moieties of the molecule, or -CnH2n+i if used in the context of a terminal moiety.
The terms unsubstituted Cn alkyl and substituted Cn alkyl include a linear alkyl comprising or being linked to a cyclical structure, for example a cyclopropane, cyclobutane, cyclopentane or cyclohexane moiety, unsubstituted or substituted depending on the annotation or the context of mention, having linear alkyl substitutions. The total number of carbon and -where appropriate- N, O or other hetero atom in the linear chain or cyclical structure adds up to n.
Where used in the context of chemical formulae, the following abbreviations may be used: Me is methyl CH3, Et is ethyl -CH2CH3, Prop is propyl -(CH2)2CH3 (n-propyl, n-pr) or -CH(CH3)2 (iso-propyl, i-pr), but is butyl -C4H9, -(CH2)3CH3, -CHCH3CH2CH3, -CH2CH(CH3)2 or -C(CH3)3.
The term substituted alkyl in its broadest sense refers to an alkyl as defined above in the broadest sense, which is covalently linked to an atom that is not carbon or hydrogen, particularly to an atom selected from N, O, F, B, Si, P, S, Cl, Br and I, which itself may be -if applicable- linked to one or several other atoms of this group, or to hydrogen, or to an unsaturated or saturated hydrocarbon (alkyl or aryl in their broadest sense). In a narrower sense, substituted alkyl refers to an alkyl as defined above in the broadest sense that is substituted in one or several carbon atoms by groups selected from amine NH2, alkylamine NHR, imide NH, alkylimide NR, amino(carboxyalkyl) NHCOR or NRCOR, hydroxyl OH, oxyalkyl OR, oxy(carboxyalkyl) OCOR, carbonyl O and its ketal or acetal (OR)2, nitril ON, isonitril NO, cyanate ONO, isocyanate NCO, thiocyanate CNS, isothiocyanate NOS, fluoride F, choride Cl, bromide Br, iodide I, phosphonate PO3H2, PO3R2, phosphate OPO3H2 and OPO3R2, sulfhydryl SH, suflalkyl SR, sulfoxide SOR, sulfonyl SO2R, sulfanylamide SO2NHR, sulfate SO3H and sulfate ester SO3R, wherein the R substituent as used in the current paragraph, different from other uses assigned to R in the body of the specification, is itself an unsubstituted or substituted Ci to C12 alkyl in its broadest sense, and in a narrower sense, R is methyl, ethyl or propyl unless otherwise specified.
The term hydroxyl substituted group refers to a group that is modified by one or several hydroxyl groups OH.
The term amino substituted group refers to a group that is modified by one or several amino groups NH2.
The term carboxyl substituted group refers to a group that is modified by one or several carboxyl groups COOH.
Non-limiting examples of amino-substituted alkyl include -CH2NH2, -CH2NHMe, -CH2NHEt, -CH2CH2NH2, -CH2CH2NHMe, -CH2CH2NHEt, -(CH2)3NH2, -(CH2)3NHMe, -(CH2)3NHEt, -CH2CH(NH2)CH3, -CH2CH(NHMe)CH3, -CH2CH(NHEt)CH3, -(CH2)3CH2NH2, -(CH2)3CH2NHMe, -(CH2)3CH2NHEt, -CH(CH2NH2)CH2CH3, -CH(CH2NHMe)CH2CH3, -CH(CH2NHEt)CH2CH3, -CH2CH(CH2NH2)CH3, -CH2CH(CH2NHMe)CH3, -CH2CH(CH2NHEt)CH3, -CH(NH2)(CH2)2NH2, -CH(NHMe)(CH2)2NHMe, -CH(NHEt)(CH2)2NHEt, -CH2CH(NH2)CH2NH2, -CH2CH(NHMe)CH2NHMe, -CH2CH(NHEt)CH2NHEt, -CH2CH(NH2)(CH2)2NH2, -CH2CH(NHMe)(CH2)2NHMe, -CH2CH(NHEt)(CH2)2NHEt, -CH2CH(CH2NH2)2, -CH2CH(CH2NHMe)2 and -CH2CH(CH2NHEt)2for terminal moieties and -CH2CHNH2-, -CH2CHNHMe-, -CH2CHNHEt- for an amino substituted alkyl moiety bridging two other moieties.
Non-limiting examples of hydroxy-substituted alkyl include -CH2OH, -(CH2)2OH, -(CH2)sOH, -CH2CH(OH)CH3, -(CH2)4OH, -CH(CH2OH)CH2CH3, -CH2CH(CH2OH)CH3, -CH(OH)(CH2)2OH, -CH2CH(OH)CH2OH, -CH2CH(OH)(CH2)2OH and -CH2CH(CH2OH)2 for terminal moieties and -CHOH-, -CH2CHOH-, -CH2CH(OH)CH2-, -(CH2)2CHOHCH2-, - CH(CH2OH)CH2CH2-, -CH2CH(CH2OH)CH2-, -CH(OH)(CH2CHOH-, -CH2CH(OH)CH2OH, - CH2CH(OH)(CH2)2OH and -CH2CHCH2OHCHOH- for a hydroxyl substituted alkyl moiety bridging two other moieties.
The term sulfoxyl substituted group refers to a group that is modified by one or several sulfoxyl groups -SO2R, or derivatives thereof, with R having the meaning as laid out in the preceding paragraph and different from other meanings assigned to R in the body of this specification.
The term amine substituted group refers to a group that is modified by one or several amine groups -NHR or -NR2, or derivatives thereof, with R having the meaning as laid out in the preceding paragraph and different from other meanings assigned to R in the body of this specification.
The term carbonyl substituted group refers to a group that is modified by one or several carbonyl groups -COR, or derivatives thereof, with R having the meaning as laid out in the preceding paragraph and different from other meanings assigned to R in the body of this specification.
An ester refers to a group that is modified by one or several ester groups -CO2R, with R being defined further in the description.
An amide refers to a group that is modified by one or several amide groups -CON HR, with R being defined further in the description.
The term halogen-substituted group refers to a group that is modified by one or several halogen atoms selected (independently) from F, Cl, Br, I.
The term fluoro substituted alkyl refers to an alkyl according to the above definition that is modified by one or several fluoride groups F. Non-limiting examples of fluoro-substituted alkyl include -CH2F, -CHF2, -CF3, -(CH2)2F, -(CHF)2H, -(CHF)2F, -C2F5, -(CH2)3F, -(CHF)3H, - (CHF)3F, -C3F7, -(CH2)4F, -(CHF)4H, -(CHF)4F and -C4F9.
Non-limiting examples of hydroxyl- and fluoro-substituted alkyl include -CHFCH2OH, - CF2CH2OH, -(CHF)2CH2OH, -(CF2)2CH2OH, -(CHF)3CH2OH, -(CF2)3CH2OH, -(CH2)3OH, -CF2CH(OH)CH3, -CF2CH(OH)CF3, -CF(CH2OH)CHFCH3, and -CF(CH2OH)CHFCF3.
The term aryl in the context of the present specification signifies a cyclic aromatic C5-C10 hydrocarbon. Examples of aryl include, without being restricted to, phenyl and naphthyl.
A heteroaryl is an aryl that comprises one or several nitrogen, oxygen and/or sulphur atoms. Examples for heteroaryl include, without being restricted to, pyrrole, thiophene, furan, imidazole, pyrazole, thiazole, oxazole, pyridine, pyrimidine, thiazin, quinoline, benzofuran and indole. An aryl or a heteroaryl in the context of the specification additionally may be substituted by one or more alkyl groups. As used herein, the term pharmaceutical composition refers to a compound of the invention, or a pharmaceutically acceptable salt thereof, together with at least one pharmaceutically acceptable carrier. In certain embodiments, the pharmaceutical composition according to the invention is provided in a form suitable for topical, parenteral or injectable administration.
As used herein, the term pharmaceutically acceptable earner includes any solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (for example, antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drugs, drug stabilizers, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, and the like and combinations thereof, as would be known to those skilled in the art (see, for example, Remington: the Science and Practice of Pharmacy, ISBN 0857110624).
As used herein, the term treating or treatment of any disease or disorder (e.g. cancer) refers in one embodiment, to ameliorating the disease or disorder (e.g. slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms thereof). In another embodiment "treating" or "treatment" refers to alleviating or ameliorating at least one physical parameter including those which may not be discernible by the patient. In yet another embodiment, "treating" or "treatment" refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both. Methods for assessing treatment and/or prevention of disease.
The term metastasis in the context of the present specification relates to the dissemination and growth of neoplastic cells outside the original tumor bed in the same organ or in an organ distant from that in which they originated. In particular embodiments, the treatment or prevention with the disclosed compounds is employed for metastasis associated with aberrant FGFR signalling. The compounds of the invention specifically reduce the motile behaviour of metastatic cells and reduce dissemination. In particular embodiments, the compounds of the invention are employed for prevention or treatment of motility and dissemination of cancerous cells.
FGFR-driven tumorigenesis
Approximately 7% of all human tumors harbor an FGFR alteration (66% gene amplification, 26% mutations, 8% gene rearrangements) (Helsten, T. et al., Clin Cancer Res., 259-268 (2016) doi:10.1158/1078-0432. CCR-14-3212). FGFR1 is frequently amplified in 20 - 25% of squamous non-small cell lung cancer (Weiss, J. et al., Science Translational Medicine (2010) doi:10.1126/scitranslmed.3001451) and 15% breast cancer (Andre, F. et al., Clin Cancer Res., 15, 441-452 (2009)) and mutated in 18% of midline gliomas (Di Stefano, A. L. et al., Journal of Clinical Oncology 36, 2005 (2018)). FGFR2 is mainly activated by gene fusions in intrahepatic cholangiocarcinomas (iCCA, 15%) and mutations in 10% of endometrial tumors have also been described (Konecny, G. E. et al., The Lancet Oncology 16, 686-694 (2015); Verlingue, L. et al., European Journal of Cancer 87, 122-130 (2017).). FGFR3 is affected by mutations in urothelial carcinomas (up to 20% in the metastatic setting7); gene fusions (mainly FGFR3-TACC3) are present in glioblastomas and gliomas (3-6% (Di Stefano, A. L. et al., Journal of Clinical Oncology 36, 2005 (2018); Singh, D. etal., Science 337, 1231-1235 (2012); Di Stefano, A. L. et al., Clinical Cancer Research 21 , 3307-3317 (2015))), as well as in bladder cancer (2-3% (Robertson, A. G. et al., Cell 171 , 540-556.e25 (2017))). FGFR1-4 signal via Fibroblast Growth Factor Receptor Substrate 2 (FRS2)-dependent (RAS/MAPK and PI3K/AKT) and FRS2-independent (PLC-y, JAK-STAT) pathways (Turner, N. & Grose, Nat Rev Cancer, 1-14 (2010) doi:10.1038/nrc2780). FRS2 interacts with FGFRs via its phosphotyrosine binding domain (PTB) (Gotoh, N., Cancer Science 99, 1319-1325 (2008)) and increased expression or activation for FRS2 is involved in tumorigenesis of several tumor entities (Zhang, K. et al., Cancer Research 73, 1298-1307 (2013); Li, J.-L. & Luo, European review for medical and pharmacological sciences 24, 97-108 (2020); Wu, S. et al., Nature Communications 1-12 (2019) doi:10.1038/s41467-019-08576-5; Liu, J. et al., Oncogene, 35, 1750-1759 (2015); Chew, N. J. et al., Cell Communication and Signaling 18, 1-17 (2020)). Targeting of FRS2 function via repressing the FRS2-directed N-Myristoyltransferase repressed FGFR signaling, cell proliferation and migration in several cancer types (Li, Q. et al., The Journal of biological chemistry 293, 6434-6448 (2018)). Pharmacological inhibition of FGFRs reduces brain invasion in medulloblastoma and reduces metastasis in hepatocellular carcinoma (Huynh, H. et al., Hepatology 69, 943-958 (2019)) and lung cancer (Preusser, M. et al., Lung Cancer 83, 83-89 (2014)). FGFR-driven invasiveness depends on FRS2 (Huynh, H. et al., Hepatology 69, 943-958 (2019)). The FGF ligands of FGFRs are highly expressed in skeletal muscle (Pedersen, B. K. & Febbraio, M. A., Nature Reviews Endocrinology vol. 8457- 465 (2012)), bone (Su, N., Du, X. L. & Chen, Frontiers in Bioscience vol. 132842-2865 (2008)) and in CSF-secreting choroid plexus (Greenwood, S. et al., Cerebrospinal Fluid Research 5, 13-20 (2008)) and can serve as chemokinetic and chemotactic factors driving local invasion and distal spread. Repression of FGFR-FRS2 signaling may thus not only suppress the proliferative potential of tumor cells but also halt their metastatic spread driven by chemokinetic or chemotactic functions of secreted FGFs in the primary tumor and the target organ, respectively.
Selective (for example AZD4547, NVP-BGJ398 and JNJ-42756493) and non-selective (for example dovitinib or ponatinib) FGFR inhibitors have been explored for cancer therapy (Facchinetti, F. et al., Clin Cancer Res, (2020) doi:10.1158/1078-0432. CCR-19-2035; Yamaoka, T. et al., Int. J. Mol. Sci. 19, 1-35 (2018)). Resistance to FGFR inhibitors can evolve similarly as to other RTK inhibitors, either by the formation of gatekeeper mutations in the catalytic domain or the activation of alternative RTKs, which enable bypass mechanism for downstream signaling activation (Yamaoka, T., et al., Int. J. Mol. Sci. 19, 1-35 (2018)). Such mutations in FGFRs can occur in the ATP binding cleft and may create a steric conflict to limit drug-binding efficacy. Examples include FGFR3_V555M, FGFR1_V561 and FGFR2_V564, which induce resistance to FGFR inhibitors in vitro (Chell, V. et al., Oncogene 32, 3059-3070 (2013); Byron S. A. et a!., Neoplasia 15, 975-988 (2013)).
The inventors’ approach to target the non-enzymatically active FGFR adaptor protein FRS2 could prevent the evolution of FGFR gatekeeper mutations or help overcoming the resistance of gatekeeper FGFR-driven tumors by blocking signaling downstream of the RTK. Targeting FRS2 is likely also effective against tumors driven by the FGFR3-TACC3 fusion, where FRS2 is phosphorylated and transmits signaling to the oncogenic MAP kinase pathway (Chew, N. J. et al., Cell Communication and Signaling 18, 1-17 (2020)). Furthermore, toxicities related to FGFR inhibitor treatments have been reported and include hyper-phosphoremia, fatigue, dry skin and mouth with stomatitis, hand-foot syndrome and gastrointestinal dysfunctions (Facchinetti, F. et al., Clin Cancer Res, (2020) doi:10.1158/1078-0432.CCR-19-2035). An approach specifically targeting FRS2 with limited off-target compound activities may reduce the severity of toxicities currently associated with FGFR inhibition.
A first aspect of the invention relates to a compound of a general formula (I) or (lb):
Figure imgf000011_0001
wherein
- one R1 or one R2 is CN;
- each other R1 is independently selected from OR°, CN, halogen, NRN1RN2, SO2RS, NHSO2RS, SRS, COORA,
- each other R2 is independently selected from OR°, CN, halogen, NRN1RN2, SO2RS, NHSO2RS, SRS, COORA,
- n is 0, 1 , 2, or 3;
- m is 0, 1, 2, 3, 4, or 5; with the proviso that n+m 0;
- with RN1 and RN2 being independently selected from H, or unsubstituted or CN- and/or hydroxyl-substituted methyl;
- with Rs, RA and R° being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl;
- o is 0, 1 , or 2, particularly o is 0 or 1 , more particularly o is 0;
- X is N or CH, particularly X is CH.
In certain embodiments, the compound is of formula (I).
In certain embodiments, each R1 is independently selected from OR°, CN, halogen, SO2RS, NHSO2RS, SRS, COORA
In certain embodiments, n is 1, 2, or 3. In certain embodiments, n is 2.
In certain embodiments, m is 0 or 1. In certain embodiments, m is 1.
In certain embodiments, the compound is described by a general formula (II):
Figure imgf000012_0001
wherein R1, R2, n and m have the same definition as above.
In certain embodiments, the compound is described by a general formula (lib):
Figure imgf000013_0001
wherein X, o, R1, R2, n and m have the same definition as above.
In certain embodiments, the compound is described by a general formula (III):
Figure imgf000013_0002
wherein R1, R2, and n have the same definition as above.
In certain embodiments, the compound is described by a general formula (lllb):
Figure imgf000013_0003
wherein X, o, R1, R2 and n have the same definition as above.
In certain embodiments, the compound is described by a general formula (IV):
Figure imgf000013_0004
wherein R1, R2, and n have the same definition as above.
In certain embodiments, the compound is described by a general formula (IVb):
Figure imgf000014_0001
wherein X, o, R1, R2 and n have the same definition as above. In certain embodiments, the compound is described by a general formula (V):
Figure imgf000014_0002
wherein R1 and n have the same definition as above.
In certain embodiments, the compound is described by a general formula (Vb):
Figure imgf000014_0003
wherein X, o, R1, R2 and n have the same definition as above.
In certain embodiments, the compound is described by a general formula (VI):
Figure imgf000015_0001
(VI).
In certain embodiments, the compound is described by a general formula (Vlb):
Figure imgf000015_0002
For formula (VI) and (Vlb): - SY is SO2 or S;
- R3 is selected from the group of H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl;
- X, o, R1, R2, and n have the same definition as above.
In certain embodiments of formulas (I) or (III), one R1 is CN, and the other R1 is/are independently selected from CN, halogen, OR°, COORA, SRS, NHSO2Rs, SC>2RS with Rs, RA, and R° being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl. In certain embodiments, the other R1 is/are halogen. In certain embodiments, the other R1 is/are is Cl.
In certain embodiments of formulas (II), (IV), or (V), each R1 is independently selected from halogen, CN, OR°, COORA, SRS, NHSO2Rs, SO2Rs with Rs, RA, and R° being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl. In certain embodiments, R1 is halogen. In certain embodiments, R1 is Cl. In certain embodiments of formulas (I) or (III), one R1 is CN, and the other R1 is/are independently selected from SRS, NHSO2Rs, SC>2Rs with each Rs being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl.
In certain embodiments of formulas (II), (IV), or (V), each R1 is independently selected from CN, SRs, NHSC>2RS, SC>2RS with each Rs being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl.
In certain embodiments of formulas (lib), (IVb), or (Vb), each R1 is independently selected from halogen, CN, OR°, COORA, SRS, NHSO2Rs, SO2RS with Rs, RA, and R° being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl. In certain embodiments, R1 is halogen. In certain embodiments, R1 is Cl.
In certain embodiments of formulas (lb) or (I lib), one R1 is CN, and the other R1 is/are independently selected from SRs, NHSO2Rs, SO2Rs with each Rs being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl.
In certain embodiments of formulas (lib), (IVb), or (Vb), each R1 is independently selected from CN, SRS, NHSO2Rs, SO2Rs with each Rs being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl.
In certain embodiments, R1 is CN. In certain embodiments, R1 is SRs. In certain embodiments, R1 is NHSO2RS. In certain embodiments, R1 is SO2Rs. In certain embodiments, R1 is NHSRS.
In certain embodiments, each R2 is independently selected from halogen, CN, OR°, SRS, NHSO2RS, SO2RS with Rs, RA, and R° being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl. In certain embodiments, R2 is halogen. In certain embodiments, R2 is Cl.
In certain embodiments, each R2 is independently selected from CN, SRs, NHSO2Rs, SO2Rs with each Rs being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl.
In certain embodiments, R2 is CN. In certain embodiments, R2 is SRS. In certain embodiments, R2 is NHSO2RS. In certain embodiments, R2 is SO2Rs In certain embodiments, R2 is NHSRs.
In certain embodiments of formulas (I) or (III) or (V) or (lb) or (111 b) or (Vb), one R1 is CN, and the other R1 is/are independently selected from NHSRs, SRS, NHSO2Rs, SO2Rs with each Rs being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl- substituted methyl. In certain embodiments, one R1 is CN and one other R1 is selected from NHSRs, SRS, NHSO2RS, SO2RS In certain embodiments, n is 2, and one R1 is CN and the other one is NHSO2Rs. A second aspect of the invention relates to a compound according to the first aspect for use as a medicament.
A third aspect of the invention relates to a compound according to the first aspect for use in treatment or prevention of cancer.
A fourth aspect of the invention relates to a compound according to the first aspect for use in treatment or prevention of metastasis.
A fifth aspect of the invention relates to a compound according to the first aspect for use as an angiogenesis antagonist, particularly an angiogenesis antagonist in treatment or prevention of cancer.
In certain embodiments, said cancer or said metastasis arises from a cancer selected from ovarian cancer, urothelial carcinomas, breast cancer, endometrial adenocarcinnoma, cholagiocarcinoma, non-small cell lung cancer, head and cancer, sarcoma, melanoma, glioma, pancreatic cancer, renal cell carcinoma, neuroendocrine carcinoma, leiomyosarcoma, liposarcoma, bladder cancer, medulloblastoma, pediatric brain tumours, multiple myeloma, colorectal cancer and gastric cancer. In certain embodiments, said cancer or said metastasis arises from a cancer selected from ovarian cancer, urothelial carcinomas, breast cancer, endometrial adenocarcinnoma, non-small cell lung cancer, head and cancer, sarcoma, melanoma, glioma, bladder cancer, medulloblastoma, pediatric brain tumours, multiple myeloma, colorectal cancer and gastric cancer.
In certain embodiments, said cancer is selected from bladder cancer, multiple myeloma, gastric cancer, pediatric brain tumour, medulloblastoma, glioblastoma, ependymoma, colorectal cancer and sarcoma. In certain embodiments, said cancer is selected from bladder cancer, pediatric brain tumour, medulloblastoma, multiple myeloma, colorectal cancer and gastric cancer.
A sixth aspect of the invention relates to a compound according to the first aspect for use in prevention or treatment of an FGFR-driven disease.
Medical treatment, Dosage Forms and Salts
Similarly, within the scope of the present invention is a method or treating cancer or metastasis in a patient in need thereof, comprising administering to the patient a compound according to the above description.
Similarly, a dosage form for the prevention or treatment of cancer is provided, comprising a non-agonist ligand or antisense molecule according to any of the above aspects or embodiments of the invention. The skilled person is aware that any specifically mentioned drug may be present as a pharmaceutically acceptable salt of said drug. Pharmaceutically acceptable salts comprise the ionized drug and an oppositely charged counterion. Non-limiting examples of pharmaceutically acceptable anionic salt forms include acetate, benzoate, besylate, bitatrate, bromide, carbonate, chloride, citrate, edetate, edisylate, embonate, estolate, fumarate, gluceptate, gluconate, hydrobromide, hydrochloride, iodide, lactate, lactobionate, malate, maleate, mandelate, mesylate, methyl bromide, methyl sulfate, mucate, napsylate, nitrate, pamoate, phosphate, diphosphate, salicylate, disalicylate, stearate, succinate, sulfate, tartrate, tosylate, triethiodide and valerate. Non-limiting examples of pharmaceutically acceptable cationic salt forms include aluminium, benzathine, calcium, ethylene diamine, lysine, magnesium, meglumine, potassium, procaine, sodium, tromethamine and zinc.
Dosage forms may be for enteral administration, such as nasal, buccal, rectal, transdermal or oral administration, or as an inhalation form or suppository. Alternatively, parenteral administration may be used, such as subcutaneous, intravenous, intrahepatic or intramuscular injection forms. Optionally, a pharmaceutically acceptable carrier and/or excipient may be present.
Pharmaceutical Composition and Administration
Another aspect of the invention relates to a pharmaceutical composition comprising a compound of the present invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. In further embodiments, the composition comprises at least two pharmaceutically acceptable carriers, such as those described herein.
In certain embodiments of the invention, the compound of the present invention is typically formulated into pharmaceutical dosage forms to provide an easily controllable dosage of the drug and to give the patient an elegant and easily handleable product.
In embodiments of the invention relating to topical uses of the compounds of the invention, the pharmaceutical composition is formulated in a way that is suitable for topical administration such as aqueous solutions, suspensions, ointments, creams, gels or sprayable formulations, e.g., for delivery by aerosol or the like, comprising the active ingredient together with one or more of solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives that are known to those skilled in the art.
The pharmaceutical composition can be formulated for oral administration, parenteral administration, or rectal administration. In addition, the pharmaceutical compositions of the present invention can be made up in a solid form (including without limitation capsules, tablets, pills, granules, powders or suppositories), or in a liquid form (including without limitation solutions, suspensions or emulsions). The dosage regimen for the compounds of the present invention will vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the species, age, sex, health, medical condition, and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; the route of administration, the renal and hepatic function of the patient, and the effect desired. In certain embodiments, the compounds of the invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three, or four times daily.
In certain embodiments, the pharmaceutical composition or combination of the present invention can be in unit dosage of about 1-1000 mg of active ingredient(s) for a subject of about 50-70 kg. The therapeutically effective dosage of a compound, the pharmaceutical composition, or the combinations thereof, is dependent on the species of the subject, the body weight, age and individual condition, the disorder or disease or the severity thereof being treated. A physician, clinician or veterinarian of ordinary skill can readily determine the effective amount of each of the active ingredients necessary to prevent, treat or inhibit the progress of the disorder or disease.
The pharmaceutical compositions of the present invention can be subjected to conventional pharmaceutical operations such as sterilization and/or can contain conventional inert diluents, lubricating agents, or buffering agents, as well as adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers and buffers, etc. They may be produced by standard processes, for instance by conventional mixing, granulating, dissolving or lyophilizing processes. Many such procedures and methods for preparing pharmaceutical compositions are known in the art, see for example L. Lachman et al. The Theory and Practice of Industrial Pharmacy, 4th Ed, 2013 (ISBN 8123922892).
Method of Manufacture and Method of Treatment according to the invention
The invention further encompasses, as an additional aspect, the use of a compound as identified herein, or its pharmaceutically acceptable salt, as specified in detail above, for use in a method of manufacture of a medicament for the treatment or prevention of cancer or metastasis.
Similarly, the invention encompasses methods of treatment of a patient having been diagnosed with a disease associated with cancer or metastasis. This method entails administering to the patient an effective amount of a compound as identified herein, or its pharmaceutically acceptable salt, as specified in detail herein.
Wherever alternatives for single separable features such as, for example, a ligand type or medical indication are laid out herein as “embodiments”, it is to be understood that such alternatives may be combined freely to form discrete embodiments of the invention disclosed herein. Thus, any of the alternative embodiments for a ligand type may be combined with any medical indication mentioned herein.
The invention is further illustrated by the following examples and figures, from which further embodiments and advantages can be drawn. These examples are meant to illustrate the invention but not to limit its scope.
Figure imgf000020_0001
Whenever the figures refer to “uM” it is to be understood that “pmol/L” is meant.
Fig. 1 shows the efficacy of the efficacy of F3.18, F18.2, F18.7, F18.8, F18.9 at 3 different concentrations - 1 .M, 5 .M and 10 .M.
Fig. 2 shows the efficacy of F3.18, F18.2, F18.7, F18.8, F18.9 at 10 .M
Fig. 3 shows the binding affinities and dissociation constant (Kd) of F3.18, F18.2,
F18.7, F18.8, F18.9. Nano diffraction scanning fluorimetry (nanoDSF) and Microscale thermophoresis (MST) are biophysical assays used to assess the binding of the compounds to the target protein. Any temperature shift above 1.5 degree Celsius is considered as indication for significant binding
Fig. 4 shows the effective inhibitory concentration of compound F3.18
Fig. 5 A and B show the Biochemical specificity of F3.18, F18.2, F18.7, F18.8, F18.9 determining the ability of the compounds to inhibit FGF signalling pathway without affecting other signalling pathways. Fig 5a shows for 1) the Control - DAOY l-A-EGFP cells unstimulated, serum starved overnight and then lysed,
2) bFGF (100ng/ml) - Overnight serum starved DAOY l-A-EGFP cells stimulated with bFGF for 10 minutes and then lysed and 3) F3.18 (10 .M) - Overnight serum starved DAOY l-A-EGFP cells treated with F3.18 for four hours, cells stimulated with bFGF for 10 minutes and then lysed. Fig 5B shows for 1) the Control - DAOY LA-EGFP cells unstimulated, serum starved overnight and then lysed, 2) bFGF (100ng/ml) - Overnight serum starved DAOY l_A-EGFP cells stimulated with bFGF for 10 minutes and then lysed and
3) F18.2 (10 .M) - Overnight serum starved DAOY l-A-EGFP cells treated with F18.2 for four hours, cells stimulated with bFGF for 10 minutes and then lysed,
4) F18.7 (10 .M) - Overnight serum starved DAOY l-A-EGFP cells treated with F18.7 for four hours, cells stimulated with bFGF for 10 minutes and then lysed,
5) F18.8 (10 .M) - Overnight serum starved DAOY l-A-EGFP cells treated with F18.8 for four hours, cells stimulated with bFGF for 10 minutes and then lysed, 6) F18.9 (10 ,M) - Overnight serum starved DAOY LA-EGFP cells treated with F18.9 for four hours, cells stimulated with bFGF for 10 minutes and then lysed.
Fig. 6 shows the structure of the compounds F3.18, F18.2, F18.7, F18.8 and F18.9.
Fig. 7 A) Binding site 1 is not involved in FGFR binding and located below the interaction site of FGFR’s N-terminus with FRS2. B) Binding site 2 is the extended surface area interacting with FGFR’s C-terminal end.
Fig. 8 Compounds of the invention.
Fig. 9 Spheroid invasion assay using DAOY cells stimulated with bFGF +/- BGJ398 or F18.7 to determine the EC50 of F18.7.
Fig. 10 Spheroid invasion assay using DAOY cells stimulated with bFGF +/- BGJ398 or F3.18 series compounds.
Fig. 11 Spheroid invasion assay using DAOY cells stimulated with bFGF +/- BGJ398 or F18.7 series compounds.
Fig. 12 Cell titer gio assay performed with DAOY cells treated with BGJ398 or FF18.7.
Fig. 13 Cell titer gio assay performed with AGS cells treated with BGJ398 or F3.118.
Fig. 14 Cell titer gio assay performed with DMS114 cells treated with BGJ398 or
F3.18.
Fig. 15 Cell titer gio assay performed with HCT 116 cells treated with BGJ398 or F3.18.
Fig. 16 Cell titer gio assay performed with M059K cells treated with BGJ398 or F3.18.
Fig. 17 Cell titer gio assay performed with RT112 cells treated with BGJ398 or F3.18.
Fig. 18 Cell titer gio assay performed with SNLI16 cells treated with BGJ398 or F3.18.
Fig. 19 Cell titer gio assay performed with SKOV3 cells treated with BGJ398 or F3.18.
Fig. 20 Table showing the in vitro absorption, distribution, metabolism, elimination and toxicity (ADMET) properties of F3.18. Efflux ration represents the permeability of F3.18, Semi-thermodynamic solubility shows the solubility of F3.18 in aqueous solutions. Intrinsic clearance and t1/2 shows the metabolic stability of F3.18 MTT shows the toxicity of F3.18 and potency shows the efficacy of F3.18.
Fig. 21 In vivo pharmacokinetics, 3 mice/treatment, serum concentration of compounds in pM. Fig. 22 Immunoblots using various FGFR-driven cell lines treated with BGJ398 or F3.18 showing the effect of the treatment on the downstream effectors of FGF signalling.
Fig. 23 Spheroid invasion assay using DAOY cells stimulated with bFGF +/- BGJ398 or F18.1 , F18.4 and F18.10 series compounds. F3.18 is used as positive control.
Fig. 24 shows chemical structure and properties of F18.7 (F2i) AND F1873 (Non-nitro analog of F2i).
Fig. 25 shows in-vitro properties of F18.7 and F1873.
Fig. 26 shows in-vitro ADME of F18.7 and F1873.
Fig. 27 shows tested compounds.
Fig. 28 shows functional assay - test for inhibition of cell invasion by the compounds using ACINDA via Spheroid invasion assay (SIA).
Fig. 29 shows functional assay - test for inhibition of cell invasion by the compounds using ACINDA via Spheroid invasion assay (SIA). Control - unstimulated cells in the form of spheroids. bFGF - Maximum invasion possible -s activated by the FGF signaling. BGJ398 - FGFR kinase inhibitor for comparison. F18.7 - F2i (lead) - parent compound. F1873 - non-nitro analogue of F2i (lead).
Fig. 30 shows nanoDSF - assay. A +ve or -ve temperature shift of 1.5°C is a significant binder to target protein.
Fig. 31 shows determination of lipophilicity - LogD. LogD (lipophilicity) of a compound is linked to the permeability, clearance and metabolic stability. Optimal logD range: 1 to 3. Most compounds showed acceptable logD values within the range of 2 to 4. Some compounds showed high logD values of >4. Few compounds were weakly ionizable and not clearly detected (ND) by LCMS.
Fig. 32 shows determination of aqueous solubility.
Fig. 33 shows determination of Kd (cell free IC50) and ADME properties.
Fig. 34 shows tested compounds.
Fig. 35 shows the percentage of inhibition in spheroid invasion assay. Control is unstimulated cells in the form of spheroids. BGJ398 is an FGFR inhibitor for comparison.
Fig. 36 shows a nanoDSF biophysical assay. A +ve or -ve temperature shift of 1.5°C is a significant binder to target protein. Fig. 37 shows KD, IC50, and EC50 data for selected compounds. KD was determined using microscale thermophoresis. IC50 was determined using cell titer gio assay. EC50 was determined using spheroid invasion assay.
The inventors designed an inhibitor of FRS2-FGFR interaction by screening a large library of fragments of small molecules. The inventors identified F3.18 as a putative small molecule inhibitor of FRS2-FGFR interaction. The inventors confirmed the binding of F3.18 to FRS2 using biophysical assays - nanoDSF, MST and NMR analysis. The inventors evaluated the efficacy of F3.18 in inhibiting cancer cell invasion and proliferation using FGFR-driven cancer cell models. Results from the spheroid invasion assay and cell titer gio assay show that F3.18 effectively inhibits cancer cell invasion and proliferation in all the FGFR-driven cancer cell lines tested. To test the effect of F3.18 on FGF signaling pathway, we used immunoblotting. F3.18 inhibits the FGF signal transduction by inhibiting the phosphorylation of the downstream effectors of FGF signaling pathway. The inventors used in vitro ADMET studies and in vivo PK studies to determine the ‘drug-like’ properties of F3.18. Results from these assays demonstrate that F3.18 has good permeability, moderate solubility and intrinsic clearance, low toxicities, and high potency. The in vivo PK studies show that F3.18 is well- tolerated in mice and could be safely administered via oral and intravenous route to living organisms for the treatment of FGFR-driven diseases.
Methods and instruments:
Spheroid invasion assay (SIA) and automated cell dissemination counter (aCDc)
1000 cells/100 pL per well were seeded in cell-repellent 96 well microplate (650790, Greiner Bio-one). The cells were incubated at 37°C overnight to form spheroids. 70 pl of the medium were removed from each well and remaining medium with spheroid overlaid with 2.5% bovine collagen 1. Following the polymerization of collagen, fresh medium was added to the cells and treated with bFGF and/or with compounds. The cells were allowed to invade the collagen matrix for 24 h, after which they were fixed with 4% PFA and stained with Hoechst. Images were acquired on an Axio Observer 2 mot plus fluorescence microscope (Zeiss, Munich, Germany) using a 5x objective. Cell invasion is determined as the average of the distance invaded by the cells from the center of the spheroid as determined using automated cell dissemination counter (aCDc) with our cell dissemination counter software aSDIcs (Kumar et al., Sci Rep 5, 15338 (2015)). Nano differential scanning fluorimetry (nanoDSF)
Purified FRS2 protein tagged with 6X Histidine residues and Guanine nucleotide-binding protein subunit beta (GB1) was diluted in the protein buffer (100mM sodium phosphate, 50mM NaCI, 0.5mM EDTA, 50mM arginine, 1 mM TCEP, pH 7.0) to final concentration of 30 .M. The compounds were dissolved in 100% at 50 or 100mM and further diluted to 1 mM with a final concentration of 100% DMSO. Compound and protein were mixed at 1 :1 ration yielding final concentrations of 15 .M and 500 .M for the compounds. The mixture was incubated at room temperature for 15 minutes before measurement. The measurement was performed on a Prometheus system in high sensitivity capillaries. Samples were subjected to a temperature gradient of 20 to 95°C with 1°C/min intervals.
Microscale thermophoresis (MST)
Purified FRS2 protein tagged with 6X Histidine residues and Guanine nucleotide-binding protein subunit beta (GB1) was labelled with 2nd generation BLUE-NHS dye. The protein was labelled at a final concentration of 20 .M with 60 .M dye. The labelling was performed in the protein buffer without arginine supplementation. Arginine was re-buffered to protein’s buffer post-labelling. The compounds were dissolved in 100% at 50 or 100mM and further diluted to 1mM with a final concentration of 100% DMSO. The compounds were then diluted. In a 1 :1 serial dilution from 1 mM to 61.04nM in protein buffer supplemented with 10% DMSO. 10 l of 50nM labelled protein was added to 10 l of each compound dilution for a final labelled protein concentration of 25nM and DMSO-concentration of 5%. The samples were incubated at room temperature for 15 minutes. The experiments were performed in premium-coated capillaries. Excitation power was set at 20%, MST power to 40% (4 Kelvin temperature gradient) with a laser-on time of 20 seconds and a laser-off time of 3 seconds. Temperature was set to 25°C. Each measurement was repeated twice. The interaction was measured in two independent duplicates.
Immunoblotting (IB)
Cancer cells were treated with bFGF (100ng/ml) and/or with compounds and lysed using Radioimmunoprecipitation assay (RIPA) buffer. RIPA buffer lysates were resolved by SDS- PAGE and transferred to a nitrocellulose membrane using a transfer apparatus according to the manufacturer’s instructions (Bio-Rad). Membranes were probed with primary antibodies against phospho-FRS2, FRS2, ERK1/2, phospho-ERK1/2, AKT, phosphor-AKT, phospho- PKC and tubulin. HRP-linked secondary antibodies (1 :5000) were used to detect the primary antibodies. Chemiluminescence detection was performed using ChemiDoc Touch Gel and Western Blot imaging system (BioRad). Cell titer gio assay
The metabolic activity and the proliferation of the cells were determined using the Cell Titer gio assay from Promega according to the manufacturer’s instructions. In brief, 250 cells/1 OO l/per well (for up to 72 h incubation) were seeded in Greiner Bio-One p-clear 384 well plates (655090, Greiner Bio-One) and incubated overnight at 37°C. The old media was then replaced with fresh serum-free media and the cells were treated with BGJ398 or F3.18 till the desired time point. Following appropriate incubation for each timepoint, 10 pl of the Cell titer gio reagent was added to each well (final concentration of cell titer gio reagent per well is 1:10) and incubated at 37° C for 30 minutes. The luminescence was then measured with a signal integration time of 0.5 to 1 second per well.
In vivo pharmacokinetics
3 Healthy non-SCID mice per group were intravenously or orally treated with F18.7. Blood samples were collected at 2, 4, 6, 8 and 24 hours after treatment. Serum from the collected blood samples were isolated and the concentration of F18.7 in the serum was measure to determine the intrinsic clearance of F18.7.
Pathway analysis
RIPA buffer FGFR-driven cell lysates were resolved by SDS-PAGE and transferred to a nitrocellulose membrane using a transfer apparatus according to the manufacturer’s instructions (Bio-Rad). Membranes were probed with primary antibodies against phospho- FRS2, FRS2, ERK1/2, phospho-ERK1/2, AKT, phospho-AKT, phospho-PKC and tubulin. HRP-linked secondary antibodies (1:5000) were used to detect the primary antibodies. Chemiluminescence detection was performed using ChemiDoc Touch Gel and Western Blot imaging system (BioRad). Integrated density of Immuno-reactive bands was quantified using Adobe Photoshop CS5.
Availability of Compounds
All compounds were purchased at ChemBridge or ChemDiv under the following vendor IDs:
F3.18 #5947468 (ChemBridge)
F18.2 4597-0445 (ChemDiv)
F18.7 2945-0019 (ChemDiv)
F18.8 8010-3211 (ChemDiv)
F18.9 8010-3214 (ChemDiv)

Claims

Claims
1. A compound of a general formula (I) or (lb), particularly the compound is of formula
Figure imgf000026_0001
wherein
- one R1 or one R2 is CN;
- each other R1 is independently selected from OR°, CN, halogen, NRN1RN2, SO2RS, NHSO2RS, SRS, COORA, particularly each R1 is independently selected from OR°, CN, halogen, SO2RS, NHSO2RS, SRS, COORA;
- each other R2 is independently selected from OR°, CN, halogen, NRN1RN2, SO2RS, NHSO2RS, SRS, COORA,
- n is 0, 1 , 2, or 3, particularly n is 1 , 2, or 3, more particularly n is 2;
- m is 0, 1 , 2, 3, 4, or 5, particularly m is 0 or 1 , more particularly m is 1 ; with the proviso that n+m 0;
- with RN1 and RN2 being independently selected from H, or unsubstituted or CN- and/or hydroxyl-substituted methyl;
- with Rs, RA and R° being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl;
25 - o is 0, 1 , or 2, particularly o is 0 or 1 , more particularly o is 0;
- X is N or CH, particularly X is CH. The compound according to claim 1, wherein the compound is described by a general formula (II) or (lib), particularly the compound is of formula (II):
Figure imgf000027_0001
wherein
- X, o, R1, R2, and m have the same definition as in claim 1;
- n is 0, 1 , or 2 particularly n is 1 , or 2 more particularly n is 1. The compound according to any one of the preceding claims 1 to 2, wherein the compound is described by a general formula (III) or (II lb), particularly the compound is of formula (III):
Figure imgf000027_0002
Figure imgf000028_0001
wherein X, o, R1, R2, and n have the same definition as in claim 1. The compound according to any one of the preceding claims, wherein the compound is described by a general formula (IV) or (IVb), particularly the compound is of formula (IV):
Figure imgf000028_0002
wherein
- X, o, R1 , and R2 have the same definition as in claim 1 ;
- n is 0, 1 , or 2 particularly n is 1 , or 2 more particularly n is 1. The compound according to any one of the preceding claims 1 to 2, wherein the compound is described by a general formula (V) or (Vb), particularly the compound is of formula (V):
Figure imgf000029_0001
wherein X, o, R1 and n have the same definition as in claim 1. The compound according to any one of the preceding claims, wherein the compound is described by a general formula (VI) or (Vlb), particularly the compound is of formula (VI):
Figure imgf000029_0002
Figure imgf000030_0001
wherein
- SY is SO2 or S, particularly SY is SO2;
- R3 is selected from the group of H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl, particularly R3 is unsubstituted methyl;
- X, o, R1, and R2 have the same definition as in claim 1 , particularly R1 is Cl;
- n is 0, 1 , or 2, particularly n is 0 or 1 , more particularly n is 0. The compound according to any one of the preceding claims 1 or 3, wherein one R1 is CN, and the other R1 is/are independently selected from CN, halogen, OR°, COORA, SRS, NHSC>2RS, SC>2RS with Rs, RA, and R° being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl, particularly the other R1 is/are halogen, more particularly the other R1 is/are is Cl. The compound according to any one of the preceding claims 2, 4, 5, or 6, wherein each R1 is independently selected from halogen, CN, OR°, COORA, SRS, NHSO2Rs, SO2RS with Rs, RA, and R° being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl, particularly R1 is halogen, more particularly R1 is Cl. The compound according to any one of the preceding claims 1 or 3, wherein one R1 is CN, and the other R1 is/are independently selected from SRs, NHSO2Rs, SO2Rs with each Rs being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl. The compound according to any one of the preceding claims 2, 4, 5, or 6, wherein each R1 is independently selected from CN, SRs, NHSO2Rs, SO2Rs with each Rs being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl. The compound according to any one of the preceding claims 1-4 or 6-10, wherein each R2 is independently selected from halogen, CN, OR°, SRS, NHSO2Rs, SO2Rs with Rs, RA, and R° being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl, particularly R2 is halogen, more particularly R2 is Cl. The compound according to any one of the preceding claims 1-4 or 6-10, wherein each R2 is independently selected from CN, SRS, NHSO2Rs, SC>2RS with each Rs being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl. The compound according to any one of the preceding claims 1 or 3 or 5, wherein one R1 is CN, and one other R1 is selected from NHSRs, SRS, NHSO2Rs, SO2RS with each Rs being independently selected from H, or unsubstituted or CN-, halogen- and/or hydroxyl-substituted methyl, particularly one R1 is CN, and one other R1 is NHSO2Rs A compound according to any one of claims 1 to 13 for use as a medicament. A compound according to any one of claims 1 to 13 for use in treatment or prevention of cancer. A compound according to any one of claims 1 to 13 for use in treatment or prevention of metastasis. A compound according to any one of claims 1 to 13 for use as an angiogenesis antagonist, particularly an angiogenesis antagonist in treatment or prevention of cancer. The compound for use according to claim 15, 16, or 17, wherein said cancer or said metastasis arises from a cancer selected from ovarian cancer, urothelial carcinomas, breast cancer, endometrial adenocarcinnoma, cholagiocarcinoma, non-small cell lung cancer, head and cancer, sarcoma, melanoma, glioma, pancreatic cancer, renal cell carcinoma, neuroendocrine carcinoma, leiomyosarcoma, liposarcoma, bladder cancer, medulloblastoma, pediatric brain tumours, multiple myeloma, colorectal cancer and gastric cancer, particularly wherein said cancer or said metastasis arises from a cancer selected from ovarian cancer, urothelial carcinomas, breast cancer, endometrial adenocarcinnoma, non-small cell lung cancer, head and cancer, sarcoma, melanoma, glioma, bladder cancer, medulloblastoma, pediatric brain tumours, multiple myeloma, colorectal cancer and gastric cancer. A compound according to any one of claims 1 to 13 for use in prevention or treatment of an FGFR-driven disease.
30
PCT/EP2022/085077 2021-12-08 2022-12-08 Small-molecule inhibitors of the frs2-fgfr interaction WO2023105008A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP21213222 2021-12-08
EP21213222.9 2021-12-08

Publications (1)

Publication Number Publication Date
WO2023105008A1 true WO2023105008A1 (en) 2023-06-15

Family

ID=78827978

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2022/085077 WO2023105008A1 (en) 2021-12-08 2022-12-08 Small-molecule inhibitors of the frs2-fgfr interaction

Country Status (1)

Country Link
WO (1) WO2023105008A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024121437A1 (en) * 2022-12-09 2024-06-13 Universität Zürich Small-molecule inhibitors of the frs2-fgfr interaction

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004072046A2 (en) * 2003-02-12 2004-08-26 Carex S.A. Quinoline derivatives and their use for modulation of lxr activity
WO2010141738A2 (en) * 2009-06-03 2010-12-09 President And Fellows Of Harvard College Compositions and method for inhibiting tumor growth
WO2013149997A1 (en) * 2012-04-02 2013-10-10 Almirall, S.A. Substituted tricyclic compounds with activity towards ep1 receptors
WO2016014847A1 (en) * 2014-07-23 2016-01-28 Northeastern University Ligands for alpha-7 nicotinic acetylcholine receptors and methods of treating neurological and inflammatory conditions
WO2021250025A1 (en) * 2020-06-08 2021-12-16 Universität Zürich Small-molecule inhibitors of the frs2-fgfr interaction and their use in medicine, in the prevention and treatment of cancer

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004072046A2 (en) * 2003-02-12 2004-08-26 Carex S.A. Quinoline derivatives and their use for modulation of lxr activity
WO2010141738A2 (en) * 2009-06-03 2010-12-09 President And Fellows Of Harvard College Compositions and method for inhibiting tumor growth
WO2013149997A1 (en) * 2012-04-02 2013-10-10 Almirall, S.A. Substituted tricyclic compounds with activity towards ep1 receptors
WO2016014847A1 (en) * 2014-07-23 2016-01-28 Northeastern University Ligands for alpha-7 nicotinic acetylcholine receptors and methods of treating neurological and inflammatory conditions
WO2021250025A1 (en) * 2020-06-08 2021-12-16 Universität Zürich Small-molecule inhibitors of the frs2-fgfr interaction and their use in medicine, in the prevention and treatment of cancer

Non-Patent Citations (60)

* Cited by examiner, † Cited by third party
Title
ANDRE, F. ET AL., CLIN CANCER RES, vol. 15, 2009, pages 441 - 452
AUSUBEL ET AL.: "Short Protocols in Molecular Biology", 1999, JOHN WILEY & SONS, INC.
BECKER, DLEE, P. LRODECK, UHERLYN, M, ONCOGENE, vol. 7, 1992, pages 2303 - 2313
BYRON S. A. ET AL., NEOPLASIA, vol. 15, 2013, pages 975 - 988
CAPPELLEN, D ET AL., NAT GENET, vol. 23, 1999, pages 18 - 20
CHELL, V ET AL., ONCOGENE, vol. 32, 2013, pages 3059 - 3070
CHEW, N. J. ET AL., CELL COMMUNICATION AND SIGNALING, vol. 18, 2020, pages 1 - 17
COLE, C. ET AL., CANCER BIOL THER, vol. 10, 2010
DI STEFANO, A. L. ET AL., CLINICAL CANCER RESEARCH, vol. 21, 2015, pages 3307 - 3317
DI STEFANO, A. L. ET AL., JOURNAL OF CLINICAL ONCOLOGY, vol. 36, 2018, pages 2005
DUTT, A. ET AL., PLOS ONE, vol. 6, 2011
FACCHINETTI, F. ET AL., CLIN CANCER RES, 2020
GOLDSTEIN, MMELLER, IORR-URTREGER, A., GENE CHROMOSOME CANC, vol. 46, 2007, pages 1028 - 1038
GOTOH, N., CANCER SCIENCE, vol. 99, 2008, pages 1319 - 1325
GREENWOOD, S ET AL., CEREBROSPINAL FLUID RESEARCH, vol. 5, 2008, pages 13 - 20
HELSTEN, T. ET AL., CLIN CANCER RES, 2016, pages 259 - 268
HUYNH, H. ET AL., HEPATOLOGY, vol. 69, 2019, pages 943 - 958
IMRICH HANS-GEORG ET AL: "From Nitrobenzenes to Substituted Tetrahydroquinolines in a Single Step by a Domino Reduction/Imine Formation/Aza-Diels-Alder Reaction", THE JOURNAL OF ORGANIC CHEMISTRY, vol. 80, no. 4, 20 February 2015 (2015-02-20), pages 2319 - 2332, XP093023086, ISSN: 0022-3263, DOI: 10.1021/jo502882y *
ISHIWATA, T. ET AL., AM J PATHOL, vol. 180, 2012, pages 1928 - 1941
JANG, J. HSHIN, K. HPARK, J. G, CANCER RES, vol. 61, 2001, pages 3541 - 3543
JIANG, X ET AL., J DIABETES RES, 2013
KALFF, ASPENCER, A, BLOOD CANCER J, vol. 2, 2012
KISELYOV ALEXANDER S ET AL: "Solid Support Synthesis of Tetrahydroquinolines via the Grieco Three Component Condensation", TETRAHEDRON LETTERS, vol. 38, no. 35, 1997, pages 6163 - 6166, XP085609606, ISSN: 0040-4039, DOI: 10.1016/S0040-4039(97)01425-1 *
KONECNY, G. E ET AL., MOL CANCER THER, vol. 12, 2013, pages 632 - 642
KONECNY, G. E. ET AL., THE LANCET ONCOLOGY, vol. 16, 2015, pages 686 - 694
KOOLE, K. ET AL.: "469", VIRCHOWS ARCH, 2016, pages S31 - S31
KUMAR ET AL., CELL REPORTS, vol. 23, 2018, pages 3798 - 3812
KUMAR ET AL., SCI REP, vol. 5, 2015, pages 15338
KUNII, K. ET AL., CANCER RES, vol. 68, 2008, pages 3549 - 3549
L. LACHMAN ET AL.: "The Theory and Practice of Industrial Pharmacy", 2013
LEE, S. ET AL., J CANCER RES CLIN, vol. 137, 2011, pages 1203 - 1211
LI, J.-L.LUO, EUROPEAN REVIEW FOR MEDICAL AND PHARMACOLOGICAL SCIENCES, vol. 24, 2020, pages 97 - 108
LI, Q. ET AL., THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 293, 2018, pages 6434 - 6448
LIU J ET AL., ONCOGENE, vol. 35, no. 14, 7 April 2016 (2016-04-07), pages 1750 - 9
LIU, J. ET AL., ONCOGENE, vol. 35, 2015, pages 1750 - 1759
MACK DANIEL J ET AL: "Distinct biological effects of golgicide a derivatives on larval and adult mosquitoes", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 22, no. 16, 5 July 2012 (2012-07-05), pages 5177 - 5181, XP028932819, ISSN: 0960-894X, DOI: 10.1016/J.BMCL.2012.06.076 *
MATSUKI M ET AL., CANCER MED, vol. 7, no. 6, June 2018 (2018-06-01), pages 2641 - 2653
MINGSHENG XIE ET AL: "Asymmetric Three-Component Inverse Electron-Demand Aza-Diels-Alder Reaction: Efficient Synthesis of Ring-Fused Tetrahydroquinolines", ANGEWANDTE CHEMIE INTERNATIONAL EDITION, vol. 49, no. 22, 14 April 2010 (2010-04-14), pages 3799 - 3802, XP055691591, ISSN: 1433-7851, DOI: 10.1002/anie.201000590 *
MORRISON, R. S. ET AL., CANCER RES, vol. 54, 1994, pages 2794 - 2799
NEMOTO, TOHASHI, KAKASHI, TJOHNSON, J. DHIROKAWA, K, PATHOBIOLOGY, vol. 65, 1997, pages 195 - 203
PEDERSEN, B. KFEBBRAIO, M. A, NATURE REVIEWS ENDOCRINOLOGY, vol. 8, 2012, pages 457 - 465
PENAULTLLORCA, F. ET AL., INT J CANCER, vol. 61, 1995, pages 170 - 176
PREUSSER, M. ET AL., LUNG CANCER, vol. 83, 2014, pages 83 - 89
ROBERTSON, A. G. ET AL., CELL, vol. 171, 2017, pages 540 - 556
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS
SANTHANA KUMAR, K ET AL., CELL REP, vol. 23, no. 3798-3812, 2018, pages e3798
SATO, T. ET AL., ONCOL REP, vol. 21, 2009, pages 211 - 216
SINGH, D. ET AL., SCIENCE, vol. 337, 2012, pages 1231 - 1235
ST BERNARD, R. ET AL., ENDOCRINOLOGY, vol. 146, 2005, pages 1145 - 1153
SU, NDU, X. LCHEN, FRONTIERS IN BIOSCIENCE, vol. 13, 2008, pages 2842 - 2865
TURNER, N.GROSE, NAT REV CANCER, 2010, pages 1 - 14
VERLINGUE, L. ET AL., EUROPEAN JOURNAL OF CANCER, vol. 87, 2017, pages 122 - 130
WEISS, J ET AL., SCI TRANSL MED, vol. 4, 2012
WEISS, J. ET AL., SCIENCE TRANSLATIONAL MEDICINE, 2010
WU SET, NAT COMMUN, vol. 10, no. 1, 12 February 2019 (2019-02-12), pages 720
WU, S. ET AL., NATURE COMMUNICATIONS, 2019, pages 1 - 12
YAMAOKA, T. ET AL., INT. J. MOL. SCI, vol. 19, 2018, pages 1 - 35
ZHANG, K. ET AL., CANCER RESEARCH, vol. 73, 2013, pages 1298 - 1307
ZHANG, K. Q. ET AL., CANCER RES, vol. 73, 2013, pages 1298 - 1307
ZHENG, NWEI, W. YWANG, Z. W. TRANSL, CANCER RES, vol. 5, 2016, pages 1 - 6

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024121437A1 (en) * 2022-12-09 2024-06-13 Universität Zürich Small-molecule inhibitors of the frs2-fgfr interaction

Similar Documents

Publication Publication Date Title
JP6963146B1 (en) Administration of KRAS inhibitors to treat cancer
TWI649081B (en) Method for treating solid tumors
Huang et al. Novel hybrid molecule overcomes the limited response of solid tumours to HDAC inhibitors via suppressing JAK1-STAT3-BCL2 signalling
JP5778735B2 (en) Use of pyrimidine derivatives to treat EGFR-dependent diseases or diseases that have acquired resistance to drugs targeting EGFR family members
CN101222850B (en) Methods for treating drug resistant cancer
Yuan et al. Discovery of new 4-indolyl quinazoline derivatives as highly potent and orally bioavailable P-glycoprotein inhibitors
WO2003101491A1 (en) PREVENTIVES AND/OR REMEDIES FOR SUBJECTS WITH THE EXPRESSION OR ACTIVATION OF Her2 AND/OR EGFR
US20230212126A1 (en) Small-molecule inhibitors of the frs2-fgfr interaction and their use in medicine, in the prevention and treatment of cancer
Fan et al. Discovery of Dioxino [2, 3-f] quinazoline derivative VEGFR-2 inhibitors exerting significant antipro-liferative activity in HUVECs and mice
JP2003535038A (en) 3-Heteroarylidenyl-2-indolinone compounds for modulating protein kinase activity and for use in cancer chemotherapy
WO2023105008A1 (en) Small-molecule inhibitors of the frs2-fgfr interaction
Listro et al. Urea-based anticancer agents. Exploring 100-years of research with an eye to the future
Moradi et al. Quinazoline-based VEGFR-2 inhibitors as potential anti-angiogenic agents: A contemporary perspective of SAR and molecular docking studies
Caligiuri et al. FT-6876, a potent and selective inhibitor of CBP/p300, is active in preclinical models of androgen receptor-positive breast cancer
JP7445609B2 (en) Thieno[2,3-B]pyridine derivatives as EPAC inhibitors and their pharmaceutical uses
US20230212124A1 (en) Small-molecule inhibitors of the frs2-fgfr interaction
EP3790541A1 (en) Inhibitors of the ras oncoprotein, methods of making and methods of use thereof
WO2024121437A1 (en) Small-molecule inhibitors of the frs2-fgfr interaction
WO2024003408A1 (en) Small-molecule inhibitors of the frs2-fgfr interaction
WO2021207352A1 (en) Compositions and methods for treatment of anticancer-drug resistant cancers
Dong et al. Design and synthesis of cantharidin derivative DCZ5418 as a TRIP13 inhibitor with anti-multiple myeloma activity in vitro and in vivo
CN100377709C (en) Methods of treating cancer and related methods
Zhang et al. YL143, a novel mutant selective irreversible EGFR inhibitor, overcomes EGFRL858R, T790M‐mutant resistance in vitro and in vivo
Karunakaran et al. Serine/threonine-protein kinase B-Raf inhibitors
Furtek TARGETING THE DNA-BINDING DOMAIN OF SIGNAL TRANSDUCER AND ACTIVATOR OF TRANSCRIPTION-3 (STAT3) WITH SMALL MOLECULE

Legal Events

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

Ref document number: 22834953

Country of ref document: EP

Kind code of ref document: A1