WO2023131677A1 - Compounds containing a hydroxyphenyl moiety and their use - Google Patents

Compounds containing a hydroxyphenyl moiety and their use Download PDF

Info

Publication number
WO2023131677A1
WO2023131677A1 PCT/EP2023/050228 EP2023050228W WO2023131677A1 WO 2023131677 A1 WO2023131677 A1 WO 2023131677A1 EP 2023050228 W EP2023050228 W EP 2023050228W WO 2023131677 A1 WO2023131677 A1 WO 2023131677A1
Authority
WO
WIPO (PCT)
Prior art keywords
hydroxyphenyl
urea
butan
dimethylphenyl
alkyl
Prior art date
Application number
PCT/EP2023/050228
Other languages
French (fr)
Inventor
Eric Chevet
Dimitrios DOULTSINOS
Leif A. Eriksson
Antonio CARLESSO
Sébastien SUERON
Xavier GUILLORY
Timothy LANGLAIS
François-Hugues PORÉE
Diana PELIZZARI-RAYMUNDO
François CARREAUX
Nicolas GOUAULT
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Universite De Rennes
Centre National De La Recherche Scientifique
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 INSERM (Institut National de la Santé et de la Recherche Médicale), Universite De Rennes, Centre National De La Recherche Scientifique filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2023131677A1 publication Critical patent/WO2023131677A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/60Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings condensed with carbocyclic rings or ring systems
    • C07D277/62Benzothiazoles
    • C07D277/68Benzothiazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • C07D277/82Nitrogen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C275/00Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups
    • C07C275/04Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of urea groups bound to acyclic carbon atoms
    • C07C275/20Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of urea groups bound to acyclic carbon atoms of an unsaturated carbon skeleton
    • C07C275/24Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of urea groups bound to acyclic carbon atoms of an unsaturated carbon skeleton containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C275/00Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups
    • C07C275/28Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of urea groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/38Nitrogen atoms

Definitions

  • the present invention relates to urea, oxalamide, amide, thiourea, carbamate or ester compounds, in particular urea compounds, containing a hydroxyphenyl or phenyl moiety, in particular a hydroxyphenyl moiety, including their pharmaceutically acceptable salts and solvates which are useful as sensitizers for chemotherapy of cancer cells, particularly in glioblastoma, and are useful as therapeutic compounds, paraticularly in the treatment of cancers that may be treated by alkylating agents, such as temozolomide.
  • Glioblastoma is the most common primary central nervous system (CNS) tumour, displaying high levels of aggressiveness, recurrence and heterogeneity; traits that contribute to a dismal prognosis of an average of 1.5 year survival post diagnosis.
  • the standard of care comprises maximal safe resection of the tumour followed by a combination of irradiation and chemotherapy with the alkylating agent temozolomide; however, all patients succumb to the disease (R. Stupp et al., N. Engl. J. Med., 2005, 352, 987-996).
  • GB cells as with most solid tumours, survives in a hostile environment which includes hypoxia, nutrient shortage, necrosis and immune infiltration, as well as having to cope with a high metabolic turnover and protein synthesis demand (D. Doultsinos et al., SLAS Discov. Adv. Life Sci. R&D, 2017, 22, 787-800).
  • URR Unfolded Protein Response
  • J. Obacz et al., Sci. Signal., 2017, 10, eaal2323 J. Obacz et al., Sci. Signal., 2017, 10, eaal2323
  • Inositol Requiring Enzyme 1 a major Unfolded Protein Response (UPR) transducer
  • URR Unfolded Protein Response
  • TNBC Triple Negative Breast Cancer
  • IRE1 activity inhibition can be mediated by compounds targeting either the ATP-binding kinase domain or the RNase domain.
  • Direct RNase pharmacological inhibitors include 4 ⁇ 8c, STF-083010, toyocamycin and a series of MKC compounds, all relying on a hydroxy-aryl aldehyde (HAA) motif, whilst kinase pharmacological inhibitors that in turn inhibit the RNase include amongst others 1-(4-(8-amino-3-isopropylimidazo[1,5-a]pyrazin-1- yl)naphthalen-1-yl)-3-(3-(trifluoro-methyl)phenyl)urea (CAS# 1414938-21-8), 1-(4-(8- amino-3-(tert-butyl)imidazo[1,5-a]pyrazin-1-yl)naphthalen-1-yl)-3-(3- (trifluoromethyl)phenyl)urea (CAS
  • the invention therefore relates to compounds of general Formula I, their pharmaceutically acceptable salts and solvates as well as methods of use of such compounds or compositions comprising such compounds as sensitizers for chemotherapy of malignant tumors.
  • the invention provides compounds of general Formula I: I, a pharmaceutically acceptable salt or a solvate thereof, wherein A is selected from –NH–, –N(Me)– and –O–; Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O–, –CH2– and –C(O)NH— or is a single bond; Z is O or S; R 1 and R 2 are independently selected from H and OH, with the proviso that at least one of R 1 and R 2 is H and that R 1 and R 2 are not both H; Cy is selected from: - R 3 is selected from H, C1-C4-alkyl and halogen; R 4 is selected from H, C1-C4-alkyl and C
  • the invention also relates to a compound of Formula I: I, a pharmaceutically acceptable salt or solvate thereof wherein A is selected from –NH–, –N(Me)– and –O–; Y is selected from –NH–, –N(Me)–, –NH–CH 2 –, –O–, –CH 2 – and –C(O)NH– or is a single bond; Z is O or S; R 1 and R 2 are independently selected from H, OH and OMe, with the proviso that at least one of R 1 and R 2 is H; Cy is selected from: - wherein R 3 is selected from H, C1-C4-alkyl, and halogen; R 4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; R 5 is H or C1-C4-alkyl; R 6 is selected from H, C1-C4-alkyl, C3-C4-cycloalkyl, hal
  • the invention futher relates to compounds of Formula I or their pharmaceutically acceptable salts and solvates for use in increasing the sensitivity of cancer cells to an anticancer agent, particularly an alkylating agent, in a treatment of cancer, particularly glioblastoma, triple- negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer.
  • an anticancer agent particularly an alkylating agent
  • the present invention provides a pharmaceutical composition comprising at least one compound of Formula I as defined above, or a pharmaceutically acceptable salt or solvate thereof, and at least one pharmaceutically acceptable carrier, diluent, excipient and/or adjuvant.
  • the invention relates to a hydroxyphenyl compound selected from the group consisting of: 1-(2,5-dimethylphenyl)-3-(6-(3-hydroxyphenyl)pyridin-3-yl)urea; 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)-1H-pyrrol-3-yl)urea; 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)-2-(prop-1-en-2-yl)-1H-indol-3-yl)urea; 1-(3,5-difluorophenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(3-methyl-5-(trifluoromethyl)phenyl)urea; and 1-(3,5-bis(trifluoromethyl)phenyl)-3-(4-(4-(4-
  • the invention relates to a pharmaceutical composition comprising a hydroxyphenyl compound as defined above, or a pharmaceutically acceptable salt or solvate thereof, and at least one pharmaceutically acceptable excipient.
  • the invention relates to a hydroxyphenyl compound as defined above, in combination with an anticancer agent, for use in treating cancer.
  • the invention relates to a compound comprising a hydroxyphenyl moiety as defined above, for use in increasing the sensitivity of cancer cells to an anticancer agent in a treatment of cancer.
  • DETAILED DESCRIPTION OF THE INVENTION As detailed above, the invention relates to compounds of Formula I, as well as their pharmaceutically acceptable salts or solvates.
  • Preferred compounds of Formula I or pharmaceutically acceptable salts or solvates thereof are those wherein one or more of A, L, Y, Z, R 1 , R 2 and Cy are defined as follows: A is selected from –NH–, –N(Me)– and –O–; in particular A is –NH–; 5
  • Y is selected from —NH–, –N(Me)–, –NH–CH 2 –, –O–, –CH 2 – and –C(O)NH–; in particular Y is selected from –NH–, –N(Me)–, –NH–CH 2 –, –O– and –CH 2 –; more particularly Y is selected from –NH–, –NH–CH2–, –O– and –CH2–; still more particularly Y is–NH–; Z is O or S; in particular Z is O; R 1 and R 2 are independently selected from H and OH, with the proviso that at least one of R 1 and R 2 is H and that R 1 and R 2 are not both H; in particular R 1 is OH and R 2 is H; Cy is selected from: - wherein R 3 is selected from H, C1-C4-alkyl and halogen; in particular R 3 is selected from H, C1-C4-alkyl and F; more particularly R
  • Halogens include a fluorine atom, an iodine atom, a chlorine atom and a bromine atom.
  • C1-C4-alkyl include butyl, in particular n-butyl, isobutyl, sec-butyl or tert-butyl; propyl, in particular n-propyl or isopropyl; ethyl or methyl.
  • C3-C4-cycloalkyl include cyclopropyl and cyclobutyl.
  • the compound of formula I may be in racemic or optically active form. In one embodiment, the compound of formula I may be in racemic form. In one embodiment, the compound of formula I may be in optically active form.
  • the compounds of Formula I are those wherein A is –NH–. In one embodiment, the compounds of Formula I are those wherein A is –N(Me)–. In one embodiment, the compounds of Formula I are those wherein A is –O–. In one embodiment, the compounds of Formula I are those wherein L is selected from In one embodiment, the compounds of Formula I are those wherein L is selected from In one embodiment, the compounds of Formula I are those wherein L is selected from In one embodiment, the compounds of Formula I are those wherein L is selected from In one embodiment, the compounds of Formula I are those wherein L is selected from In one embodiment, the compounds of Formula I are those wherein L is . In one embodiment, the compounds of Formula I are those wherein L is . In one embodiment, the compounds of Formula I are those wherein L is . In one embodiment, the compounds of Formula I are those wherein L is .
  • the compounds of Formula I are those wherein L is . In one embodiment, the compounds of Formula I are those wherein L is . In one embodiment, the compounds of Formula I are those wherein L is . In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein L is .
  • the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein Y is selected from –NH– , –N(Me)–, –NH–CH2–, –O– and –CH2–; in particular Y is selected from –NH–, –NH–CH2– , –O– and –CH 2 –.
  • the compounds of Formula I are those wherein Y is –NH–. In one embodiment, the compounds of Formula I are those wherein Y is –N(Me)–. In one embodiment, the compounds of Formula I are those wherein Y is –NH–CH 2 –. In one embodiment, the compounds of Formula I are those wherein Y is –O–. In one embodiment, the compounds of Formula I are those wherein Y is –CH2–. In one embodiment, the compounds of Formula I are those wherein Z is O. In one embodiment, the compounds of Formula I are those wherein Z is S. In one embodiment, the compounds of Formula I are those wherein R 1 is OH and R 2 is H.
  • the compounds of Formula I are those wherein R 1 is H and R 2 is OH.
  • the compounds of Formula I are those wherein wherein R 3 is selected from H, C1-C4-alkyl and halogen; in particular R 3 is selected from H, C1-C4-alkyl and F; more particularly R 3 is H or C1-C4-alkyl; still more particularly R 3 is Me or t-Bu;
  • R 4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; in particular R 4 is selected from H, C1-C3-alkyl and cyclopropyl; more particularly R 4 is H or C1- C3-alkyl; still more particularly R 4 is H;
  • R 5 is H or C1-C4-alkyl; in particular R 5 is H or C1-C3-alkyl; more particularly R 5 is H or Me; still more particularly R 5 is H;
  • R 6 is selected from H, C1-C4-al
  • the compounds of Formula I are those wherein wherein R 11 and R 12 are independently selected from hydrogen and C1-C4-alkyl; in particular R 11 and R 12 are independently selected from hydrogen and C1-C3- alkyl; more particularly R 11 and R 12 are H or Me; still more particularly R 11 and R 12 are Me.
  • the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein Cy is selected from In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those of Formula II: II, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, Y and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula IIa: IIa, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A and Y are as defined above with respect to Formula I and any of its embodiments.
  • Particular compounds of Formula IIa are those wherein R 1 , R 2 , L, A and Y are defined as follows: R 1 and R 2 are independently selected from H and OH, with the proviso that at least one of R 1 and R 2 is H and that R 1 and R 2 are not both H; in particular R 1 is OH and R 2 is H;
  • the compounds of Formula I are those of Formula IIb: IIb, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A, Y and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula IIc: IIc, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula III: III, or pharmaceutically acceptable salts or solvates thereof, wherein L, A, Z, Y and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula IIIa: IIIa, or pharmaceutically acceptable salts or solvates thereof, wherein L, A, Z and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula IIIb: IIIb, or pharmaceutically acceptable salts or solvates thereof, wherein A, Z, Y and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula IIIc: IIIc, or pharmaceutically acceptable salts or solvates thereof, wherein A, Z and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula IV: IV, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, Y and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula IVa: IVa, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula IVb: IVb, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , Y and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula IVc: IVc, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula V: V, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula Va: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L and A are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula Vb: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula Vc: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and A are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula VI: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula VIa: VIa, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , and L are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula VIb: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula VIc: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula VId: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 and R 2 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula VII: VII, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, Z, Y, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula VIIa: VIIa, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, Z and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula VIIb: VIIb, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A, Z, Y, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula VIIc: VIIc, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A, Z and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula VIII: VIII, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, Y, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • R 1 and R 2 are independently selected from H and OH, with the proviso that at least one of R 1 and R 2 is H and that R 1 and R 2 are not both H; in particular R 1 is OH and R 2 is H; A is selected from —NH–, –N(Me)– and –O–; in particular A is –NH–; 5 more particularly L is selected from , Y is selected from –NH–, –N(Me)–, –NH–CH 2 –, –O–, –CH 2 – and –C(O)NH–; in particular Y is selected from –NH–, –N(Me)–, –NH–CH 2 –, –O– and –CH 2 –; more particularly Y is
  • the compounds of Formula I are those of Formula VIIIb: VIIIb, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A, Y, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula VIIIc: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula IX: IX, or pharmaceutically acceptable salts or solvates thereof, wherein L, A, Y, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula IXa: or pharmaceutically acceptable salts or solvates thereof, wherein L, A and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula IXb: IXb, or pharmaceutically acceptable salts or solvates thereof, wherein A, Y, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula IXc: or pharmaceutically acceptable salts or solvates thereof, wherein A and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula X: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula Xa:
  • the compounds of Formula I are those of Formula Xb: Xb, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula Xc: Xc, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and A are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula XI: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula XIa: XIa, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L and A are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula XIb:
  • the compounds of Formula I are those of Formula XIc: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and A are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula XII: XII, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula XIIa: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula XIIb: XIIb, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula XIIc: XIIc, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and A are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula XIII: XIII, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula XIIIa: XIIIa, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L and A are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula XIIIb: XIIIb, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula XIIIc: XIIIc, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and A are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula XIV: XIV, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula XIVa: XIVa, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and L are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula XIVb: XIVb, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds of Formula I are those of Formula XIVc: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 and R 2 are as defined above with respect to Formula I and any of its embodiments.
  • Particularly preferred compounds of the invention are those listed in Table 1 hereafter: Table 1
  • Table 1 The invention further concerns hydroxyphenyl compounds listed in Table 1b hereafter.
  • hydroxyphenyl compound as used herein means a compound comprising a hydroxyphenyl moiety.
  • Table 1b The compounds of the invention can be prepared by different ways with reactions known by the person skilled in the art. Typical routes of synthesis are described thereafter The compounds of the invention (i.e.
  • the compounds of Formula I and its subformulae as described above and the hydroxyphenyl compounds of Table 1b above) are indeed capable of inhibiting IRE RNase activity and sensitizing cancer cells to anticancer drugs. They further have the advantage of sensitizing cancer cells, in particular GGM cells to anticancer drugs, in particular alkylating agents.
  • the invention thus also provides the use of the compounds of the invention, or pharmaceutically acceptable salts or solvates thereof, as sensitizers for chemotherapy of cancer cells. Accordingly, the invention relates to the use of compounds of the invention, or pharmaceutically acceptable salts or solvates thereof, for the treatment of cancer, particularly as sensitizers for chemotherapy of cancer cells.
  • the compounds of the invention may be used as inhibitors of IRE1 RNase activity and have the potentential of increasing the sensitivity of cancer cells to an anticancer agent in a treatment of cancer.
  • the inventors think the specific structure of the compounds of the invention allow them to interact with the ATP kinase binding pocket of IRE1.
  • the compounds of the invention are able to inhibit IRE1 RNase activity and to increase the sensitivity of cancer cells to an anticancer agent in a treatment of cancer, in particular glioblastoma, triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer.
  • the compounds of the invention as defined above can thus be used for treating cancer, particularly as sensitizers for chemotherapy of cancer cells, aiming at improving the chemotherapy effect of the cancer treatment, preventing tolerance and decreasing toxicity and adverse effects.
  • the invention thus relates to a compound of Formula I: I, a pharmaceutically acceptable salt or solvate thereof, wherein A is selected from –NH–, –N(Me)– and –O–; , Y is selected from –NH–, –N(Me)–, –NH–CH 2 –, –O–, –CH 2 – and –C(O)NH– or is a single bond; Z is O or S; R 1 and R 2 are independently selected from H and OH, with the proviso that at least one of R 1 and R 2 is H; Cy is selected from: - R 3 is selected from H, C1-C4-alkyl, and halogen; R 4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; R 5 is H or C1-C4-alkyl; R 6 is selected from H, C1-C4-alkyl, C3-C4-cycloalkyl, halogen, C
  • the compound for use of formula I may be in racemic or optically active form. In one embodiment, the compound for use of formula I may be in racemic form. In one embodiment, the compound for use of formula I is in optically active form.
  • particular compounds for use according to the invention are compounds of formula I, or pharmaceutically acceptable salts or solvates thereof, wherein A, L, Y, Z, R 1 , R 2 and Cy are defined as follows: A is selected from –NH–, –N(Me)– and –O–; in particular A is –NH–; 5 more particularly L is selected from , Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O–, –CH2– and –C(O)NH– or is a single bond; in particular Y is selected from –NH–, –N(Me)–, –NH–CH 2 –, –O–, –CH 2 – and – C(O)NH— or is a
  • the compounds for use of Formula I are those wherein A is –NH–. In one embodiment, the compounds for use of Formula I are those wherein A is –N(Me)–. In one embodiment, the compounds for use of Formula I are those wherein A is –O–. In one embodiment, the compounds for use of Formula I are those wherein L is selected , In one embodiment, the compounds of Formula I are those wherein . In one embodiment, the compounds for use of Formula I are those wherein Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O– and –CH2–; in particular Y is selected from –NH– , –NH–CH 2 –, –O– and –CH 2 –.
  • the compounds for use of Formula I are those wherein Y is –NH–. In one embodiment, the compounds for use of Formula I are those wherein Y is –N(Me)–. In one embodiment, the compounds for use of Formula I are those wherein Y is –NH–CH2– . In one embodiment, the compounds for use of Formula I are those wherein Y is –O–. In one embodiment, the compounds for use of Formula I are those wherein Y is –CH2–. In one embodiment, the compounds for use of Formula I are those wherein Z is O. In one embodiment, the compounds for use of Formula I are those wherein Z is S. In one embodiment, the compounds for use for use of Formula I are those wherein R 1 is OH and R 2 is H.
  • the compounds for use of Formula I are those wherein R 1 is H and R 2 is OH. In one embodiment, the compounds for use of Formula I are those of Formula II: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, Y and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula IIa: IIa, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula IIb: IIb, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A, Y and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula IIc: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula III: or pharmaceutically acceptable salts or solvates thereof, wherein L, A, Z, Y and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula IIIa: IIIa, or pharmaceutically acceptable salts or solvates thereof, wherein L, A, Z and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula IIIb: IIIb, or pharmaceutically acceptable salts or solvates thereof, wherein A, Z, Y and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula IIIc: IIIc, or pharmaceutically acceptable salts or solvates thereof, wherein A, Z and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula IV: IV, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, Y and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula IVa: IVa, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula IVb: IVb, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , Y and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula IVc: IVc, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula V: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula Va: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L and A are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula Vb: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula Vc: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and A are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula VI: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula VIa: VIa, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , and L are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula VIb: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula VIc: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula VId: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 and R 2 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula VII: VII, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, Z, Y, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula VIIa: VIIa, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, Z and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula VIIb: VIIb, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A, Z, Y, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula VIIc: VIIc, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A, Z and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula VIII: VIII, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, Y, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • R 1 and R 2 are independently selected from H and OH, with the proviso that at least one of R 1 and R 2 is H; in particular R 1 is OH and R 2 is H; A is selected from —NH–, –N(Me)– and –O–; in particular A is –NH–; 5 more particularly L is selected from , Y is selected from –NH–, –N(Me)–, –NH–CH 2 –, –O–, –CH 2 – and –C(O)NH– or is a single bond; in particular Y is selected from –NH–, –N(Me)–, –NH–CH 2 –, –O– and –CH 2 –; more particularly Y is selected from
  • the compounds for use of Formula I are those of Formula VIIIb: VIIIb, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A, Y, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula VIIIc: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula IX: IX, or pharmaceutically acceptable salts or solvates thereof, wherein L, A, Y, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula IXa: or pharmaceutically acceptable salts or solvates thereof, wherein L, A and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula IXb: IXb, or pharmaceutically acceptable salts or solvates thereof, wherein A, Y, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula IXc: or pharmaceutically acceptable salts or solvates thereof, wherein A and Y are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula X: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula Xa:
  • the compounds for use of Formula I are those of Formula Xb: Xb, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula Xc: Xc, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and A are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula XI: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula XIa: XIa, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L and A are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula XIb:
  • the compounds for use of Formula I are those of Formula XIc: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and A are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula XII: XII, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, Y, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula XIIa: or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, Y, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula XIIb: XIIb, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula XIIc: XIIc, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and A are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula XIII: XIII, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, A, Y, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula XIIIa: XIIIa, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L and A are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula XIIIb: XIIIb, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , A, R 3 , R 4 , R 5 , R 6 and R 7 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula XIIIc: XIIIc, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and A are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula XIV: XIV, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 , L, and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula XIVa: XIVa, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and L are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use of Formula I are those of Formula XIVb: XIVb, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 , R 2 and Cy are as defined above with respect to Formula I and any of its embodiments.
  • the compoundsfor use of Formula I are those of Formula XIVc: XIVc, or pharmaceutically acceptable salts or solvates thereof, wherein R 1 and R 2 are as defined above with respect to Formula I and any of its embodiments.
  • the compounds for use according to the invention therefore include compounds of Formula I and subformulae as defined above, in particular compounds of Table 2 below.
  • Table 2 The invention also concerns hydroxyphenyl compounds as listed in Table 2b hereafter, in combination with an anticancer agent, for use in treating cancer: Table 2b
  • the invention also relates to the compounds of the invention as described above for use in increasing the sensitivity of cancer cells to an anticancer agent in a treatment of cancer.
  • the compounds for use according to the invention are selected from compounds 2, 3, 4, 5, 6, 7, 9, 11, 12, 14, 16, 17, 18, 20, 22, 23, 24, 27, 28, S-28, R-28, 30, 31, 32, 40, 41, 42, 43, 44, 46, 47, 50, 52, 54, 57, 58, 59, 63, 64 and 66 of Tables 2 and 2b above. In one embodiment, the compounds for use according to the invention are selected from compounds 2, 3, 4, 5, 6, 7, 9, 11, 12, 14, 16, 17, 18, 20, 22, 23, 24, 27, 28, S-28, R-28, 30, 31, 32, 40, 41, 42, 43, 44, 46, 47, 50, 52, 54, 63, 64 and 66 of Table 2 above.
  • the compounds for use according to the invention are selected from compounds 57, 58 and 59 of Table 2b above. In one embodiment, the compounds for use according to the invention are selected from compounds 28, 33, 34, 35, 36, 37 and 38 of Table 2 above, in particular compounds 28 and 33 of Table 2 above. In one embodiment, the compound for use according to the invention is compound 28 of Table 2 above.
  • Treatment of cancer, in particular chemotherapy is a type of treatment that uses one or more anticancer agents.
  • Anticancer agents, or cytotoxic agents, within the meaning of the present invention include, but are not limited to, alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents.
  • the anticancer agent is an alkylating agent.
  • Preferred alkylating agent is temozolamide.
  • the compounds of the invention are therefore useful in the treatment of cancers, and particularly cancers that may be treated by anticancer agents selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly cancers that may be treated by alkylating agents.
  • Cancers that may be treated by anticancer agents selected from alkylating agents, anti- microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, within the meaning of the present invention include, but are not limited to, glioblastoma, triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer.
  • a preferred cancer that may be treated by alkylating agents is glioblastoma.
  • the invention thus also relates to a compound of the present invention, i.e.
  • an anticancer agent in particular an anticancer agent selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly an alkylating agent, even more particularlry temozolamide, for use in treating cancer, preferably cancers that may be treated by anticancer agents selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly a cancer selected from glioblastoma (GB), triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer, even more particularly cancers that may be treated by alkylating agents, still more
  • the invention also relates to a method of treating cancer, in particular cancers that may be treated by anticancer agents selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly a cancer selected from glioblastoma, triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer, even more particularly cancers that may be treated by alkylating agents, still more particularly glioblastoma, comprising the administration of a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt or solvate thereof, in combination with an anticancer agent, in particular an anticancer agent selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly an alkylating agent, even more particularly temozolamide, to a patient in need of such
  • the patient is a warm-blooded animal, more preferably a human.
  • the cancers that may be treated by an alkylating agent are preferably those defined above.
  • the invention further provides the use of a compound of the present invention, or a pharmaceutically acceptable salt or solvates thereof, in combination with an anticancer agent, in particular an anticancer agent selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly an alkylating agent, even more particularly temozolamide, for the manufacture of a medicament for use in treating cancer, in particular cancers that may be treated by anticancer agents selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly a cancer selected from glioblastoma, triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer
  • the patient is a warm-blooded animal, more preferably a human.
  • the cancers that may be treated by an alkylating agent are preferably those defined above.
  • a compound of the invention or a pharmaceutically acceptable salt or solvate thereof and the anticancer agent are administered to the patient in the same preparation or in a separate form, either simultaneously or sequentially, for the treatment of cancer.
  • a compound of the present invention for use in increasing the sensitivity of cancer cells to an anticancer agent in a treatment of cancer.
  • the invention thus relates to a compound of the present invention, i.e.
  • the invention also relates to a method for increasing the sensitivity of cancer cells to an anticancer agent, in particular an anticancer agent selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly an alkylating agent, even more particularly temozolomide, in a treatment of cancer, in particular cancers that may be treated by anticancer agents selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly a cancer selected from glioblastoma, triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer, even more particularly cancers that may be treated by alkylating agents, still more particularly glioblastoma, comprising the administration of a therapeutically effective amount of a compound of the present invention, i.e.
  • an anticancer agent in particular an anticancer agent selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly an alkylating agent, even more particularly temozolamide, to a patient in need thereof.
  • an anticancer agent selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly an alkylating agent, even more particularly temozolamide, to a patient in need thereof.
  • the patient is a warm-blooded animal, more preferably a human.
  • the invention further provides the use of a compound of the present invention, i.e.
  • an anticancer agent selected from alkylating agents, anti- microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly an alkylating agent, even more particularly temozolomide, in a treatment of cancer, in particular cancers that may be treated by anticancer agents selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly a cancer selected from glioblastoma, triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer, even more particularly cancers that may be treated by alkyl
  • a compound of the present invention i.e. a compound of Formula I and any of its embodiments, or any of its subformulae as defined above, or a hydrophenyl compound of Table 2b, or a pharmaceutically acceptable salt or solvate thereof, for inhibiting IRE1 RNase activity, in a patient in need of such treatment, comprising administering to said patient an effective amount of a compound of the present invention, or a pharmaceutically acceptable salt or solvate thereof.
  • the invention also provides a method for inhibiting IRE1 RNase activity, in a patient in need of such treatment, which comprises the step of administering to said patient an effective amount of a compound of the present invention, i.e.
  • the patient is a warm blooded animal, and even more preferably a human.
  • the compound of the invention or the compound for use according to the invention may be administered as a pharmaceutical formulation in a therapeutically effective amount by any of the accepted modes of administration, preferably by intravenous or oral route.
  • Therapeutically effective amount ranges are typically from 0.1 to 50 000 ⁇ g/kg of body weight daily, preferably from 1000 to 40000 ⁇ g/kg of body weight daily, depending upon numerous factors such as the severity of the disease to be treated, the age and relative health of the subject, the potency of the compound, the route and the form of administration, the indication towards which the administration is directed, and the preferences and experience of the medical practitioner involved.
  • One of ordinary skill in the art of treating such diseases will be able in reliance upon personal knowledge, to ascertain a therapeutically effective amount of the anticancer agent of the present invention for a given cancer.
  • the compounds of the invention, their pharmaceutical acceptable salts or solvates may be administered as part of a combination therapy.
  • compositions and medicaments which contain, in addition to a compound of the present invention, a pharmaceutically acceptable salt or solvate thereof as active ingredient, additional therapeutic agents and/or active ingredients.
  • Such multiple drug regimens often referred to as combination therapy, may be used in the treatment of cancer, particularly those defined above.
  • the methods of treatment and pharmaceutical compositions of the present invention may employ the compounds of the invention or their pharmaceutical acceptable salts or solvates thereof in the form of monotherapy, but said methods and compositions may also be used in the form of multiple therapy in which one or more compounds of the invention or their pharmaceutically acceptable salts or solvates are co-administered in combination with one or more other therapeutic agents.
  • Such additional therapeutic agents include, but are not limited to, alkylating agents, and preferably temozolomide.
  • the methods of treatment and pharmaceutical compositions of the present invention may employ the compounds of the present invention, or their pharmaceutical acceptable salts or solvates thereof, in combination with radiation therapy.
  • the compounds of the invention, their pharmaceutical acceptable salts or solvates may be administered in combination with radiation therapy.
  • radiation therapies include, but are not limited to, external beam radiation therapy, brachytherapy and systemic radioisotope therapy.
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the present invention, i.e. a compound of Formula I and any of its embodiments, or any of its subformulae as defined above, or a hydrophenyl compound of Table 2b, or a pharmaceutically acceptable salt or solvate thereof, and at least one pharmaceutically acceptable carrier, diluent, excipient and/or adjuvant.
  • the invention also covers pharmaceutical compositions which contain, in addition to a compound of the present invention, a pharmaceutically acceptable salt or solvate thereof as active ingredient, additional therapeutic agents and/or active ingredients, in particular an anticancer agent.
  • the invention also provides a compound of the invention, or a pharmaceutically acceptable salt or solvate thereof, for use in a therapeutic treatment in humans or animals.
  • Another object of this invention is a medicament comprising at least one compound of the invention, or a pharmaceutically acceptable salt or solvate thereof, as active ingredient.
  • the compounds of the invention may be formulated as a pharmaceutical preparation comprising at least one compound of the invention and at least one pharmaceutically acceptable carrier, diluent, excipient and/or adjuvant, and optionally one or more further pharmaceutically active compounds.
  • such a formulation may be in a form suitable for oral administration, for parenteral administration (such as by intravenous, intramuscular or subcutaneous injection or intravenous infusion), for topical administration (including ocular), cerebral administration, for administration by inhalation, by a skin patch, by an implant, by a suppository, etc.
  • parenteral administration such as by intravenous, intramuscular or subcutaneous injection or intravenous infusion
  • topical administration including ocular
  • cerebral administration for administration by inhalation, by a skin patch, by an implant, by a suppository, etc.
  • suitable administration forms — which may be solid, semi-solid or liquid, depending on the manner of administration – as well as methods and carriers, diluents and excipients for use in the preparation thereof, will be clear to the skilled person; reference is made to the latest edition of Remington’s Pharmaceutical Sciences.
  • the compound of the invention or a pharmaceutical composition comprising a compound of the invention can be administered orally in the form of tablets, coated tablets, pills, capsules, soft gelatin capsules, oral powders, granules, ovules, elixirs, solutions or suspensions, which may contain flavouring or colouring agents, for immediate-, delayed-, modified-, sustained-, pulsed- or controlled-release applications.
  • the tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, a disintegrant such as starch (preferably corn, potato or tapioca starch), sodium starch glycollate, croscarmellose sodium and certain complex silicates, a binder such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxypropylcellulose (HPC), sucrose, gelatin and acacia, a lubricant such as magnesium stearate, stearic acid, glyceryl behenate.
  • solid compositions of a similar type may also be employed as fillers in hard gelatin capsules.
  • Preferred excipients in this regard include lactose, saccharose, sorbitol, mannitol, potato starch, corn starch, amylopectin, cellulose derivatives or gelatin.
  • Hard gelatin capsules may contain granules of the compound of the invention.
  • Soft gelatin capsules may be prepared with capsules containing the compound of the invention, vegetable oil, waxes, fat, or other suitable vehicle for soft gelatin capsules.
  • the acceptable vehicle can be an oleaginous vehicle, such as a long chain triglyceride vegetable oil (e.g. corn oil).
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water may contain the active ingredient in a mixture with dispersing agents, wetting agents, and suspending agents and one or more preservatives. Additional excipients, for example sweetening, flavouring and colouring agents, may also be present. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Liquid dosage forms for oral administration may include pharmaceutically acceptable, solutions, emulsions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art, such as water or an oleaginous vehicle. Liquid dosage form may be presented as a dry product for constitution with water or other suitable vehicle before use.
  • compositions may also comprise adjuvants, such as wetting agents, emulsifying and suspending agents, complexing agents such as 2-hydroxypropyl-beta-cyclodextrin, sulfobutylether-beta-cylodextrin, and sweetening, flavouring, perfuming agents, colouring matter or dyes with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
  • adjuvants such as wetting agents, emulsifying and suspending agents, complexing agents such as 2-hydroxypropyl-beta-cyclodextrin, sulfobutylether-beta-cylodextrin, and sweetening, flavouring, perfuming agents, colouring matter or dyes with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
  • These compositions may be preserved by the addition of an anti-
  • the compound of the invention may be milled using known milling procedures such as wet milling to obtain a particle size appropriate for tablet formation and for other formulation types.
  • examples of such administration include one or more of: intravenously, intraarterially, intraperitoneally, intrathecally, intraventricularly, intraurethrally, intrasternally, intracranially, intramuscularly or subcutaneously administering the agent; and/or by using infusion techniques.
  • the compound of the invention can be administered via the parenteral route with a readily available or a depot-type formulation.
  • compositions for the parenteral administration of a readily available formulation may be in the form of a sterile injectable aqueous or oleagenous solution or suspension in a non-toxic parenterally-acceptable diluent or solvent and may contain formulatory agents such as suspending, stabilising dispersing, wetting and/or complexing agents such as cyclodextrin e.g. 2-hydroxypropyl-beta-cyclodextrin, sulfobutylether-beta- cylodextrin.
  • the depot-type formulation for the parenteral administration may be prepared by conventional techniques with pharmaceutically acceptable excipient including without being limited to, biocompatible and biodegradable polymers (e.g.
  • poly( ⁇ -caprolactone), poly(ethylene oxide), poly(glycolic acid), poly[(lactic acid)-co-(glycolic acid)...)], poly(lactic acid)...), non-biodegradable polymers e.g. ethylene vinylacetate copolymer, polyurethane, polyester(amide), polyvinyl chloride
  • aqueous and non-aqueous vehicles e.g. water, sesame oil, cottonseed oil, soybean oil, castor oil, almond oil, oily esters, ethyl alcohol or fractionated vegetable oils, propylene glycol, DMSO, THF, 2-pyrrolidone, N- methylpyrrolidinone, N-vinylpyrrolidinone... ).
  • the active ingredient may be in dry form such as a powder, crystalline or freeze-dried solid for constitution with a suitable vehicle.
  • suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.
  • the compound of the present invention can be administered intranasally or by inhalation and is conveniently delivered in the form of a dry powder inhaler or an aerosol spray presentation from a pressurised container, pump, spray or nebuliser with the use of a suitable propellant, e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, (for example from Ineos Fluor), carbon dioxide or other suitable gas.
  • a suitable propellant e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, (for example from Ineos Fluor), carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the pressurised container, pump, spray or nebuliser may contain a solution or suspension of the active compound.
  • Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated to contain a powder mix of the compound and a suitable powder base such as lactose or starch.
  • a suitable powder base such as lactose or starch.
  • the compound or salt of the invention is in a particle-size-reduced form, and more preferably the size-reduced form is obtained or obtainable by micronisation.
  • the preferable particle size of the size-reduced (e.g. micronised) compound or salt or solvate is defined by a D50 value of about 0.5 to about 50 microns (for example as measured using laser diffraction).
  • the compound of the present invention can be administered in the form of a suppository or pessary, or it may be applied topically in the form of a gel, hydrogel, lotion, solution, cream, ointment or dusting powder.
  • the compound of the present invention may also be dermally or transdermally administered, for example, by the use of a skin patch. They may also be administered by the pulmonary or rectal routes. It may also be administered by the ocular route.
  • the compound can be formulated as micronised suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride. Alternatively, it may be formulated in an ointment such as petrolatum.
  • the agent of the present invention can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • it can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • any reference to compounds of the invention herein means the compounds as such as well as their pharmaceutically acceptable salts and solvates.
  • the terms used are to be construed in accordance with the following definitions, unless indicated otherwise.
  • the term “unsubstituted” as used herein means that a radical, a group or a residue carries no substituents.
  • substituted means that a radical, a group or a residue carries one or more substituents.
  • halo refers to the atoms of the group 17 of the periodic table (halogens) and includes in particular fluorine, chlorine, bromine and iodine atom. Preferred halo groups in the context of the invention are fluoro and iodo, fluoro being particularly preferred.
  • alkyl by itself or as part of another substituent refers to a hydrocarbyl group of Formula C n H 2n+1 wherein n is a number greater than or equal to 1. Alkyl groups may thus comprise 1 or more carbon atoms and generally, according to this invention comprise from 1 to 12, more preferably from 1 to 8 carbon atoms, and still more preferably from 1 to 6 carbon atoms.
  • Alkyl groups within the meaning of the invention may be linear or branched.
  • alkyl groups include but are not limited to methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, neopenyl, isopentyl, sec-pentyl, tert-pentyl, n-hexyl, neohexyl, isohexyl, sec-hexyl and tert-hexyl.
  • alkyl groups in the context of the invention include methyl, ethyl, isopropyl and tert-butyl.
  • haloalkyl alone or in combination, refers to an alkyl group having the meaning as defined above wherein one or more hydrogens are replaced with a halogen as defined above.
  • Non-limiting examples of such haloalkyl groups include chloromethyl, 1- bromoethyl, fluoromethyl, difluoromethyl, trifluoromethyl, 1,1,1-trifluoroethyl and the like.
  • cycloalkyl as used herein is a monovalent, saturated, or unsaturated monocyclic or bicyclic hydrocarbyl group.
  • Cycloalkyl groups may comprise 3 or more carbon atoms in the ring and generally, according to this invention comprise from 3 to 10, more preferably from 3 to 8 carbon atoms, and still more preferably from 3 to 6 carbon atoms.
  • Examples of cycloalkyl groups include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • heteroatom refers to any atom that is not carbon or hydrogen. Non-limiting examples of such heteroatoms include nitrogen, oxygen, sulfur, and phosphorus. Preferred heteroatoms according to the invention are nitrogen, oxygen and sulfur.
  • heterocyclyl refers to non-aromatic, fully saturated or partially unsaturated cyclic groups (for example, 3- to 7-membered monocyclic, 7- to 11-membered bicyclic, or containing a total of 3 to 10 ring atoms) which have at least one heteroatom in at least one carbon atom-containing ring.
  • Each ring of the heterocyclic group containing a heteroatom may have 1, 2, 3 or 4 heteroatoms selected from nitrogen, oxygen and/or sulfur atoms, where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized.
  • heterocyclic group may be attached at any heteroatom or carbon atom of the ring or ring system, where valence allows.
  • heterocyclyl groups include but are not limited to aziridinyl, azetidinyl, pyrrolidinyl, piperidinyl, azepanyl, piperazinyl, morpholinyl.
  • aryl refers to a polyunsaturated, aromatic hydrocarbyl group having a single ring (e.g. phenyl) or multiple aromatic rings fused together (e.g. naphthyl), typically containing 5 to 12 atoms; preferably 6 to 10, wherein at least one ring is aromatic.
  • aryl groups include but are not limited to phenyl, biphenyl, 1-naphthyl (or naphthalene-1-yl), 2-naphthyl (or naphthalene-2-yl), anthracenyl, indanyl, indenyl, 1,2,3,4- tetrahydronaphthyl.
  • heteroaryl refers but is not limited to 5 to 12 carbon-atom aromatic rings or ring systems containing 1 to 2 rings which are fused together, each ring typically containing 5 to 6 atoms; at least one of which is aromatic, in which one or more carbon atoms in one or more of these rings is replaced by oxygen, nitrogen and/or sulfur atoms where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized.
  • heteroaryl groups include but are not limited to pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, quinolinyl, quinoxalinyl, quinazolinyl, furanyl, benzofuranyl, pyrrolyl, indolyl, thiophenyl, benzothiophenyl, imidazolyl, benzimidazolyl, pyrazolyl, indazolyl, oxazolyl, benzoxazolyl, isoxazolyl, benzisoxazolyl, thiazolyl, and benzothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, dioxazolyl, dithiazolyl and tetrazolyl.
  • the compounds of the invention containing a basic functional group may be in the form of pharmaceutically acceptable salts.
  • Pharmaceutically acceptable salts of the compounds of the invention containing one or more basic functional groups include in particular the acid addition salts thereof. Suitable acid addition salts are formed from acids which form non- toxic salts.
  • Examples include the acetate, adipate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, cinnamate, citrate, cyclamate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, pyroglutamate, saccharate, stearate, succinate, tannate,
  • Pharmaceutically acceptable salts of compounds of Formula I and subformulae, or a hydrophenyl compounds of Table 2b may for example be prepared as follows: (i) reacting the compound of Formula I or any of its subformulae, or a hydrophenyl compounds of Table 2b, with the desired acid; or (ii) converting one salt of the compound of Formula I or any of its subformulae, or a hydrophenyl compounds of Table 2b, to another by reaction with an appropriate acid or by means of a suitable ion exchange column. All these reactions are typically carried out in solution.
  • the salt may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent.
  • the degree of ionization in the salt may vary from completely ionized to almost non-ionized.
  • solvate is used herein to describe a molecular complex comprising the compound of the invention and one or more pharmaceutically acceptable solvent molecules, for example, ethanol.
  • hydrate is employed when said solvent is water.
  • the compounds of the invention include compounds of the invention as hereinbefore defined, including all polymorphs and crystal habits thereof, prodrugs and isomers thereof (including optical, geometric and tautomeric isomers) and isotopically-labeled compounds of the invention.
  • the invention in its broadest sense also includes non-pharmaceutically acceptable salts, which may for example be used in the isolation and/or purification of the compounds of the invention.
  • non-pharmaceutically acceptable salts which may for example be used in the isolation and/or purification of the compounds of the invention.
  • salts formed with optically active acids or bases may be used to form diastereoisomeric salts that can facilitate the separation of optically active isomers of the compounds of the invention.
  • patient refers to a warm-blooded animal, more preferably a human, who/which is awaiting or receiving medical care or is or will be the object of a medical procedure.
  • human refers to subjects of both genders and at any stage of development (i.e.
  • the human is an adolescent or adult, preferably an adult.
  • the terms “treat”, “treating” and “treatment”, as used herein, are meant to include alleviating or abrogating a condition or disease and/or its attendant symptoms.
  • the term “therapeutically effective amount” (or more simply an “effective amount”) as used herein means the amount of active agent or active ingredient which is sufficient to achieve the desired therapeutic or prophylactic effect in the individual to which it is administered.
  • administration means providing the active agent or active ingredient, alone or as part of a pharmaceutically acceptable composition, to the patient in whom/which the condition, symptom, or disease is to be treated.
  • pharmaceutically acceptable is meant that the ingredients of a pharmaceutical composition are compatible with each other and not deleterious to the patient thereof.
  • excipient as used herein means a substance formulated alongside the active agent or active ingredient in a pharmaceutical composition or medicament. Acceptable excipients for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington’s Pharmaceutical Sciences, 21 st Edition 2011. The choice of excipient can be selected with regard to the intended route of administration and standard pharmaceutical practice.
  • the excipient must be acceptable in the sense of being not deleterious to the recipient thereof.
  • the at least one pharmaceutically acceptable excipient may be for example, a binder, a diluent, a carrier, a lubricant, a disintegrator, a wetting agent, a dispersing agent, a suspending agent, and the like.
  • pharmaceutical vehicle as used herein means a carrier or inert medium used as solvent or diluent in which the pharmaceutically active agent is formulated and/or administered.
  • Non-limiting examples of pharmaceutical vehicles include creams, gels, lotions, solutions, and liposomes.
  • cancer refers to the physiological condition in subjects that is characterized by unregulated or dysregulated cell growth with the potential to invade or spread to other parts of the body.
  • cancer includes solid tumors and blood born tumors, whether malignant or benign.
  • cancer examples include, but are not limited to: Acinar adenocarcinoma, acinar carcinoma, acral-lentiginous melanoma, actinic keratosis, adenocarcinoma, adenocystic carcinoma, adenosquamous carcinoma, adnexal carcinoma, adrenal rest tumor, adrenocortical carcinoma, aldosterone secreting carcinoma, alveolar soft part sarcoma, amelanotic melanoma, ameloblastic thyroid carcinoma, angiosarcoma, apocrine carcinoma, Askin’s tumor, astrocytoma, basal cell carcinoma, basaloid carcinoma, basosquamous cell carcinoma, biliary cancer, bone cancer, bone marrow cancer, botryoid sarcoma, brain cancer, breast cancer, bronchioalveolar carcinoma, bronchogenic adenocarcinoma, bronchogenic carcinoma, carcinoma ex pleomorphic adenom
  • Preferred cancers according to the invention are glioblastoma, triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer, more preferred cancer is glioblastoma.
  • anticancer agent refers to a chemical agent used to treat cancer, administered in regimens of one or more cycles, alone or combined with one or more agents over a period of days to weeks. Such agents are toxic to cells with high proliferative rates, such as cancer cells.
  • anticancer agents include, but are not limited to: - alkylating agents, such as for example cyclophosphamide, mechlorethamine, chlorambucil, melphalan, dacarbazine, dacarbazine, nitrosoureas or temozolomide; - antimetabolites, including antifolates, such as for example methotrexate, pemetrexed, pralatrexate or trimetrexate; pyrimidine analogues, such as for example azacitidine, capecitabine, cytarabine, decitabine, floxurinine, fluorouracil, gemcitabine or trifluridine; and purine analogues such as for example azathioprine, cladribine, fludarabine, mercaptopurine or tioguanine (formerly thioguanine); - anti-microtubule agents, including taxanes, such as for example paclitaxel,
  • FIGURES Figure 1 In vitro inhibition of IRE1 activity by the compounds of the invention. Measure of IRE1 activity in vitro in the presence of increasing concentrations of MKC (A) and compounds 34 (Z4A), 35 (Z4B), 36 (Z4C), 37 (Z4D) and 38 (Z4E) (B-F). FRET signals were measured upon fluorescent probe cleavage over a 25 minutes incubation.
  • FIG. 1 The IC50 calculated from the fitting step are shown in the figure. Symbols and error bars represent mean values ⁇ SD.
  • Figure 2 Compound 33 (Z4) and compound 28 (Z4P) inhibit IRE1 activity in cellular models.
  • A Protein levels of IRE1 and phospho-IRE1 in U87 cells treated with compound 33 (Z4) and compound 28 (Z4P) over 24 hours. Fold change of protein expression between IRE1 and phospho-IRE1 is represented in bar chart form normalized to untreated U87 cells.
  • Typical procedure 7 Acyl azide formation from carboxylic derivative
  • Carboxylic derivative (1 equiv.) was dissolved in dry toluene (2 mL / 100 mg) and NEt3 (1.05 equiv.) was added at room temperature. After 15 minutes, DPPA (1.05 equiv.) was slowly added. The mixture was stirred until complete consumption of starting material, and monitored by TLC. Water was added and the solution was extracted twice by EtOAc. Combined organic layer was washed with brine, dried by MgSO4 and concentrated under vacuo. The crude material was used without purification or was purified using SCC, Cyclohexane/EtOAc to afford pure product.
  • Typical procedure 8 Urea formation from acyl azide derivative through from Curtius rearrangment
  • Acyl azide derivative (1 equiv.) was dissolved in dry toluene (2 mL / 100 mg) and was heated at reflux overnight. The mixture was allowed to return at room temperature and 2,5-dimethylaniline (1 equiv.) was added. The mixture was stirred overnight at room temperature. The precipitate was filtered and washed with toluene to afford desired compound or water was added and the mixture was extracted twice by EtOAc. Combined organic layer was washed with brine, dried by MgSO4 and concentrated under vacuo.
  • Typical procedure 12 Reductive amination of 4-(4-hydroxyphenyl)butan-2-one derivatives [Adapted from ACS Chem. Neurosci., 2017, 8, 486-500] To a 500 mL round bottom flask filled with methanol (150 mL) was added the ketone starting material (28.05 mmol, 1 equiv.) and ammonium acetate (12.97 g, 168.3 mmol, 6 equiv.).
  • reaction mixture was left to stir at room temperature for 30 min before lowering the temperature to 0°C with an ice bath.
  • Sodium cyanoborohydride (2.64 g, 42.07 mmol, 1.5 equiv.) was then added portion wise.
  • the ice batch was removed and the reaction left to stir overnight at room temperature.
  • TLC monitoring indicated full consumption of the starting material and the reaction was subsequently quenched at 0°C by addition of HCl 1M (200 mL). After 15 min of stirring, the reaction mixture was concentrated by rotary evaporation to remove most of the methanol and then extracted with diethyl ether (2 ⁇ 100 mL).
  • the aqueous layer is isolated and basified by addition of concentrated NaOH until pH ⁇ 10, and then sodium chloride is added until saturation, followed by extraction with DCM (3 ⁇ 100 mL).
  • the combined organic layers were dried over anh. MgSO 4 , filtered, evaporated to dryness by rotary-evaporation and the residue dried under high-vacuum to yield the crude product (80 to 85% yield on average).
  • Crude product was usually pure enough to be used without further purification, but if needed, purification can be performed by silica gel column chromatography with gradient elution (100:0-90:10 DCM/MeOH).
  • the reaction was monitored by TLC and stopped upon complete consumption of the amine derivative ( ⁇ 5h).
  • the reaction mixture was concentrated in vacuo, then filtered with Et 2 O (3 ⁇ 30 mL), the organic layer was concentrated in vacuo to yield to pure isocyanate product (425 mg, 74 %) with sufficient purity to be used directly in the next step without further purification.
  • Typical procedure 14 – Urea synthesis using isocyanatobenzene derivatives To a solution of anhydrous dichloromethane (6 mL) in an oven dried 25 mL round bottom flask under argon atmosphere and at room temperature was added 4-(4- methoxyphenyl)butan-2-amine (1 equiv.) and the corresponding isocyanatobenzene derivative (1 equiv.). The reaction mixture was stirred at room temperature, overnight, monitored by TLC and stopped upon complete consumption of the amine derivative. The treatment is specific for each compound: please refer to the protocol of each analogue. Typical procedure 15 – Saponification of ester group.
  • Typical procedure 16 – Deprotection of benzyl protecting group To a solution of benzyl protected compound (1.0 equiv.) in MeOH (10 mL / 0.2 mmol) under argon, 10% Pd/C was added (1/4 of alkyne substrate’s mass). The flask was evacuated and filled with hydrogen gas by balloon. The mixture was stirred at room temperature for 2 h. The Pd/C was removed by vacuum filtration through Celite and the Celite layer was washed with MeOH (3 ⁇ 10 mL).
  • EXAMPLE 3 1-(2,5-dimethylbenzyl)-3-(4-hydroxyphenethyl)urea 2,5-dimethylphenyl)methanamine (1.458 mmol, 1.0 equiv., 0.21 mL) and 4-(2- aminoethyl)phenol as primary amine (1.458 mmol, 1.0 equiv., 200 mg) were reacted according to Typical procedure 5 Bis. Crude was purified with SCC (100% DCM to 98/2 DCM/MeOH) to afford compound 3, 107 mg, yield: 24.6%. A fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound for characterization and biological assays.
  • EXAMPLE 16 3-(2,5-dimethylphenyl)-1-(4-(4-hydroxyphenyl)butan-2-yl)-1- methylurea (B3-1)
  • STEP 1 4-(4-(benzyloxy)phenyl)-N-methylbutan-2-amine (L1) -
  • L1 4-(4-(benzyloxy)phenyl)-N-methylbutan-2-amine
  • 4-(4-(benzyloxy)phenyl)butan-2-one 500 mg, 1.966 mmol, 1 equiv.
  • aqueous methylamine 40 wt. % in water, 680 ⁇ L, 7.864 mmol, 4 equiv).
  • EXAMPLE 18 2,5-dimethylphenyl (4-(4-hydroxyphenyl)butan-2-yl)carbamate step 2 step 3 STEP 1 4-nitrophenyl (4-(4-methoxyphenyl)butan-2-yl)carbamate (N1) – To a solution of 4-(4- methoxyphenyl)butan-2-amine (2.568 g, 14.326 mmol, 1 equiv.) and DIPEA (4.99 mL, 28.611 mmol, 2 equiv.) in anh.
  • EXAMPLE 19 4-(4-hydroxyphenyl)butan-2-yl 2-(2,5-dimethylphenyl)acetate
  • STEP 1 4-(4-methoxyphenyl)butan-2-yl 2-(2,5-dimethylphenyl)acetate (O1) –
  • 2- (2,5-dimethylphenyl)acetic acid 80 mg, 0.487 mmol, 1 equiv.
  • N,N'- dicyclohexylcarbodiimide 110.6 mg, 0.536 mmol 1.1 equiv.
  • 4-Dimethylaminopyridine 9 mg, 0.073 mmol, 0.15 equiv.
  • EXAMPLE 20 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)thiourea STEP 1 1-(2,5-dimethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)thiourea (P1) – Due to its toxicity, 2,5-dimethylphenylisothyanate (60 mg, 0.368 mmol, 1 equiv.) was weighted under the fume hood in an oven dry 5 mL round bottom flask, rapidly put under argon atmosphere and to which was added anh. DCM (2 mL).
  • EXAMPLE 21 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(o-tolyl)urea
  • STEP 1 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(o-tolyl)urea (Q1) -
  • Q1 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(o-tolyl)urea (Q1) -
  • Q1 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(o-tolyl)urea (Q1) -
  • Q1 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(o-tolyl)urea (Q1) -
  • 1-(3-isocyanatobutyl)-4-methoxybenzene 80 mg, 0.390 mmol, 1 equiv.
  • o-toluidine 41.77 mg,
  • EXAMPLE 22 1-(3,5-dimethyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea STEP 1 1-(3,5-dimethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea (R1) - According to typical procedure 13 part 2 using 4-(4-methoxyphenyl)butan-2-amine (80 mg, 0,446 mmol, 1 equiv.) and isocyanato-3,5-dimethylbenzene (59.7 mg, 0.446 mmol, 1 equiv.).
  • EXAMPLE 24 1-(benzo[d]thiazol-6-yl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea STEP 1 6-isocyanatobenzo[d]thiazole (T1) - To a solution of anhydrous dichloromethane (6 mL) under inert atmosphere with NEt 3 (161.69 mg, 1.598 mmol, 3 equiv.) was added the benzo[d]thiazol-6-amine (80 mg, 0.533 mmol, 1 equiv.), the mixture was cooled to 0-5°C with an ice bath.
  • Triphosgene was added (79 mg, 0.266 mmol, 0.5 equiv.), and the mixture was allowed to return at room temperature. The reaction was monitored by TLC and stopped upon complete consumption of the amine derivative (overnight). The reaction mixture was concentrated in vacuo, then filtered with Et2O (3 ⁇ 15 mL). The organic layer was concentrated in vacuo to yield to pure isocyanate product T1 (62 mg, 66%) with sufficient purity to be used directly in step 3 without further purification.
  • STEP 2 4-(3-aminobutyl)phenyl acetate (T2) – Synthesized according to typical procedure: Reductive amination of 4-(4-hydroxyphenyl)butan-2-one derivatives – To a solution of anhydrous dichloromethane (8 mL) and TFA (trifluoroacetic acid – 8 mL) was added acetic anhydride (810 mg, 7.93 mmol, 4 equiv.) and 4-(4-hydroxyphenyl)butan-2-aminium chloride (400 mg, 1.98 mmol, 1 equiv.) at room temperature.
  • STEP 3 4-(3-(3-benzo[d]thiazol-6-yl)ureido)butyl)phenyl acetate (T3) -
  • 6-isocyanatobenzo[d]thiazole T1 62 mg, 0.352 mmol, 1 equiv.
  • NEt3 42.7 mg, 0.422 mmol, 1.2 equiv.
  • 4-(3-aminobutyl)phenyl acetate T2 80 mg, 0.387 mmol, 1.1 equiv.
  • reaction mixture was stirred at room temperature, overnight, monitored by TLC and stopped upon complete consumption of the amine derivative.
  • the resulting mixture was evaporated by rotary evaporation and the crude product was purified by SCC (97:3 dichloromethane/methanol), yielded the pure desired product T3: 55 mg, 40%.
  • EXAMPLE 25 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(quinolin-7-yl)urea U1 U 3 25 STEP 1 7-isocyanatoquinoline (U1) -
  • anhydrous dichloromethane (6 mL) under inert atmosphere with NEt3 (309.9 mg, 0.613 mmol, 5 equiv.) was added the quinolin-7-amine (133 mg, 0.613 mmol, 1 equiv.), the mixture was cooled to 0-5°C with an ice bath.
  • Triphosgene was added (181.8 mg, 0.613 mmol, 1 equiv.), and the mixture was allowed to return at room temperature.
  • reaction mixture was then quenched with H 2 O (4 mL), and the biphasic reaction mixture was extracted with ethyl acetate (3 ⁇ 10 mL). The combined organic layers were dried over MgSO 4 and concentrated to afford crude product, which was purified by SCC (9:1 to 7:3 petroleum ether/acetone), yielded the pure desired compound 25: 18 mg, 88%.
  • EXAMPLE 26 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(2-methylnaphtalen-1-yl)urea
  • STEP 1 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(2-methylnaphtalen-1-yl)urea (V1) -
  • V1 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(2-methylnaphtalen-1-yl)urea (V1) -
  • typical procedure 14 using 1-(3-isocyanatobutyl)-4-methoxybenzene (50 mg, 0.244 mmol, 1 equiv.) and 2-methylnaphtalen-1-amine (38.2 mg, 0.244 mmol, 1 equiv.).
  • EXAMPLE 27 1-(2-(tert-butyl)phenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea STEP 1 1-(4-(4-(benzyloxy)phenyl)butan-2-yl)-3-(2-(tert-butyl)phenyl)urea (W1) - According to typical procedure 14 using 1-(benzyloxy)-4-(3-isocyanatobutyl)benzene (70 mg, 0.249 mmol, 1 equiv.) and 2-(tert-butyl)aniline (40.84 mg, 0.273 mmol, 1.1 equiv.).
  • EXAMPLE 28 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea 4-(4-methoxyphenyl)butan-2-amine (W1) – To a 250 mL round bottom flask filled with methanol (150 mL) was added 4-(4-methoxyphenyl)butan-2-one (5 g, 28.05 mmol, 1 equiv.) and ammonium acetate (12.97 g, 168.3 mmol, 6 equiv.) and the reaction mixture was left to stir at room temperature for 30 min.
  • reaction mixture is diluted with EtOAc (100 mL), transferred in a separatory funnel and washed successively with 1M citric acid (100 mL), sat. sodium carbonate (100 mL), and brine (100 mL).
  • 1M citric acid 100 mL
  • sat. sodium carbonate 100 mL
  • brine 100 mL
  • the organic layer is dried over anh. MgSO 4 , filtered, evaporated to dryness by rotary-evaporation and the residue dried under high-vacuum to afford 1.9 g of W2 (74% yield) as a pale beige solid, used without further purification.
  • EXAMPLE 30 1-(2,5-dimethylphenyl)-3-(4-hydroxyphenethyl)urea 1-(2,5-dimethylphenyl)-3-(4-hydroxyphenethyl)urea (30) - 2,5-dimethylaniline (1,458 mmol, 1.0 equiv., 0.18 mL) and 4-(2-aminoethyl)phenol as primary amine (1.458 mmol, 1.0 equiv., 200 mg) were reacted according to Typical procedure 5. Crude was purified with SCC (100% DCM to 98/2 DCM/MeOH) to afford compound 30, 297 mg, yield: 71.6%.
  • EXAMPLE 31 N-(2,5-dimethylphenyl)-4-(3-hydroxyphenyl)piperazine-1- carboxamide
  • STEP 1 N-(2,5-dimethylphenyl)-4-(3-methoxyphenyl)piperazine-1-carboxamide (Z1) - According to Typical procedure 5 using 2-isocyanato-1,4-dimethylbenzene (0.219 mmol, 1.0 equiv., 32 mg) and 1-(3-methoxyphenyl)piperazine hydrochloride (0.219 mmol, 1.0 equiv., 50 mg) in dry DCM (3 mL) followed by triethylamine (0.219 mmol, 1.0 equiv., 30 ⁇ L).
  • EXAMPLE 32 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(p-tolyl)urea
  • STEP 1 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(p-tolyl)urea (AA1) -
  • AA1 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(p-tolyl)urea
  • AA1 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(p-tolyl)urea
  • the crude product was purified by SCC with solid deposition (75:25 petroleum ether/acetone), yielded the pure desired product AA1: 40 mg, 52%.
  • EXAMPLE 39 1-(2,5-dimethylbenzyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea
  • STEP 1 1-(2,5-dimethylbenzyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea (AB1) -
  • AB1 1-(2,5-dimethylbenzyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea (AB1) -
  • Typical procedure 5 Bis using 2,5-dimethylphenyl)methanamine (0.75 mmol, 1.0 equiv., 0.1 mL) and 4-(4-methoxyphenyl)butan-2-amine W1 (0.75 mmol, 1.0 equiv., 101 mg).
  • EXAMPLE 40 1-(2,5-dimethylphenyl)-3-(3-(3-hydroxyphenyl)prop-2-yn-1-yl)urea 1-(2,5-dimethylphenyl)-3-(3-(3-hydroxyphenyl)prop-2-yn-1-yl)urea (40) - According to Typical procedure 2 using 1-(2,5-dimethylphenyl)-3-(prop-2-yn-1-yl)urea D1 (0.227 mmol, 1.0 equiv., 46 mg) as alkyne derivative and 3-iodophenol (0.227 mmol, 1.0 equiv., 50 mg) as iodine derivative. The mixture was stirred overnight.
  • EXAMPLE 42 1-(2,5-dimethylphenyl)-3-(4-(3-hydroxyphenyl)-2-methylbut-3-yn-2- yl)urea 1-(2,5-dimethylphenyl)-3-(4-(3-hydroxyphenyl)-2-methylbut-3-yn-2-yl)urea (44) - According to Typical procedure 2 using 1-(2,5-dimethylphenyl)-3-(2-methylbut-3-yn-2- yl)urea E1 (0.227 mmol, 1.0 equiv., 52 mg) as alkyne derivative and 3-iodophenol (0.227 mmol, 1.0 equiv., 50 mg) as iodine derivative.
  • EXAMPLE 46 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)piperidin-4-yl)urea STEP 1 tert-butyl (1-(4-methoxyphenyl)piperidin-4-yl)carbamate (AG1) - According to Typical procedure 9 using 1-iodo-4-methoxybenzene (1.67 mmol, 1.0 equiv., 390 mg) as iodine derivative and tert-butyl piperidin-4-ylcarbamate (2.0 mmol, 1.2 equiv., 400 mg) as piperidine derivative.
  • EXAMPLE 50 4-(4-hydroxyphenyl)butan-2-yl (2,5-dimethylphenyl)carbamate
  • STEP 1 4-(4-((tert-butyldimethylsilyl)oxy)phenyl)butan-2-one (AK1) -
  • EXAMPLE 56 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(1H-indol-6-yl)urea STEP 1 tert-butyl 6-isocyanato-1H-indole-1-carboxylate (AQ1) - According to Typical procedure 5 Bis part 1 using tert-butyl 6-amino-1H-indole-1-carboxylate (0.86 mmol, 1.0 equiv., 200 mg) in a solution of DCM (8 mL) under argon atmosphere with triethylamine (5.166 mmol, 6.0 equiv., 0.73 mL).
  • STEP 1 Bis 4-(4-acetoxyphenyl)butan-2-aminium chloride AQ2 - To a solution of DCM (8 mL) and TFA (8 mL) was added acetic anhydride (7.93 mmol, 4.0 equiv., 810 mg) and 4-(4- hydroxyphenyl)butan-2-aminium chloride (obtained from reductive amination of frambinone without basification, 1.98 mmol, 1.0 equiv., 400 mg) at room temperature. The resulting mixture was stirred (Monitored by TLC, around 4h), and stopped upon complete consumption.
  • tert-butyl 6-isocyanato-1H-indole-1-carboxylate AQ1 (0.86 mmol, 1.0 equiv., 222 mg) in THF (4 mL) was added and the reaction mixture was stirred at room temperature overnight. Solvent were evaporated and the crude was purified by SCC (Cyclohexane/EtOAc – 100:0 to 70:30) to obtain tert-butyl 6-(3-(4-(4- acetoxyphenyl)butan-2-yl)ureido)-1H-indole-1-carboxylate AQ3, 178 mg, yield: 45%, as a beige solid.
  • STEP 3 Bis 4-(3-(3-(1H-indol-6-yl)ureido)butyl)phenyl acetate hydrochloride (AQ5) - According to Typical procedure 10 using tert-butyl 6-(3-(4-(4-acetoxyphenyl)butan-2-yl)ureido)-1H- indole-1-carboxylate AQ4 (0.189 mmol, 1.0 equiv., 88 mg). The mixture was stirred at room temperature for 24h. The resulting solution was concentrated to give the product AQ5 which was engaged directly in the next step.
  • EXAMPLE 58 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)-1H-pyrrol-3-yl)urea
  • STEP 1 Methyl 1-(4-methoxyphenyl)-1H-pyrrole-3-carboxylate (AS1) -
  • methyl 1H-pyrrole-3-carboxylate 8 mmol, 1.0 equiv., 1 g
  • 4- iodoanisole (10.4 mmol, 1.3 equiv., 2.43 g)
  • CuI (10 mol%, 150 mg) K3PO4 (16 mmol, 2.0 equiv., 3.39 g)
  • EXAMPLE 60 1-(3,5-difluorophenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea STEP 1 1,3-difluoro-5-isocyanatobenzene (AT1) - According to Typical procedure 5 Bis part 1 using 3,5-difluoroaniline (0.775 mmol, 1.0 equiv., 100 mg) in a solution of DCM (10 mL) under argon atmosphere with triethylamine (1.55 mmol, 2.0 equiv., 0.211 ml).
  • STEP 2 1-(3,5-difluorophenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea (AT2) -
  • Typical procedure 5 Bis part 2 using a solution of 4-(4-methoxyphenyl)butan-2-amine W1 (0.775 mmol, 1.0 equiv., 139 mg) in DCM (4 mL) and the freshly prepared 1,3-difluoro-5- isocyanatobenzene AT1 in DCM (2 mL) which was added to the solution. The reaction was putted under argon atmosphere and stirred overnight.
  • STEP 2 4-(3-(3-(benzo[d][1,3]dioxol-5-yl)ureido)butyl)phenyl acetate (AZ2) - According to Typical procedure 5 Bis part 2 using 5-isocyantobenzo[d][1,3]dioxole AZ1 (0.981 mmol, 1.0 equiv., 160 mg), triethylamine (2.944 mmol, 3.0 equiv., 0.38 mL) and 4-(4- acetoxyphenyl)butan-2-aminium chloride AR2 (0.981 mmol, 1.0 equiv., 203 mg) in DCM (8 mL).
  • BIOLOGICAL EVALUATION Materials and methods Materials – IRE1 wild-type recombinant protein encoding the cytoplasmic domain (amino acids 465–977) with N-terminal polyhistidine-tag and GST tag was from Sinobiological (Sino Biological Europe GmbH, Eschborn, Germany, #11905-H20B). The fluorescent probe used for the in vitro IRE1 RNase assay was from Eurogentec. Tunicamycin was purchased from Calbiochem (Merck KGaA, Darmstadt, Germany).
  • MicroScale Thermophoresis The direct binding of compound 33 (Z4) and compound 28 (Z4P) to IRE1 protein was measured using MicroScale Thermophoresis (MST).
  • MST MicroScale Thermophoresis
  • IRE recombinant protein was labelled using RED-Tris-NTA fluorescent dye (RED-Tris-NTA 2 nd Generation, NanoTemper, Kunststoff, Germany; # MO-L018).
  • 100 ⁇ L of 20 nM protein solution is mixed with 100 ⁇ L of 10 nM RED-Tris-NTA dye in PBST buffer (PBS with 0.05% Tween-20) and incubated for 30 min at RT.
  • the protein–dye mixture was centrifuged for 10 min at 4°C and 15000xg.
  • the compounds were analyzed in a 16-point dilution series mixed in a 1:1 ratio with the labelled protein in PBST buffer.
  • the assay was performed in standard Monolith NT.115 Capillaries (NanoTemper; #MO-K022), and all measurements were performed at 60% MST power and 60% excitation power using the Monolith NT.115 Pico machine (NanoTemper).
  • the dissociation constant (Kd) was calculated by taking the average of triplicate normalized fluorescence data using NANOTEMPER analysis software (MO.Affinity Analysis v2.3).
  • IRE1-mediated in vitro RNase assay – Organic molecules (Compounds 28 (Z4P), and 33 (Z4), 34 (Z4A), 35 (Z4B), 36 (Z4C), 37 (Z4D) and 38 (Z4E)) were diluted in minimal volume of DMSO and subsequently re-diluted in reaction buffer (20 mM HEPES pH 7.5; 1 mM MgOAc; 50 mM KOAc). Maximum volume of DMSO per reaction never exceeded 1%. Reaction volume was 25 ⁇ L.
  • Recombinant IRE1 (0.6 ⁇ g/ reaction) was incubated at room temperature for 10 minutes with varying concentrations (0-100 ⁇ M) of inhibitor and reaction buffer.
  • the assay relied on the use of fluorescence resonance energy transfer (FRET)— quenched mini Xbp1 RNA substrate probe, which when cleaved by IRE1 emits fluorescence at 590 nm (cy3) wavelength (F. Prischi et al., Nature Communications, 2014, 5, 3554).
  • FRET fluorescence resonance energy transfer
  • U87 were grown in DMEM Glutamax (Invitrogen, Carlsbad, CA, USA) supplemented with 10% FBS.
  • GB immortalized U251 and primary RADH87 (T. Avril et al., Brain Pathol., 2012, 22, 159–174) cells were grown in DMEM supplemented with 10% FBS in a 5% CO2 humidified atmosphere at 37°C.
  • GB cell lines were modified for IRE1 activity by overexpressing dominant negative (DN) forms of IRE1 that lack the RNase domain (IRE1.NCK or IRE1 Q780stop) as previously described (S. Lhomond et al., EMBO Molecular Medicine, 2018, 10, e7929; J.
  • DN dominant negative
  • TMZ sensitivity assays cells were plated in a 96 well plate at 5000 cells per well and co-treated with 0, 5, 10, 25, 50, 100, 250, 500, 1000 and 2500 ⁇ M of TMZ plus a non-toxic dose of inhibitor. After 6 days of incubation, WST1 reagent (Roche) was added to each well and post 2-hour incubation the plate was read using a Tecan 200 colorimeter. Western blotting - All IRE1 signaling analyses were carried out as described previously (S. Lhomond et al., Methods Mol. Biol., 2015, 1292, 177-194).
  • IRE1 and phosphorylated IRE1 were stained using anti ⁇ IRE1 antibody (Anti- human; rabbit polyclonal; SantaCruz Biotechnologies, H-190) and pS724-IRE1 antibody (Anti-human; rabbit polyclonal; Abcam, ab48187), respectively.
  • the phosphorylated form of eIF2 ⁇ was stained with anti-phospho-eIF2 ⁇ (Ser51) Antibody #9721 (CellSignalling®). Actin was used as a loading control ( ⁇ -Actin (C4): sc-47778; SantaCruz Biotechnologies).
  • Cell extracts were resolved by SDS-PAGE and transferred to nitrocellulose membranes for 30 minutes using a Trans-Blot® TurboTM (BioRad® Transfer System #1704150).
  • the resulting membranes were incubated with primary antibodies for 16 hours at 4°C, washed with PBST, and incubated for 1 hour with goat anti ⁇ rabbit or goat anti ⁇ mouse secondary antibodies at room temperature (Invitrogen, Carlsbad, CA, USA) prior revelation using chemiluminescence (ECL RevelBlOt® Intense, Ozyme).
  • Washed pellet were then denatured with 8 M urea in Tris-HCl 0.1 mM, reduced with 5 mM TCEP for 30 minutes, and then alkylated with 10 mM iodoacetamide for 30 minutes in the dark.
  • Double digestion was performed with endoproteinase Lys-C (Ref 125-05061, Wako) at a ratio 1/100 (enzyme/proteins) in 8 M urea for 4h, followed by an overnight modified trypsin digestion (Ref V511A, Promega) at a ratio 1/100 (enzyme/proteins) in 2 M urea. Both Lys- C and Trypsin digestions were performed at 37°C.
  • Peptide mixtures were then desalted on C18 spin-column and dried on Speed-Vacuum before LC-MS/MS analysis.
  • Samples were analyzed using an Ultimate 3000 nano-RSLC (Thermo Scientific, San Jose California) coupled in line with a LTQ-Orbitrap ELITE mass spectrometer via a nano-electrospray ionization source (Thermo Scientific, San Jose California).
  • Proteins were identified and quantified by database searching using SequestHT (Thermo Fisher Scientific) with Proteome Discoverer 2.4 software (PD2.4, Thermo Fisher Scientific) against Homo sapiens reviewed SwissProt database.
  • Tumor cell orthotopic implantation - Tumor cells (U87-Luc (A. Jabouille, et al., Oncotarget, 2015, 6, 24922–24934)) were implanted into the brain of immunodeficient NMRI-Foxn1nu/Foxn1nu, 8 weeks old male mice (Janvier Laboratories, Laval, France).
  • This framework makes it possible to manipulate the brains of living animals, and to reach isolated areas of the brain precisely relative to markings visible to the naked eye through the use of three- dimensional coordinates.
  • the stereotaxic coordinates were calculated for injection of tumor cells into a specific point of the brain, and reproducible for all the mice used.
  • the tumor cells (5 ⁇ 10 4 cells per mice in 1 ⁇ L) were injected at Bregma 0, 2.2 mm to the left of the bregma and 3.2 mm deep to perform the implantation at the level of the striatum.
  • Mouse treatments – Four days after tumor cells implantation, compound 28 (Z4P) treatments were started consisting in treatments of 300 ⁇ g/kg/day intraperitonially.
  • RNA quantification experiment performed on tumors generated in mice, three mice per group were treated according to the methods above. Mice were sacrificed at day 34 and the brains were removed and extract the tumor. The samples were used to extract RNA for the analysis, as described below.
  • Quantitative real ⁇ time PCR - Total RNA was prepared using TRIzol reagent (Invitrogen, Carlsbad, CA, USA). All RNAs were reverse ⁇ transcribed with Maxima Reverse Transcriptase (Thermo Scientific, Waltham, MA, USA), according to manufacturer protocol. qPCR was performed via a StepOnePlusTM Real ⁇ Time PCR Systems from Applied Biosystems and the SYBR Green PCR Core reagents kit (Takara). Analysis was carried out using QuantStudio TM Design and Analysis software version 1.3.1. Three technical repeats were performed per experiment and at least three biological repeats were performed per point per experiment. Each sample was extract individually for RNA and performed the quantification of mRNA levels by qPCR for several targets.
  • lysis buffer 25 mM Tricine pH 7.8; 15 mM Potassium Phosphate pH 7.8; 15 mM MgSO 4 ; 4 mM EGTA; 1% Triton X-100; 1 mM DTT
  • substrate buffer 25 mM Tricine pH 7.8; 15 mM Potassium Phosphate pH 7.8; 15 mM MgSO 4 ; 4 mM EGTA; 1% Triton X-100; 1 mM DTT; 1 mM ATP; 0.2 mM luciferin
  • IRE1-fragment-derived organic molecules dock onto the ATP binding pocket of IRE1 in vitro
  • MST micro-scale thermophoresis
  • proteomes of these GB lines exposed to compound 33 was then functionally compared to those of the same cell lines in which IRE1 activity was invalidated genetically either using a dominant negative construct reported previously (B. Drogat, et al., Cancer Res., 2007, 67, 6700–6707) or a truncated mutant variant of IRE1 lacking an RNase domain (Q780stop) (S. Lhomond, et al., EMBO Molecular Medicine, 2018, 10, e7929) (data not shown). Genes that were down- or up-regulated were compared according to the cell lines and conditions.
  • compound 28 (Z4P) displayed zero toxicity in WT mice, it was then sought to (i) characterize the effect of compound 28 (Z4P) on IRE1 biology in an in vivo tumor; (ii) test whether compound 28 (Z4P) was able to cross the BBB and reach tumors cells; (iii) determine if co-treatment of compound 28 (Z4P) alongside SOC chemotherapy TMZ conferred any anti-tumorigenic, anti-relapse or pro-survival advantages. To do so, U87-luc cells were orthotopically implanted in the mouse brain and the tumor was allowed to grow. At day 4 post implantation, a small tumor formation was detected using bioluminescence (data not shown).
  • mice were randomized in different groups and compound 28 (Z4P) daily treatments (i.p.300 ⁇ g/kg) were administered for 34 consecutive days (Figure 5A).
  • compound 28 (Z4P) daily treatments i.p.300 ⁇ g/kg
  • Figure 5A mice were randomized in different groups and compound 28 (Z4P) daily treatments (i.p.300 ⁇ g/kg) were administered for 34 consecutive days ( Figure 5A).
  • mouse brains were entirely excised and dissected.
  • the corresponding tumors were resected, dissociated and RNA was extracted from them.
  • the splicing of Xbp1 mRNA was evaluated in control tumors and in animals treated with compound 28 (Z4P).
  • Treatment with compound 28 (Z4P) decreased the splicing of Xbp1 mRNA thus demonstrating that compound 28 (Z4P) mediates IRE1 inhibition in vivo when administered IP, also implying that it crossed the BBB ( Figure 5B).
  • mice treated with TMZ alone relapsed (Figure 5D, black curve) forming new tumors; a result consistent with what is observed in clinic.
  • no tumor relapse was observed in the group of animals treated with both TMZ and compound 28 (Z4P) ( Figure 5D, grey curve).
  • compound 28 (Z4P) administered intraperitoneally, sensitizes orthotopically implanted GB tumors to TMZ in mice. It prevents tumor relapse when used as an adjuvant therapy alongside TMZ. This has major translational clinical implications as GB tumor relapse is the major disease evolution characteristic causing death in human patients.
  • IRE1-mediated in vitro RNase assay and Luciferase assay Results of the IRE1-mediated in vitro RNase assay and Luciferase assay for the compounds of the invention are listed in the Table 5 below: Table 5

Abstract

The inventors have succeeded in developing urea, oxalamide, amide, thiourea, carbamate or ester compounds, in particular urea compounds, bearing two side groups, one of which carries a hydroxyphenyl or phenyl moiety, in particular a hydroxyphenyl moiety. These compounds have the advantage of inhibiting IRE1 RNase activity and sensitizing cancer cells, in particular GB cells, to chemotherapy. The present invention relates to urea, oxalamide, amide, thiourea, carbamate or ester compounds, in particular urea compounds containing a hydroxyphenyl or phenyl moiety, in particular a hydroxyphenyl moiety, including their pharmaceutically acceptable salts and solvates which are useful as sensitizers for chemotherapy of cancer cells, particularly in glioblastoma, and are useful as therapeutic compounds, particularly in the treatment of cancers that may be treated by alkylating agents, such as temozolomide.

Description

COMPOUNDS CONTAINING A HYDROXYPHENYL MOIETY AND THEIR USE The present invention relates to urea, oxalamide, amide, thiourea, carbamate or ester compounds, in particular urea compounds, containing a hydroxyphenyl or phenyl moiety, in particular a hydroxyphenyl moiety, including their pharmaceutically acceptable salts and solvates which are useful as sensitizers for chemotherapy of cancer cells, particularly in glioblastoma, and are useful as therapeutic compounds, paraticularly in the treatment of cancers that may be treated by alkylating agents, such as temozolomide. BACKGROUND OF THE INVENTION Glioblastoma (GB) is the most common primary central nervous system (CNS) tumour, displaying high levels of aggressiveness, recurrence and heterogeneity; traits that contribute to a dismal prognosis of an average of 1.5 year survival post diagnosis. The standard of care comprises maximal safe resection of the tumour followed by a combination of irradiation and chemotherapy with the alkylating agent temozolomide; however, all patients succumb to the disease (R. Stupp et al., N. Engl. J. Med., 2005, 352, 987-996). GB cells, as with most solid tumours, survives in a hostile environment which includes hypoxia, nutrient shortage, necrosis and immune infiltration, as well as having to cope with a high metabolic turnover and protein synthesis demand (D. Doultsinos et al., SLAS Discov. Adv. Life Sci. R&D, 2017, 22, 787-800). As such, the Unfolded Protein Response (UPR) is inextricably linked to GB pathophysiology (J. Obacz et al., Sci. Signal., 2017, 10, eaal2323). It has been shown that in particular Inositol Requiring Enzyme 1 (IRE1), a major Unfolded Protein Response (UPR) transducer, plays a decisive role in tumorigenesis and aggressiveness as through XBP1s signalling it is promoting tumour infiltration by immune cells, angiogenesis and invasion. GB tumours displaying high levels of IRE1/XBP1 activity have a worse prognosis than those with low activity (S. Lhomond et al., EMBO Mol. Med., 2018, 10, 139-308). This pertains to the possibility that attenuating IRE1 activity could lead to sensitization of tumours to current therapies as GB cells would exhibit reduced capacity to cope with the hostile environment. Indeed, such studies have been performed in Triple Negative Breast Cancer (TNBC) showing that inhibition of IRE1 RNase activity with salicylaldehyde MKC8866 increased paclitaxel-dependent attenuation of TNBC development in mouse xenograft models (S.E. Logue et al., Nat. Commun., 2018, 9, 3267). Further to this, MKC8866 treatment greatly enhanced the efficacy of docetaxel in regressing MYC-overexpressing tumours in breast cancer PDX models (N. Zhao et al., J. Clin. Invest., 2018, 128, 1283-1299). This inhibitor is currently tested on other types of cancers. IRE1 activity inhibition can be mediated by compounds targeting either the ATP-binding kinase domain or the RNase domain. Direct RNase pharmacological inhibitors include 4μ8c, STF-083010, toyocamycin and a series of MKC compounds, all relying on a hydroxy-aryl aldehyde (HAA) motif, whilst kinase pharmacological inhibitors that in turn inhibit the RNase include amongst others 1-(4-(8-amino-3-isopropylimidazo[1,5-a]pyrazin-1- yl)naphthalen-1-yl)-3-(3-(trifluoro-methyl)phenyl)urea (CAS# 1414938-21-8), 1-(4-(8- amino-3-(tert-butyl)imidazo[1,5-a]pyrazin-1-yl)naphthalen-1-yl)-3-(3- (trifluoromethyl)phenyl)urea (CAS# 1589527-65-0), 1-(4-(8-amino-3-(1- methylcyclopropyl)imidazo[1,5-a]pyrazin-1-yl)naphthalen-1-yl)-3-(3-fluorophenyl)urea (CAS# 1937235-76-1), 6-chloro-3-(6-fluoro-2-(phenylamino)-1H-benzo[d]imidazol-5-yl)- N-((1-methylpiperidin-4-yl)methyl)imidazo[1,2-b]pyridazin-8-amine (CAS# 2328097-41- 0), 2-(3,4-dichlorobenzyl)-N-(4-methylbenzyl)-2-azaspiro[4.5]decane-8-carboxamide (CAS# 2121989-91-9), 2-chloro-N-(6-methyl-5-((3-(2-(piperidin-3-ylamino)pyrimidin-4- yl)pyridin-2-yl)oxy)naphthalen-1-yl)benzenesulfonamide (CAS# 1630086-20-2) (T. Langlais, Biochem. J., 2021, 478, 2953-2975; D. Pelizzari-Raymundo et al., Trends in Cancer, 2020, 6, 1018-1030) and, although unclear as to its effect on IRE1 activity, sunitinib (C. Hetz et al., Nat. Rev. Drug Discov., 2013, 12, 703-719). The description of an allosteric IRE1 RNase inhibitory mechanism by ATP competitive ligands was provided through the discovery of Kinase inhibiting RNase attenuators (KIRAs) showing that inhibition of the kinase site may have an inhibitory effect on the RNase activity (L. Wang et al., Nat. Chem. Biol., 2012, 8, 982-989; H.C. Feldman et al., CS Chem. Biol., 2016, 11, 219-2205). Other studies indicated that large (18-50 amino acid long) peptides derived from the cytosolic domain of IRE1 could affect its oligomerisation and subsequent RNase activity (M. Bouchecareilh et al., FASEB J., 2011, 25, 3115-3129). However, such peptides, even in their reduced 18 amino acid form, presented a plethora of issues such as bioavailability, proteasomal degradation, sheer size, crossing the blood brain barrier and stability when considering use in in vivo CNS settings. However, there is still a need for compounds having the ability to inhibit IRE1 RNase activity and to sensitize cancer cells, in particular GB cells, to anticancer drugs, in particular alkylating agents, and that may be of therapeutic value in the treatment of cancers that may be treated by alkylating agents. SUMMARY OF THE INVENTION The inventors have now succeeded in developing urea, oxalamide, amide, thiourea, carbamate or ester compounds, in particular urea compounds, bearing two side groups, one of which carries a hydroxyphenyl or phenyl moiety, in particular a hydroxyphenyl moiety. These compounds have the advantage of inhibiting IRE1 RNase activity and sensitizing cancer cells, in particular GB cells, to chemotherapy. The invention therefore relates to compounds of general Formula I, their pharmaceutically acceptable salts and solvates as well as methods of use of such compounds or compositions comprising such compounds as sensitizers for chemotherapy of malignant tumors. In a general aspect, the invention provides compounds of general Formula I:
Figure imgf000004_0001
I, a pharmaceutically acceptable salt or a solvate thereof, wherein A is selected from –NH–, –N(Me)– and –O–; Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O–, –CH2– and –C(O)NH– or is a single bond; Z is O or S; R1 and R2 are independently selected from H and OH, with the proviso that at least one of R1 and R2 is H and that R1 and R2 are not both H; Cy is selected from: - R3 is selected from H, C1-C4-alkyl and halogen; R4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; R5 is H or C1-C4-alkyl; R6 is selected from H, C1-C4-alkyl, C3-C4-cycloalkyl, halogen, C(O)NR8R9, NHC(O)R10 and C(O)OR11 wherein R8 are R9 are independently selected from hydrogen, C1-C4-alkyl and C3- C4-cycloalkyl; R9 is C1-C4-alkyl; and R10 is selected from H and C1-C4-alkyl; and R7 is H or C1-C4-alkyl; with the proviso that R3, R4, R5, R6 and R7, are not all H; -
Figure imgf000006_0001
wherein R11 and R12 are independently selected from hydrogen and C1-C4-alkyl; - -
Figure imgf000006_0002
- - - with the proviso that, when
Figure imgf000007_0001
are not all H, and the compound of Formula I is none of the following: (R)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; (S)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N,4-dimethylbenzamide; (R)-4-fluoro-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)benzamide; (S)-N-ethyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; (S)-N-cyclopropyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; (S)-N1-(5-carbamoyl-2-fluorophenyl)-N2-(4-(4-hydroxyphenyl)butan-2-yl)oxalamide; (R)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N,4-dimethylbenzamide; (S)-N-(3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylphenyl)acetamide; (S)-1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(5-iodo-2-methylphenyl)urea; (S)-N-(3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylphenyl)propionamide; (S)-3-acetamido-N-(4-(4-hydroxyphenyl)butan-2-yl)-4-methylbenzamide; (S)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)benzamide; (R)-N-ethyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; Methyl (S)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzoate; (R)-N-(3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylphenyl)isobutyramide; (S)-1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; (S)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N,N,4-trimethylbenzamide; (S)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; (R)-N-(3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylphenyl)acetamide; (R)-N1-(5-carbamoyl-2-methylphenyl)-N2-(4-(4-hydroxyphenyl)butan-2- yl)oxalamide; N-ethyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; 3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N,4-dimethylbenzamide; 3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; 3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N,2-dimethylbenzamide; 4-fluoro-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)benzamide; 4-fluoro-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N-methylbenzamide; N-cyclopropyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; N-(3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)phenyl)isobutyramide; N-(5-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-2-methylphenyl)acetamide; (S)-1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(m-tolyl)urea; 1-(2,5-dimethylphenyl)-3-(4-phenylbutan-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-hydroxyphenethyl)urea; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(2-fluorophenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(2-ethyl-6-methylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; N-(2,5-dimethylphenyl)-4-(3-hydroxyphenyl)piperazine-1-carboxamide; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-phenylurea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(m-tolyl)urea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(p-tolyl)urea; 1-cyclohexyl-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; and 1-(benzo[d][1,3]dioxol-5-yl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea. The invention also relates to a compound of Formula I: I, a pharmaceutically acceptable salt or solvate thereof wherein A is selected from –NH–, –N(Me)– and –O–;
Figure imgf000009_0001
Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O–, –CH2– and –C(O)NH– or is a single bond; Z is O or S; R1 and R2 are independently selected from H, OH and OMe, with the proviso that at least one of R1 and R2 is H; Cy is selected from: -
Figure imgf000010_0001
wherein R3 is selected from H, C1-C4-alkyl, and halogen; R4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; R5 is H or C1-C4-alkyl; R6 is selected from H, C1-C4-alkyl, C3-C4-cycloalkyl, halogen, C(O)NR8R9, NHC(O)R10 and C(O)OR11 wherein R8 are R9 are independently selected from hydrogen, C1-C4-alkyl and C3- C4-cycloalkyl; R9 is C1-C4-alkyl; and R10 is selected from H and C1-C4-alkyl; and R7 is H or C1-C4-alkyl; - wherein R11 and R12 are independently selected from hydrogen and C1-C4-alkyl; - - - - -
Figure imgf000011_0001
in combination with an anticancer agent, particularly an alkylating agent, for use in treating cancer, particularly glioblastoma, triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer. The invention futher relates to compounds of Formula I or their pharmaceutically acceptable salts and solvates for use in increasing the sensitivity of cancer cells to an anticancer agent, particularly an alkylating agent, in a treatment of cancer, particularly glioblastoma, triple- negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer. In another aspect, the present invention provides a pharmaceutical composition comprising at least one compound of Formula I as defined above, or a pharmaceutically acceptable salt or solvate thereof, and at least one pharmaceutically acceptable carrier, diluent, excipient and/or adjuvant. In another aspect, the invention relates to a hydroxyphenyl compound selected from the group consisting of: 1-(2,5-dimethylphenyl)-3-(6-(3-hydroxyphenyl)pyridin-3-yl)urea; 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)-1H-pyrrol-3-yl)urea; 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)-2-(prop-1-en-2-yl)-1H-indol-3-yl)urea; 1-(3,5-difluorophenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(3-methyl-5-(trifluoromethyl)phenyl)urea; and 1-(3,5-bis(trifluoromethyl)phenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; or a pharmaceutically acceptable salt or solvate thereof. In another aspect, the invention relates to a pharmaceutical composition comprising a hydroxyphenyl compound as defined above, or a pharmaceutically acceptable salt or solvate thereof, and at least one pharmaceutically acceptable excipient. In another aspect, the invention relates to a hydroxyphenyl compound as defined above, in combination with an anticancer agent, for use in treating cancer. In another aspect, the invention relates to a compound comprising a hydroxyphenyl moiety as defined above, for use in increasing the sensitivity of cancer cells to an anticancer agent in a treatment of cancer. DETAILED DESCRIPTION OF THE INVENTION As detailed above, the invention relates to compounds of Formula I, as well as their pharmaceutically acceptable salts or solvates. Preferred compounds of Formula I or pharmaceutically acceptable salts or solvates thereof are those wherein one or more of A, L, Y, Z, R1, R2 and Cy are defined as follows: A is selected from –NH–, –N(Me)– and –O–; in particular A is –NH–;
Figure imgf000013_0001
5
Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O–, –CH2– and –C(O)NH–; in particular Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O– and –CH2–; more particularly Y is selected from –NH–, –NH–CH2–, –O– and –CH2–; still more particularly Y is–NH–; Z is O or S; in particular Z is O; R1 and R2 are independently selected from H and OH, with the proviso that at least one of R1 and R2 is H and that R1 and R2 are not both H; in particular R1 is OH and R2 is H; Cy is selected from: -
Figure imgf000015_0001
wherein R3 is selected from H, C1-C4-alkyl and halogen; in particular R3 is selected from H, C1-C4-alkyl and F; more particularly R3 is H or C1-C4-alkyl; still more particularly R3 is Me or t-Bu; R4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; in particular R4 is selected from H, C1-C3-alkyl and cyclopropyl; more particularly R4 is H or C1- C3-alkyl; still more particularly R4 is H; R5 is H or C1-C4-alkyl; in particular R5 is H or C1-C3-alkyl; more particularly R5 is H or Me; still more particularly R5 is H; R6 is selected from H, C1-C4-alkyl, C3-C4-cycloalkyl, halogen, C(O)NR8R9, NHC(O)R10 and C(O)OR11; in particular R6 is selected from H, C1-C3-alkyl, I, C(O)NR8R9, NHC(O)R10 and C(O)OR11; more particularly R6 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl, still more particularly R6 is selected from H, C1-C3-alkyl and cyclopropyl; even more particularly R6 is H or C1-C3-alkyl; wherein R8 are R9 are independently selected from hydrogen, C1-C4-alkyl and C3- C4-cycloalkyl; in particular R8 are R9 are independently selected from hydrogen, C1-C3-alkyl and cyclopropyl; R9 is C1-C4-alkyl; in particular R9 is C1-C3-alkyl; and R10 is selected from H and C1-C4-alkyl; in particular R10 is selected from H and C1-C3-alkyl; and R7 is H or C1-C4-alkyl; in particular R7 is H or C1-C3-alkyl; more particularly R7 is H or Me; still more particularly R7 is H; with the proviso that R3, R4, R5, R6 and R7, are not all H; -
Figure imgf000016_0001
wherein R11 and R12 are independently selected from hydrogen and C1-C4-alkyl; in particular R11 and R12 are independently selected from hydrogen and C1-C3- alkyl; more particularly R11 and R12 are H or Me; still more particularly R11 and R12 are Me; - - - - - with the proviso that, when
Figure imgf000017_0001
are not all H; in particular Cy is selected from: - wherein R3 is selected from H, C1-C4-alkyl and halogen; in particular R3 is selected from H, C1-C4-alkyl and F; more particularly R3 is H or C1-C4-alkyl; still more particularly R3 is Me or t-Bu; R4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; in particular R4 is selected from H, C1-C3-alkyl and cyclopropyl; more particularly R4 is H or C1- C3-alkyl; still more particularly R4 is H; R5 is H or C1-C4-alkyl; in particular R5 is H or C1-C3-alkyl; more particularly R5 is H or Me; still more particularly R5 is H; R6 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl, in particular R6 is selected from H, C1-C3-alkyl and cyclopropyl; more particularly R6 is H or C1- C3-alkyl; R7 is H or C1-C4-alkyl; in particular R7 is H or C1-C3-alkyl; more particularly R7 is H or Me; still more particularly R7 is H; with the proviso that R3, R4, R5, R6 and R7, are not all H; -
Figure imgf000018_0001
wherein R11 and R12 are independently selected from hydrogen and C1-C4-alkyl; in particular R11 and R12 are independently selected from hydrogen and C1-C3- alkyl; more particularly R11 and R12 are H or Me; still more particularly R11 and R12 are Me; - - - - with the proviso that, when
Figure imgf000019_0001
are not all H; more particularly Cy is selected from: -
Figure imgf000019_0002
wherein R3 is selected from H, C1-C4-alkyl and halogen; in particular R3 is selected from H, C1-C4-alkyl and F; more particularly R3 is H or C1-C4-alkyl; still more particularly R3 is Me or t-Bu; R4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; in particular R4 is selected from H, C1-C3-alkyl and cyclopropyl; more particularly R4 is H or C1- C3-alkyl; still more particularly R4 is H; R5 is H or C1-C4-alkyl; in particular R5 is H or C1-C3-alkyl; more particularly R5 is H or Me; still more particularly R5 is H; R6 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl, in particular R6 is selected from H, C1-C3-alkyl and cyclopropyl; more particularly R6 is H or C1- C3-alkyl; R7 is H or C1-C4-alkyl; in particular R7 is H or C1-C3-alkyl; more particularly R7 is H or Me; still more particularly R7 is H; with the proviso that R3, R4, R5, R6 and R7, are not all H; - - -
Figure imgf000020_0001
with the proviso that, when are not all H, and the compound of Formula I is none of the following: (R)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; (S)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N,4-dimethylbenzamide; (R)-4-fluoro-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)benzamide; (S)-N-ethyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; (S)-N-cyclopropyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; (S)-N1-(5-carbamoyl-2-fluorophenyl)-N2-(4-(4-hydroxyphenyl)butan-2-yl)oxalamide; (R)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N,4-dimethylbenzamide; (S)-N-(3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylphenyl)acetamide; (S)-1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(5-iodo-2-methylphenyl)urea; (S)-N-(3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylphenyl)propionamide; (S)-3-acetamido-N-(4-(4-hydroxyphenyl)butan-2-yl)-4-methylbenzamide; (S)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)benzamide; (R)-N-ethyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; Methyl (S)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzoate; (R)-N-(3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylphenyl)isobutyramide; (S)-1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; (S)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N,N,4-trimethylbenzamide; (S)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; (R)-N-(3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylphenyl)acetamide; (R)-N1-(5-carbamoyl-2-methylphenyl)-N2-(4-(4-hydroxyphenyl)butan-2-yl)oxalamide; N-ethyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; 3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N,4-dimethylbenzamide; 3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; 3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N,2-dimethylbenzamide; 4-fluoro-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)benzamide; 4-fluoro-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N-methylbenzamide; N-cyclopropyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; N-(3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)phenyl)isobutyramide; N-(5-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-2-methylphenyl)acetamide; (S)-1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(m-tolyl)urea; 1-(2,5-dimethylphenyl)-3-(4-phenylbutan-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-hydroxyphenethyl)urea; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(2-fluorophenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(2-ethyl-6-methylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; N-(2,5-dimethylphenyl)-4-(3-hydroxyphenyl)piperazine-1-carboxamide; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-phenylurea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(m-tolyl)urea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(p-tolyl)urea; 1-cyclohexyl-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; and 1-(benzo[d][1,3]dioxol-5-yl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea. Halogens include a fluorine atom, an iodine atom, a chlorine atom and a bromine atom. C1-C4-alkyl include butyl, in particular n-butyl, isobutyl, sec-butyl or tert-butyl; propyl, in particular n-propyl or isopropyl; ethyl or methyl. C3-C4-cycloalkyl include cyclopropyl and cyclobutyl. In one embodiment, the compound of formula I may be in racemic or optically active form. In one embodiment, the compound of formula I may be in racemic form. In one embodiment, the compound of formula I may be in optically active form. In one embodiment, the compounds of Formula I are those wherein A is –NH–. In one embodiment, the compounds of Formula I are those wherein A is –N(Me)–. In one embodiment, the compounds of Formula I are those wherein A is –O–. In one embodiment, the compounds of Formula I are those wherein L is selected from
Figure imgf000023_0001
In one embodiment, the compounds of Formula I are those wherein L is selected from
Figure imgf000023_0002
In one embodiment, the compounds of Formula I are those wherein L is selected from
Figure imgf000024_0001
In one embodiment, the compounds of Formula I are those wherein L is selected from
Figure imgf000024_0002
In one embodiment, the compounds of Formula I are those wherein L is
Figure imgf000025_0001
. In one embodiment, the compounds of Formula I are those wherein L is
Figure imgf000025_0002
. In one embodiment, the compounds of Formula I are those wherein L is
Figure imgf000025_0003
. In one embodiment, the compounds of Formula I are those wherein L is
Figure imgf000025_0004
. In one embodiment, the compounds of Formula I are those wherein L is
Figure imgf000025_0005
. In one embodiment, the compounds of Formula I are those wherein L is
Figure imgf000025_0006
. In one embodiment, the compounds of Formula I are those wherein
Figure imgf000025_0007
In one embodiment, the compounds of Formula I are those wherein
Figure imgf000025_0008
In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein
Figure imgf000026_0001
In one embodiment, the compounds of Formula I are those wherein
Figure imgf000026_0002
In one embodiment, the compounds of Formula I are those wherein L is
Figure imgf000026_0003
. In one embodiment, the compounds of Formula I are those wherein
Figure imgf000026_0004
In one embodiment, the compounds of Formula I are those wherein
Figure imgf000026_0005
In one embodiment, the compounds of Formula I are those wherein
Figure imgf000026_0006
In one embodiment, the compounds of Formula I are those wherein
Figure imgf000026_0007
In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein
Figure imgf000027_0001
In one embodiment, the compounds of Formula I are those wherein
Figure imgf000027_0002
In one embodiment, the compounds of Formula I are those wherein Y is selected from –NH– , –N(Me)–, –NH–CH2–, –O– and –CH2–; in particular Y is selected from –NH–, –NH–CH2– , –O– and –CH2–. In one embodiment, the compounds of Formula I are those wherein Y is –NH–. In one embodiment, the compounds of Formula I are those wherein Y is –N(Me)–. In one embodiment, the compounds of Formula I are those wherein Y is –NH–CH2–. In one embodiment, the compounds of Formula I are those wherein Y is –O–. In one embodiment, the compounds of Formula I are those wherein Y is –CH2–. In one embodiment, the compounds of Formula I are those wherein Z is O. In one embodiment, the compounds of Formula I are those wherein Z is S. In one embodiment, the compounds of Formula I are those wherein R1 is OH and R2 is H. In one embodiment, the compounds of Formula I are those wherein R1 is H and R2 is OH. In one embodiment, the compounds of Formula I are those wherein wherein R3 is selected from H, C1-C4-alkyl and halogen; in particular R3 is selected from H, C1-C4-alkyl and F; more particularly R3 is H or C1-C4-alkyl; still more particularly R3 is Me or t-Bu; R4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; in particular R4 is selected from H, C1-C3-alkyl and cyclopropyl; more particularly R4 is H or C1- C3-alkyl; still more particularly R4 is H; R5 is H or C1-C4-alkyl; in particular R5 is H or C1-C3-alkyl; more particularly R5 is H or Me; still more particularly R5 is H; R6 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl, in particular R6 is selected from H, C1-C3-alkyl and cyclopropyl; more particularly R6 is H or C1- C3-alkyl; R7 is H or C1-C4-alkyl; in particular R7 is H or C1-C3-alkyl; more particularly R7 is H or Me; still more particularly R7 is H; with the proviso that R3, R4, R5, R6 and R7, are not all H. In one embodiment, the compounds of Formula I are those wherein
Figure imgf000028_0001
wherein R11 and R12 are independently selected from hydrogen and C1-C4-alkyl; in particular R11 and R12 are independently selected from hydrogen and C1-C3- alkyl; more particularly R11 and R12 are H or Me; still more particularly R11 and R12 are Me. In one embodiment, the compounds of Formula I are those wherein In one embodiment, the compounds of Formula I are those wherein
Figure imgf000029_0001
In one embodiment, the compounds of Formula I are those wherein
Figure imgf000029_0002
In one embodiment, the compounds of Formula I are those wherein
Figure imgf000029_0003
In one embodiment, the compounds of Formula I are those wherein
Figure imgf000029_0004
In one embodiment, the compounds of Formula I are those wherein Cy is selected from
Figure imgf000029_0005
In one embodiment, the compounds of Formula I are those wherein
Figure imgf000030_0001
In one embodiment, the compounds of Formula I are those of Formula II:
Figure imgf000030_0002
II, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, Y and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula IIa:
Figure imgf000031_0001
IIa, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A and Y are as defined above with respect to Formula I and any of its embodiments. Particular compounds of Formula IIa are those wherein R1, R2, L, A and Y are defined as follows: R1 and R2 are independently selected from H and OH, with the proviso that at least one of R1 and R2 is H and that R1 and R2 are not both H; in particular R1 is OH and R2 is H; In one embodiment, the compounds of Formula I are those of Formula IIb:
Figure imgf000032_0001
IIb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A, Y and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula IIc:
Figure imgf000032_0002
IIc, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula III:
Figure imgf000033_0001
III, or pharmaceutically acceptable salts or solvates thereof, wherein L, A, Z, Y and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula IIIa:
Figure imgf000033_0002
IIIa, or pharmaceutically acceptable salts or solvates thereof, wherein L, A, Z and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula IIIb: IIIb, or pharmaceutically acceptable salts or solvates thereof, wherein A, Z, Y and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula IIIc:
Figure imgf000034_0001
IIIc, or pharmaceutically acceptable salts or solvates thereof, wherein A, Z and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula IV: IV, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, Y and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula IVa:
Figure imgf000035_0001
IVa, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula IVb:
Figure imgf000036_0001
IVb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, Y and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula IVc:
Figure imgf000036_0002
IVc, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula V:
Figure imgf000037_0001
V, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula Va:
Figure imgf000037_0002
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula Vb:
Figure imgf000038_0001
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula Vc:
Figure imgf000038_0002
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula VI:
Figure imgf000039_0001
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula VIa:
Figure imgf000039_0002
VIa, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, and L are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula VIb:
Figure imgf000040_0001
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula VIc:
Figure imgf000040_0002
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula VId: or pharmaceutically acceptable salts or solvates thereof, wherein R1 and R2 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula VII:
Figure imgf000041_0001
VII, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, Z, Y, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula VIIa:
Figure imgf000042_0001
VIIa, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, Z and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula VIIb:
Figure imgf000042_0002
VIIb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A, Z, Y, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula VIIc:
Figure imgf000043_0001
VIIc, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A, Z and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula VIII:
Figure imgf000044_0001
VIII, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, Y, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. Preferred compounds of Formula VIII or pharmaceutically acceptable salts or solvates thereof are those wherein one or more of R1, R2, L, A, Y, R3, R4, R5, R6 and R7 are defined as follows: R1 and R2 are independently selected from H and OH, with the proviso that at least one of R1 and R2 is H and that R1 and R2 are not both H; in particular R1 is OH and R2 is H; A is selected from –NH–, –N(Me)– and –O–; in particular A is –NH–;
Figure imgf000044_0002
5 more particularly L is selected from
Figure imgf000045_0001
,
Figure imgf000045_0002
Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O–, –CH2– and –C(O)NH–; in particular Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O– and –CH2–; more particularly Y is selected from –NH–, –NH–CH2–, –O– and –CH2–; still more particularly Y is–NH–; R3 is selected from H, C1-C4-alkyl and halogen; in particular R3 is selected from H, C1-C4- alkyl and F; more particularly R3 is H or C1-C4-alkyl; still more particularly R3 is Me or t- Bu; R4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; in particular R4 is selected from H, C1-C3-alkyl and cyclopropyl; more particularly R4 is H or C1-C3-alkyl; still more particularly R4 is H; R5 is H or C1-C4-alkyl; in particular R5 is H or C1-C3-alkyl; more particularly R5 is H or Me; still more particularly R5 is H; R6 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl, in particular R6 is selected from H, C1-C3-alkyl and cyclopropyl; more particularly R6 is H or C1-C3-alkyl; R7 is H or C1-C4-alkyl; in particular R7 is H or C1-C3-alkyl; more particularly R7 is H or Me; still more particularly R7 is H; with the proviso that R3, R4, R5, R6 and R7, are not all H. In one embodiment, the compounds of Formula I are those of Formula VIIIb:
Figure imgf000047_0001
VIIIb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A, Y, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula VIIIc:
Figure imgf000047_0002
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula IX:
Figure imgf000048_0001
IX, or pharmaceutically acceptable salts or solvates thereof, wherein L, A, Y, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula IXa:
Figure imgf000048_0002
or pharmaceutically acceptable salts or solvates thereof, wherein L, A and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula IXb:
Figure imgf000049_0001
IXb, or pharmaceutically acceptable salts or solvates thereof, wherein A, Y, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula IXc:
Figure imgf000049_0002
or pharmaceutically acceptable salts or solvates thereof, wherein A and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula X:
Figure imgf000050_0001
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula Xa:
Xa, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula Xb:
Figure imgf000051_0001
Xb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula Xc:
Figure imgf000052_0001
Xc, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula XI:
Figure imgf000052_0002
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula XIa:
Figure imgf000053_0001
XIa, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula XIb:
Figure imgf000054_0001
XIb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula XIc:
Figure imgf000054_0002
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula XII:
Figure imgf000055_0001
XII, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula XIIa:
Figure imgf000055_0002
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula XIIb:
Figure imgf000056_0001
XIIb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula XIIc: XIIc, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula XIII:
Figure imgf000057_0001
XIII, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula XIIIa:
Figure imgf000058_0001
XIIIa, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula XIIIb:
Figure imgf000058_0002
XIIIb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula XIIIc:
Figure imgf000059_0001
XIIIc, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula XIV:
Figure imgf000059_0002
XIV, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula XIVa:
Figure imgf000060_0001
XIVa, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and L are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula XIVb:
Figure imgf000060_0002
XIVb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds of Formula I are those of Formula XIVc:
Figure imgf000061_0001
or pharmaceutically acceptable salts or solvates thereof, wherein R1 and R2 are as defined above with respect to Formula I and any of its embodiments. Particularly preferred compounds of the invention are those listed in Table 1 hereafter: Table 1
Figure imgf000061_0002
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
The invention further concerns hydroxyphenyl compounds listed in Table 1b hereafter. The term “hydroxyphenyl compound” as used herein means a compound comprising a hydroxyphenyl moiety. Table 1b
Figure imgf000066_0002
Figure imgf000067_0001
The compounds of the invention can be prepared by different ways with reactions known by the person skilled in the art. Typical routes of synthesis are described thereafter The compounds of the invention (i.e. the compounds of Formula I and its subformulae as described above and the hydroxyphenyl compounds of Table 1b above) are indeed capable of inhibiting IRE RNase activity and sensitizing cancer cells to anticancer drugs. They further have the advantage of sensitizing cancer cells, in particular GGM cells to anticancer drugs, in particular alkylating agents. The invention thus also provides the use of the compounds of the invention, or pharmaceutically acceptable salts or solvates thereof, as sensitizers for chemotherapy of cancer cells. Accordingly, the invention relates to the use of compounds of the invention, or pharmaceutically acceptable salts or solvates thereof, for the treatment of cancer, particularly as sensitizers for chemotherapy of cancer cells. APPLICATIONS Unexpectedly, the inventors have discovered that the compounds of the invention, including the 41 compounds listed in the proviso above, may be used as inhibitors of IRE1 RNase activity and have the potentential of increasing the sensitivity of cancer cells to an anticancer agent in a treatment of cancer. In fact, and without wanting to be tied to any theory whatsoever, the inventors think the specific structure of the compounds of the invention allow them to interact with the ATP kinase binding pocket of IRE1. The compounds of the invention are able to inhibit IRE1 RNase activity and to increase the sensitivity of cancer cells to an anticancer agent in a treatment of cancer, in particular glioblastoma, triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer. The compounds of the invention as defined above, can thus be used for treating cancer, particularly as sensitizers for chemotherapy of cancer cells, aiming at improving the chemotherapy effect of the cancer treatment, preventing tolerance and decreasing toxicity and adverse effects. The invention thus relates to a compound of Formula I:
Figure imgf000068_0001
I, a pharmaceutically acceptable salt or solvate thereof, wherein A is selected from –NH–, –N(Me)– and –O–;
Figure imgf000068_0002
, Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O–, –CH2– and –C(O)NH– or is a single bond; Z is O or S; R1 and R2 are independently selected from H and OH, with the proviso that at least one of R1 and R2 is H; Cy is selected from: -
Figure imgf000069_0001
R3 is selected from H, C1-C4-alkyl, and halogen; R4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; R5 is H or C1-C4-alkyl; R6 is selected from H, C1-C4-alkyl, C3-C4-cycloalkyl, halogen, C(O)NR8R9, NHC(O)R10 and C(O)OR11 wherein R8 are R9 are independently selected from hydrogen, C1-C4-alkyl and C3- C4-cycloalkyl; R9 is C1-C4-alkyl; and R10 is selected from H and C1-C4-alkyl; and R7 is H or C1-C4-alkyl; with the proviso that R3, R4, R5, R6 and R7, are not all H; -
Figure imgf000070_0001
wherein R11 and R12 are independently selected from hydrogen and C1-C4-alkyl; - - - -
Figure imgf000070_0002
- in combination with an anticancer agent, for use in treating cancer. In one embodiment, the compound for use of formula I may be in racemic or optically active form. In one embodiment, the compound for use of formula I may be in racemic form. In one embodiment, the compound for use of formula I is in optically active form. In one embodiment, particular compounds for use according to the invention are compounds of formula I, or pharmaceutically acceptable salts or solvates thereof, wherein A, L, Y, Z, R1, R2 and Cy are defined as follows: A is selected from –NH–, –N(Me)– and –O–; in particular A is –NH–;
Figure imgf000071_0001
5 more particularly L is selected from
Figure imgf000072_0001
,
Figure imgf000072_0002
Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O–, –CH2– and –C(O)NH– or is a single bond; in particular Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O–, –CH2– and – C(O)NH–; more particularly Y is selected from –NH– or –C(O)NH–; Z is O or S; in particular Z is O; R1 and R2 are independently selected from H and OH, with the proviso that at least one of R1 and R2 is H; in particular R1 is OH and R2 is H; Cy is selected from: -
Figure imgf000073_0001
wherein R3 is selected from H, C1-C4-alkyl and halogen; in particular R3 is selected from H, C1-C4-alkyl and F; more particularly R3 is C1-C4-alkyl or F; still more particularly R3 is Me or F; R4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; in particular R4 is selected from H, C1-C3-alkyl and cyclopropyl; more particularly R4 is H or C1- C3-alkyl; still more particularly R4 is H; R5 is H or C1-C4-alkyl; in particular R5 is H or C1-C3-alkyl; more particularly R5 is H or Me; still more particularly R5 is H; R6 is selected from H, C1-C4-alkyl, C3-C4-cycloalkyl, halogen, C(O)NR8R9, NHC(O)R10 and C(O)OR11; in particular R6 is selected from H, C1-C3-alkyl, I, C(O)NR8R9, NHC(O)R10 and C(O)OR11; more particularly R6 is selected from C1-C3-alkyl, C(O)NR8R9, NHC(O)R10 and C(O)OR11; still more particularly R6 is selected from C1-C3-alkyl and C(O)NR8R9; wherein R8 are R9 are independently selected from hydrogen, C1-C4-alkyl and C3- C4-cycloalkyl; in particular R8 are R9 are independently selected from hydrogen, C1-C3-alkyl and cyclopropyl; R9 is C1-C4-alkyl; in particular R9 is C1-C3-alkyl; and R10 is selected from H and C1-C4-alkyl; in particular R10 is selected from H and C1-C3-alkyl; and R7 is H or C1-C4-alkyl; in particular R7 is H or C1-C3-alkyl; more particularly R7 is H or Me; still more particularly R7 is H; -
Figure imgf000074_0001
wherein R11 and R12 are independently selected from hydrogen and C1-C4-alkyl; in particular R11 and R12 are independently selected from hydrogen and C1-C3- alkyl; more particularly R11 and R12 are H or Me; still more particularly R11 and R12 are Me; - - -
Figure imgf000074_0002
R3 is selected from H, C1-C4-alkyl and halogen; in particular R3 is selected from H, C1-C4-alkyl and F; more particularly R3 is C1-C4-alkyl or F; still more particularly R3 is Me or F; R4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; in particular R4 is selected from H, C1-C3-alkyl and cyclopropyl; more particularly R4 is H or C1- C3-alkyl; still more particularly R4 is H; R5 is H or C1-C4-alkyl; in particular R5 is H or C1-C3-alkyl; more particularly R5 is H or Me; still more particularly R5 is H; R6 is selected from H, C1-C4-alkyl, C3-C4-cycloalkyl, halogen, C(O)NR8R9, NHC(O)R10 and C(O)OR11; in particular R6 is selected from H, C1-C3-alkyl, I, C(O)NR8R9, NHC(O)R10 and C(O)OR11; more particularly R6 is selected from C1-C3-alkyl, C(O)NR8R9, NHC(O)R10 and C(O)OR11; still more particularly R6 is selected from C1-C3-alkyl and C(O)NR8R9; wherein R8 are R9 are independently selected from hydrogen, C1-C4-alkyl and C3- C4-cycloalkyl; in particular R8 are R9 are independently selected from hydrogen, C1-C3-alkyl and cyclopropyl; R9 is C1-C4-alkyl; in particular R9 is C1-C3-alkyl; and R10 is selected from H and C1-C4-alkyl; in particular R10 is selected from H and C1-C3-alkyl; and R7 is H or C1-C4-alkyl; in particular R7 is H or C1-C3-alkyl; more particularly R7 is H or Me; still more particularly R7 is H. In one embodiment, the compounds for use of Formula I are those wherein A is –NH–. In one embodiment, the compounds for use of Formula I are those wherein A is –N(Me)–. In one embodiment, the compounds for use of Formula I are those wherein A is –O–. In one embodiment, the compounds for use of Formula I are those wherein L is selected
Figure imgf000076_0001
, In one embodiment, the compounds of Formula I are those wherein
Figure imgf000077_0001
. In one embodiment, the compounds for use of Formula I are those wherein Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O– and –CH2–; in particular Y is selected from –NH– , –NH–CH2–, –O– and –CH2–. In one embodiment, the compounds for use of Formula I are those wherein Y is –NH–. In one embodiment, the compounds for use of Formula I are those wherein Y is –N(Me)–. In one embodiment, the compounds for use of Formula I are those wherein Y is –NH–CH2– . In one embodiment, the compounds for use of Formula I are those wherein Y is –O–. In one embodiment, the compounds for use of Formula I are those wherein Y is –CH2–. In one embodiment, the compounds for use of Formula I are those wherein Z is O. In one embodiment, the compounds for use of Formula I are those wherein Z is S. In one embodiment, the compounds for use for use of Formula I are those wherein R1 is OH and R2 is H. In one embodiment, the compounds for use of Formula I are those wherein R1 is H and R2 is OH. In one embodiment, the compounds for use of Formula I are those of Formula II:
Figure imgf000078_0001
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, Y and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula IIa:
Figure imgf000078_0002
IIa, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula IIb:
Figure imgf000079_0001
IIb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A, Y and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula IIc:
Figure imgf000079_0002
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula III:
Figure imgf000080_0001
or pharmaceutically acceptable salts or solvates thereof, wherein L, A, Z, Y and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula IIIa:
Figure imgf000080_0002
IIIa, or pharmaceutically acceptable salts or solvates thereof, wherein L, A, Z and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula IIIb:
Figure imgf000081_0001
IIIb, or pharmaceutically acceptable salts or solvates thereof, wherein A, Z, Y and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula IIIc:
Figure imgf000081_0002
IIIc, or pharmaceutically acceptable salts or solvates thereof, wherein A, Z and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula IV: IV, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, Y and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula IVa:
Figure imgf000082_0001
IVa, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula IVb:
Figure imgf000083_0001
IVb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, Y and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula IVc:
Figure imgf000083_0002
IVc, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula V:
Figure imgf000084_0001
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula Va:
Figure imgf000084_0002
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula Vb:
Figure imgf000085_0001
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula Vc:
Figure imgf000085_0002
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula VI:
Figure imgf000086_0001
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula VIa:
Figure imgf000086_0002
VIa, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, and L are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula VIb:
Figure imgf000087_0001
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula VIc:
Figure imgf000087_0002
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula VId: or pharmaceutically acceptable salts or solvates thereof, wherein R1 and R2 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula VII:
Figure imgf000088_0001
VII, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, Z, Y, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula VIIa:
Figure imgf000089_0001
VIIa, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, Z and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula VIIb:
Figure imgf000089_0002
VIIb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A, Z, Y, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula VIIc:
Figure imgf000090_0001
VIIc, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A, Z and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula VIII:
Figure imgf000091_0001
VIII, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, Y, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. Preferred compounds for use of Formula VI or pharmaceutically acceptable salts or solvates thereof are those wherein one or more of R1, R2, L, A, Y, R3, R4, R5, R6 and R7 are defined as follows: R1 and R2 are independently selected from H and OH, with the proviso that at least one of R1 and R2 is H; in particular R1 is OH and R2 is H; A is selected from –NH–, –N(Me)– and –O–; in particular A is –NH–;
Figure imgf000091_0002
5 more particularly L is selected from
Figure imgf000092_0001
,
Figure imgf000092_0002
Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O–, –CH2– and –C(O)NH– or is a single bond; in particular Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O– and –CH2–; more particularly Y is selected from –NH–, –NH–CH2–, –O– and –CH2–; still more particularly Y is–NH–; R3 is selected from H, C1-C4-alkyl and halogen; in particular R3 is selected from H, C1-C4- alkyl and F; more particularly R3 is H or C1-C4-alkyl; still more particularly R3 is Me or t- Bu; R4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; in particular R4 is selected from H, C1-C3-alkyl and cyclopropyl; more particularly R4 is H or C1-C3-alkyl; still more particularly R4 is H; R5 is H or C1-C4-alkyl; in particular R5 is H or C1-C3-alkyl; more particularly R5 is H or Me; still more particularly R5 is H; R6 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl, in particular R6 is selected from H, C1-C3-alkyl and cyclopropyl; more particularly R6 is H or C1-C3-alkyl; R7 is H or C1-C4-alkyl; in particular R7 is H or C1-C3-alkyl; more particularly R7 is H or Me; still more particularly R7 is H; with the proviso that R3, R4, R5, R6 and R7, are not all H. In one embodiment, the compounds for use of Formula I are those of Formula VIIIb:
Figure imgf000094_0001
VIIIb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A, Y, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula VIIIc:
Figure imgf000094_0002
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula IX:
Figure imgf000095_0001
IX, or pharmaceutically acceptable salts or solvates thereof, wherein L, A, Y, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula IXa:
Figure imgf000095_0002
or pharmaceutically acceptable salts or solvates thereof, wherein L, A and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula IXb:
Figure imgf000096_0001
IXb, or pharmaceutically acceptable salts or solvates thereof, wherein A, Y, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula IXc:
Figure imgf000096_0002
or pharmaceutically acceptable salts or solvates thereof, wherein A and Y are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula X:
Figure imgf000097_0001
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula Xa:
Xa, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula Xb:
Figure imgf000098_0001
Xb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula Xc:
Figure imgf000099_0001
Xc, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula XI:
Figure imgf000099_0002
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula XIa:
Figure imgf000100_0001
XIa, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula XIb:
Figure imgf000101_0001
XIb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula XIc:
Figure imgf000101_0002
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula XII:
Figure imgf000102_0001
XII, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, Y, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula XIIa:
Figure imgf000102_0002
or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, Y, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula XIIb:
Figure imgf000103_0001
XIIb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula XIIc: XIIc, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula XIII:
Figure imgf000104_0001
XIII, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, A, Y, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula XIIIa:
Figure imgf000105_0001
XIIIa, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula XIIIb:
Figure imgf000105_0002
XIIIb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, A, R3, R4, R5, R6 and R7 are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula XIIIc:
Figure imgf000106_0001
XIIIc, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and A are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula XIV:
Figure imgf000106_0002
XIV, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2, L, and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula XIVa:
Figure imgf000107_0001
XIVa, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and L are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compounds for use of Formula I are those of Formula XIVb:
Figure imgf000107_0002
XIVb, or pharmaceutically acceptable salts or solvates thereof, wherein R1, R2 and Cy are as defined above with respect to Formula I and any of its embodiments. In one embodiment, the compoundsfor use of Formula I are those of Formula XIVc:
Figure imgf000108_0001
XIVc, or pharmaceutically acceptable salts or solvates thereof, wherein R1 and R2 are as defined above with respect to Formula I and any of its embodiments. The compounds for use according to the invention therefore include compounds of Formula I and subformulae as defined above, in particular compounds of Table 2 below. Table 2
Figure imgf000108_0002
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
The invention also concerns hydroxyphenyl compounds as listed in Table 2b hereafter, in combination with an anticancer agent, for use in treating cancer: Table 2b
Figure imgf000115_0001
The invention also relates to the compounds of the invention as described above for use in increasing the sensitivity of cancer cells to an anticancer agent in a treatment of cancer. In one embodiment, the compounds for use according to the invention are selected from compounds 2, 3, 4, 5, 6, 7, 9, 11, 12, 14, 16, 17, 18, 20, 22, 23, 24, 27, 28, S-28, R-28, 30, 31, 32, 40, 41, 42, 43, 44, 46, 47, 50, 52, 54, 57, 58, 59, 63, 64 and 66 of Tables 2 and 2b above. In one embodiment, the compounds for use according to the invention are selected from compounds 2, 3, 4, 5, 6, 7, 9, 11, 12, 14, 16, 17, 18, 20, 22, 23, 24, 27, 28, S-28, R-28, 30, 31, 32, 40, 41, 42, 43, 44, 46, 47, 50, 52, 54, 63, 64 and 66 of Table 2 above. In one embodiment, the compounds for use according to the invention are selected from compounds 57, 58 and 59 of Table 2b above. In one embodiment, the compounds for use according to the invention are selected from compounds 28, 33, 34, 35, 36, 37 and 38 of Table 2 above, in particular compounds 28 and 33 of Table 2 above. In one embodiment, the compound for use according to the invention is compound 28 of Table 2 above. Treatment of cancer, in particular chemotherapy, is a type of treatment that uses one or more anticancer agents. Anticancer agents, or cytotoxic agents, within the meaning of the present invention include, but are not limited to, alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents. Preferably, the anticancer agent is an alkylating agent. Preferred alkylating agent is temozolamide. The compounds of the invention are therefore useful in the treatment of cancers, and particularly cancers that may be treated by anticancer agents selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly cancers that may be treated by alkylating agents. Cancers that may be treated by anticancer agents selected from alkylating agents, anti- microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, within the meaning of the present invention include, but are not limited to, glioblastoma, triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer. A preferred cancer that may be treated by alkylating agents is glioblastoma. Thus, in one embodiment, there is provided a compound of the present invention, or a pharmaceutically acceptable salt or solvate thereof, in combination with an anticancer agent, for use in treating cancer. The invention thus also relates to a compound of the present invention, i.e. a compound of Formula I, or any of its subformulae as defined above, in particular a compound of Table 2 above, or a hydroxyphenyl compound of Table 2b, or a pharmaceutically acceptable salt or solvate thereof, in combination with an anticancer agent, in particular an anticancer agent selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly an alkylating agent, even more particularlry temozolamide, for use in treating cancer, preferably cancers that may be treated by anticancer agents selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly a cancer selected from glioblastoma (GB), triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer, even more particularly cancers that may be treated by alkylating agents, still more preferably glioblastoma. In other terms, the invention also relates to a method of treating cancer, in particular cancers that may be treated by anticancer agents selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly a cancer selected from glioblastoma, triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer, even more particularly cancers that may be treated by alkylating agents, still more particularly glioblastoma, comprising the administration of a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt or solvate thereof, in combination with an anticancer agent, in particular an anticancer agent selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly an alkylating agent, even more particularly temozolamide, to a patient in need of such treatment. Preferably the patient is a warm-blooded animal, more preferably a human. The cancers that may be treated by an alkylating agent are preferably those defined above. The invention further provides the use of a compound of the present invention, or a pharmaceutically acceptable salt or solvates thereof, in combination with an anticancer agent, in particular an anticancer agent selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly an alkylating agent, even more particularly temozolamide, for the manufacture of a medicament for use in treating cancer, in particular cancers that may be treated by anticancer agents selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly a cancer selected from glioblastoma, triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer, even more particularly cancers that may be treated by an alkylating agent, still more particularly glioblastoma. Preferably the patient is a warm-blooded animal, more preferably a human. The cancers that may be treated by an alkylating agent are preferably those defined above. By “in combination”, it is meant a combined preparation wherein the active ingredients are physically together in the same preparation or physically separated for use in a combined therapy by simultaneous administration or sequential administration to the patient. Hence, according to the present invention, a compound of the invention or a pharmaceutically acceptable salt or solvate thereof and the anticancer agent are administered to the patient in the same preparation or in a separate form, either simultaneously or sequentially, for the treatment of cancer. According to a further feature of the present invention, there is provided a compound of the present invention, or a pharmaceutically acceptable salt or solvate thereof, for use in increasing the sensitivity of cancer cells to an anticancer agent in a treatment of cancer. The invention thus relates to a compound of the present invention, i.e. a compound of Formula I and any of its embodiments, or any of the subformulae of Formula I as defined above, in particular a compound of Table 2 above, or a hydrophenyl compound of Table 2b, or a pharmaceutically acceptable salt or solvate thereof, for use in increasing the sensitivity of cancer cells to an anticancer agent, in particular an anticancer agent selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly an alkylating agent, still more particularly temozolamide, in a treatment of cancer, in particular cancers that may be treated by anticancer agents selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly a cancer selected from glioblastoma, triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer, even more particularly cancer that may be treated by an alkylating agent, still more particularly glioblastoma. In other terms, the invention also relates to a method for increasing the sensitivity of cancer cells to an anticancer agent, in particular an anticancer agent selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly an alkylating agent, even more particularly temozolomide, in a treatment of cancer, in particular cancers that may be treated by anticancer agents selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly a cancer selected from glioblastoma, triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer, even more particularly cancers that may be treated by alkylating agents, still more particularly glioblastoma, comprising the administration of a therapeutically effective amount of a compound of the present invention, i.e. a compound of Formula I and any of its embodiments, or any of its subformulae as defined above, or a hydrophenyl compound of Table 2b, or a pharmaceutically acceptable salt or solvate thereof, in combination with an anticancer agent, in particular an anticancer agent selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly an alkylating agent, even more particularly temozolamide, to a patient in need thereof. Preferably the patient is a warm-blooded animal, more preferably a human. The invention further provides the use of a compound of the present invention, i.e. a compound of Formula I and any of its embodiments, or any of its subformulae as defined above, or a hydrophenyl compound of Table 2b, or a pharmaceutically acceptable salt or solvates thereof, for the manufacture of a medicament for use in sensitizing cancer cells to an anticancer agent, in particular an anticancer agent selected from alkylating agents, anti- microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly an alkylating agent, even more particularly temozolomide, in a treatment of cancer, in particular cancers that may be treated by anticancer agents selected from alkylating agents, anti-microtubule agents, antimetabolites, topoisomerase inhibitors, cytotoxic antibiotics and antiangiogenic agents, more particularly a cancer selected from glioblastoma, triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer, even more particularly cancers that may be treated by alkylating agents, still more particularly glioblastoma. According to a further feature of the present invention, there is provided the use of a compound of the present invention, i.e. a compound of Formula I and any of its embodiments, or any of its subformulae as defined above, or a hydrophenyl compound of Table 2b, or a pharmaceutically acceptable salt or solvate thereof, for inhibiting IRE1 RNase activity, in a patient in need of such treatment, comprising administering to said patient an effective amount of a compound of the present invention, or a pharmaceutically acceptable salt or solvate thereof. In other terms, the invention also provides a method for inhibiting IRE1 RNase activity, in a patient in need of such treatment, which comprises the step of administering to said patient an effective amount of a compound of the present invention, i.e. a compound of Formula I and any of its embodiments, or a hydrophenyl compound of Table 2b, or any of its subformulae as defined above, or a pharmaceutically acceptable salt or solvate thereof. Preferably, the patient is a warm blooded animal, and even more preferably a human. According to the present invention, the compound of the invention or the compound for use according to the invention may be administered as a pharmaceutical formulation in a therapeutically effective amount by any of the accepted modes of administration, preferably by intravenous or oral route. Therapeutically effective amount ranges are typically from 0.1 to 50 000 µg/kg of body weight daily, preferably from 1000 to 40000 µg/kg of body weight daily, depending upon numerous factors such as the severity of the disease to be treated, the age and relative health of the subject, the potency of the compound, the route and the form of administration, the indication towards which the administration is directed, and the preferences and experience of the medical practitioner involved. One of ordinary skill in the art of treating such diseases will be able in reliance upon personal knowledge, to ascertain a therapeutically effective amount of the anticancer agent of the present invention for a given cancer. According to one embodiment, the compounds of the invention, their pharmaceutical acceptable salts or solvates may be administered as part of a combination therapy. Thus, are included within the scope of the present invention embodiments comprising co- administration of, and compositions and medicaments which contain, in addition to a compound of the present invention, a pharmaceutically acceptable salt or solvate thereof as active ingredient, additional therapeutic agents and/or active ingredients. Such multiple drug regimens, often referred to as combination therapy, may be used in the treatment of cancer, particularly those defined above. Thus, the methods of treatment and pharmaceutical compositions of the present invention may employ the compounds of the invention or their pharmaceutical acceptable salts or solvates thereof in the form of monotherapy, but said methods and compositions may also be used in the form of multiple therapy in which one or more compounds of the invention or their pharmaceutically acceptable salts or solvates are co-administered in combination with one or more other therapeutic agents. Such additional therapeutic agents include, but are not limited to, alkylating agents, and preferably temozolomide. In one embodiment, the methods of treatment and pharmaceutical compositions of the present invention may employ the compounds of the present invention, or their pharmaceutical acceptable salts or solvates thereof, in combination with radiation therapy. According to this embodiment, the compounds of the invention, their pharmaceutical acceptable salts or solvates may be administered in combination with radiation therapy. Thus, there is provided a compound of Formula I and any of its embodiments, or any of its subformulae as defined above, or a hydrophenyl compound of Table 2b, for use in the treatment of cancers as defined above in combination with radiation therapy. Such radiation therapies include, but are not limited to, external beam radiation therapy, brachytherapy and systemic radioisotope therapy. The invention also provides a pharmaceutical composition comprising a compound of the present invention, i.e. a compound of Formula I and any of its embodiments, or any of its subformulae as defined above, or a hydrophenyl compound of Table 2b, or a pharmaceutically acceptable salt or solvate thereof, and at least one pharmaceutically acceptable carrier, diluent, excipient and/or adjuvant. As indicated above, the invention also covers pharmaceutical compositions which contain, in addition to a compound of the present invention, a pharmaceutically acceptable salt or solvate thereof as active ingredient, additional therapeutic agents and/or active ingredients, in particular an anticancer agent. The invention also provides a compound of the invention, or a pharmaceutically acceptable salt or solvate thereof, for use in a therapeutic treatment in humans or animals. Another object of this invention is a medicament comprising at least one compound of the invention, or a pharmaceutically acceptable salt or solvate thereof, as active ingredient. Generally, for pharmaceutical use, the compounds of the invention may be formulated as a pharmaceutical preparation comprising at least one compound of the invention and at least one pharmaceutically acceptable carrier, diluent, excipient and/or adjuvant, and optionally one or more further pharmaceutically active compounds. By means of non-limiting examples, such a formulation may be in a form suitable for oral administration, for parenteral administration (such as by intravenous, intramuscular or subcutaneous injection or intravenous infusion), for topical administration (including ocular), cerebral administration, for administration by inhalation, by a skin patch, by an implant, by a suppository, etc. Such suitable administration forms – which may be solid, semi-solid or liquid, depending on the manner of administration – as well as methods and carriers, diluents and excipients for use in the preparation thereof, will be clear to the skilled person; reference is made to the latest edition of Remington’s Pharmaceutical Sciences. For example, the compound of the invention or a pharmaceutical composition comprising a compound of the invention can be administered orally in the form of tablets, coated tablets, pills, capsules, soft gelatin capsules, oral powders, granules, ovules, elixirs, solutions or suspensions, which may contain flavouring or colouring agents, for immediate-, delayed-, modified-, sustained-, pulsed- or controlled-release applications. The tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, a disintegrant such as starch (preferably corn, potato or tapioca starch), sodium starch glycollate, croscarmellose sodium and certain complex silicates, a binder such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxypropylcellulose (HPC), sucrose, gelatin and acacia, a lubricant such as magnesium stearate, stearic acid, glyceryl behenate. Solid compositions of a similar type may also be employed as fillers in hard gelatin capsules. Preferred excipients in this regard include lactose, saccharose, sorbitol, mannitol, potato starch, corn starch, amylopectin, cellulose derivatives or gelatin. Hard gelatin capsules may contain granules of the compound of the invention. Soft gelatin capsules may be prepared with capsules containing the compound of the invention, vegetable oil, waxes, fat, or other suitable vehicle for soft gelatin capsules. As an example, the acceptable vehicle can be an oleaginous vehicle, such as a long chain triglyceride vegetable oil (e.g. corn oil). Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water may contain the active ingredient in a mixture with dispersing agents, wetting agents, and suspending agents and one or more preservatives. Additional excipients, for example sweetening, flavouring and colouring agents, may also be present. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid. Liquid dosage forms for oral administration may include pharmaceutically acceptable, solutions, emulsions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art, such as water or an oleaginous vehicle. Liquid dosage form may be presented as a dry product for constitution with water or other suitable vehicle before use. Such compositions may also comprise adjuvants, such as wetting agents, emulsifying and suspending agents, complexing agents such as 2-hydroxypropyl-beta-cyclodextrin, sulfobutylether-beta-cylodextrin, and sweetening, flavouring, perfuming agents, colouring matter or dyes with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof. These compositions may be preserved by the addition of an anti- oxidant such as butylated hydroxyanisol or alpha-tocopherol. Finely divided powder of the compound of the invention may be prepared for example by micronisation or by processes known in the art. The compound of the invention may be milled using known milling procedures such as wet milling to obtain a particle size appropriate for tablet formation and for other formulation types. If the compound of the present invention is administered parenterally, then examples of such administration include one or more of: intravenously, intraarterially, intraperitoneally, intrathecally, intraventricularly, intraurethrally, intrasternally, intracranially, intramuscularly or subcutaneously administering the agent; and/or by using infusion techniques. The compound of the invention can be administered via the parenteral route with a readily available or a depot-type formulation. The pharmaceutical compositions for the parenteral administration of a readily available formulation may be in the form of a sterile injectable aqueous or oleagenous solution or suspension in a non-toxic parenterally-acceptable diluent or solvent and may contain formulatory agents such as suspending, stabilising dispersing, wetting and/or complexing agents such as cyclodextrin e.g. 2-hydroxypropyl-beta-cyclodextrin, sulfobutylether-beta- cylodextrin. The depot-type formulation for the parenteral administration may be prepared by conventional techniques with pharmaceutically acceptable excipient including without being limited to, biocompatible and biodegradable polymers (e.g. poly(β-caprolactone), poly(ethylene oxide), poly(glycolic acid), poly[(lactic acid)-co-(glycolic acid)...)], poly(lactic acid)...), non-biodegradable polymers (e.g. ethylene vinylacetate copolymer, polyurethane, polyester(amide), polyvinyl chloride...) aqueous and non-aqueous vehicles (e.g. water, sesame oil, cottonseed oil, soybean oil, castor oil, almond oil, oily esters, ethyl alcohol or fractionated vegetable oils, propylene glycol, DMSO, THF, 2-pyrrolidone, N- methylpyrrolidinone, N-vinylpyrrolidinone... ). Alternatively, the active ingredient may be in dry form such as a powder, crystalline or freeze-dried solid for constitution with a suitable vehicle. The preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well known to those skilled in the art. As indicated, the compound of the present invention can be administered intranasally or by inhalation and is conveniently delivered in the form of a dry powder inhaler or an aerosol spray presentation from a pressurised container, pump, spray or nebuliser with the use of a suitable propellant, e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, (for example from Ineos Fluor), carbon dioxide or other suitable gas. In the case of a pressurised aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. The pressurised container, pump, spray or nebuliser may contain a solution or suspension of the active compound. Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated to contain a powder mix of the compound and a suitable powder base such as lactose or starch. For compositions suitable and/or adapted for inhaled administration, it is preferred that the compound or salt of the invention is in a particle-size-reduced form, and more preferably the size-reduced form is obtained or obtainable by micronisation. The preferable particle size of the size-reduced (e.g. micronised) compound or salt or solvate is defined by a D50 value of about 0.5 to about 50 microns (for example as measured using laser diffraction). Alternatively, the compound of the present invention can be administered in the form of a suppository or pessary, or it may be applied topically in the form of a gel, hydrogel, lotion, solution, cream, ointment or dusting powder. The compound of the present invention may also be dermally or transdermally administered, for example, by the use of a skin patch. They may also be administered by the pulmonary or rectal routes. It may also be administered by the ocular route. For ophthalmic use, the compound can be formulated as micronised suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride. Alternatively, it may be formulated in an ointment such as petrolatum. For topical application to the skin, the agent of the present invention can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, it can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water. DEFINITIONS The definitions and explanations below are for the terms as used throughout the entire application, including both the specification and the claims. Unless otherwise stated, any reference to compounds of the invention herein, means the compounds as such as well as their pharmaceutically acceptable salts and solvates. When describing the compounds of the invention, the terms used are to be construed in accordance with the following definitions, unless indicated otherwise. The term “unsubstituted” as used herein means that a radical, a group or a residue carries no substituents. The term “substituted” means that a radical, a group or a residue carries one or more substituents. The term “halo” or “halogen” refers to the atoms of the group 17 of the periodic table (halogens) and includes in particular fluorine, chlorine, bromine and iodine atom. Preferred halo groups in the context of the invention are fluoro and iodo, fluoro being particularly preferred. The term “alkyl” by itself or as part of another substituent refers to a hydrocarbyl group of Formula CnH2n+1 wherein n is a number greater than or equal to 1. Alkyl groups may thus comprise 1 or more carbon atoms and generally, according to this invention comprise from 1 to 12, more preferably from 1 to 8 carbon atoms, and still more preferably from 1 to 6 carbon atoms. Alkyl groups within the meaning of the invention may be linear or branched. Examples of alkyl groups include but are not limited to methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, neopenyl, isopentyl, sec-pentyl, tert-pentyl, n-hexyl, neohexyl, isohexyl, sec-hexyl and tert-hexyl. Particular examples of alkyl groups in the context of the invention include methyl, ethyl, isopropyl and tert-butyl. The term “haloalkyl” alone or in combination, refers to an alkyl group having the meaning as defined above wherein one or more hydrogens are replaced with a halogen as defined above. Non-limiting examples of such haloalkyl groups include chloromethyl, 1- bromoethyl, fluoromethyl, difluoromethyl, trifluoromethyl, 1,1,1-trifluoroethyl and the like. The term “cycloalkyl” as used herein is a monovalent, saturated, or unsaturated monocyclic or bicyclic hydrocarbyl group. Cycloalkyl groups may comprise 3 or more carbon atoms in the ring and generally, according to this invention comprise from 3 to 10, more preferably from 3 to 8 carbon atoms, and still more preferably from 3 to 6 carbon atoms. Examples of cycloalkyl groups include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. The term “heteroatom” as used herein refers to any atom that is not carbon or hydrogen. Non-limiting examples of such heteroatoms include nitrogen, oxygen, sulfur, and phosphorus. Preferred heteroatoms according to the invention are nitrogen, oxygen and sulfur. The terms “heterocyclyl”, “heterocycloalkyl” or “heterocyclo” as used herein by itself or as part of another group refer to non-aromatic, fully saturated or partially unsaturated cyclic groups (for example, 3- to 7-membered monocyclic, 7- to 11-membered bicyclic, or containing a total of 3 to 10 ring atoms) which have at least one heteroatom in at least one carbon atom-containing ring. Each ring of the heterocyclic group containing a heteroatom may have 1, 2, 3 or 4 heteroatoms selected from nitrogen, oxygen and/or sulfur atoms, where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized. The heterocyclic group may be attached at any heteroatom or carbon atom of the ring or ring system, where valence allows. Examples of heterocyclyl groups include but are not limited to aziridinyl, azetidinyl, pyrrolidinyl, piperidinyl, azepanyl, piperazinyl, morpholinyl. The term “aryl” as used herein refers to a polyunsaturated, aromatic hydrocarbyl group having a single ring (e.g. phenyl) or multiple aromatic rings fused together (e.g. naphthyl), typically containing 5 to 12 atoms; preferably 6 to 10, wherein at least one ring is aromatic. Examples of aryl groups include but are not limited to phenyl, biphenyl, 1-naphthyl (or naphthalene-1-yl), 2-naphthyl (or naphthalene-2-yl), anthracenyl, indanyl, indenyl, 1,2,3,4- tetrahydronaphthyl. The term “heteroaryl” as used herein by itself or as part of another group refers but is not limited to 5 to 12 carbon-atom aromatic rings or ring systems containing 1 to 2 rings which are fused together, each ring typically containing 5 to 6 atoms; at least one of which is aromatic, in which one or more carbon atoms in one or more of these rings is replaced by oxygen, nitrogen and/or sulfur atoms where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized. Examples of heteroaryl groups include but are not limited to pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, quinolinyl, quinoxalinyl, quinazolinyl, furanyl, benzofuranyl, pyrrolyl, indolyl, thiophenyl, benzothiophenyl, imidazolyl, benzimidazolyl, pyrazolyl, indazolyl, oxazolyl, benzoxazolyl, isoxazolyl, benzisoxazolyl, thiazolyl, and benzothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, dioxazolyl, dithiazolyl and tetrazolyl. The compounds of the invention containing a basic functional group may be in the form of pharmaceutically acceptable salts. Pharmaceutically acceptable salts of the compounds of the invention containing one or more basic functional groups include in particular the acid addition salts thereof. Suitable acid addition salts are formed from acids which form non- toxic salts. Examples include the acetate, adipate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, cinnamate, citrate, cyclamate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, pyroglutamate, saccharate, stearate, succinate, tannate, tartrate, tosylate, trifluoroacetate and xinofoate salts. Pharmaceutically acceptable salts of compounds of Formula I and subformulae, or a hydrophenyl compounds of Table 2b, may for example be prepared as follows: (i) reacting the compound of Formula I or any of its subformulae, or a hydrophenyl compounds of Table 2b, with the desired acid; or (ii) converting one salt of the compound of Formula I or any of its subformulae, or a hydrophenyl compounds of Table 2b, to another by reaction with an appropriate acid or by means of a suitable ion exchange column. All these reactions are typically carried out in solution. The salt may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent. The degree of ionization in the salt may vary from completely ionized to almost non-ionized. The term “solvate” is used herein to describe a molecular complex comprising the compound of the invention and one or more pharmaceutically acceptable solvent molecules, for example, ethanol. The term “hydrate” is employed when said solvent is water. The compounds of the invention include compounds of the invention as hereinbefore defined, including all polymorphs and crystal habits thereof, prodrugs and isomers thereof (including optical, geometric and tautomeric isomers) and isotopically-labeled compounds of the invention. In addition, although generally, with respect to the salts of the compounds of the invention, pharmaceutically acceptable salts are preferred, it should be noted that the invention in its broadest sense also includes non-pharmaceutically acceptable salts, which may for example be used in the isolation and/or purification of the compounds of the invention. For example, salts formed with optically active acids or bases may be used to form diastereoisomeric salts that can facilitate the separation of optically active isomers of the compounds of the invention. The term “patient” refers to a warm-blooded animal, more preferably a human, who/which is awaiting or receiving medical care or is or will be the object of a medical procedure. The term “human” refers to subjects of both genders and at any stage of development (i.e. neonate, infant, juvenile, adolescent, adult). In one embodiment, the human is an adolescent or adult, preferably an adult. The terms “treat”, “treating” and “treatment”, as used herein, are meant to include alleviating or abrogating a condition or disease and/or its attendant symptoms. The term “therapeutically effective amount” (or more simply an “effective amount”) as used herein means the amount of active agent or active ingredient which is sufficient to achieve the desired therapeutic or prophylactic effect in the individual to which it is administered. The term “administration”, or a variant thereof (e.g., “administering”), means providing the active agent or active ingredient, alone or as part of a pharmaceutically acceptable composition, to the patient in whom/which the condition, symptom, or disease is to be treated. By “pharmaceutically acceptable” is meant that the ingredients of a pharmaceutical composition are compatible with each other and not deleterious to the patient thereof. The term “excipient” as used herein means a substance formulated alongside the active agent or active ingredient in a pharmaceutical composition or medicament. Acceptable excipients for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington’s Pharmaceutical Sciences, 21st Edition 2011. The choice of excipient can be selected with regard to the intended route of administration and standard pharmaceutical practice. The excipient must be acceptable in the sense of being not deleterious to the recipient thereof. The at least one pharmaceutically acceptable excipient may be for example, a binder, a diluent, a carrier, a lubricant, a disintegrator, a wetting agent, a dispersing agent, a suspending agent, and the like. The term “pharmaceutical vehicle” as used herein means a carrier or inert medium used as solvent or diluent in which the pharmaceutically active agent is formulated and/or administered. Non-limiting examples of pharmaceutical vehicles include creams, gels, lotions, solutions, and liposomes. The term “cancer” as used herein refers to the physiological condition in subjects that is characterized by unregulated or dysregulated cell growth with the potential to invade or spread to other parts of the body. The term “cancer” includes solid tumors and blood born tumors, whether malignant or benign. Examples of cancer include, but are not limited to: Acinar adenocarcinoma, acinar carcinoma, acral-lentiginous melanoma, actinic keratosis, adenocarcinoma, adenocystic carcinoma, adenosquamous carcinoma, adnexal carcinoma, adrenal rest tumor, adrenocortical carcinoma, aldosterone secreting carcinoma, alveolar soft part sarcoma, amelanotic melanoma, ameloblastic thyroid carcinoma, angiosarcoma, apocrine carcinoma, Askin’s tumor, astrocytoma, basal cell carcinoma, basaloid carcinoma, basosquamous cell carcinoma, biliary cancer, bone cancer, bone marrow cancer, botryoid sarcoma, brain cancer, breast cancer, bronchioalveolar carcinoma, bronchogenic adenocarcinoma, bronchogenic carcinoma, carcinoma ex pleomorphic adenoma, cervical cancer, chloroma, cholangiocellular carcinoma, chondrosarcoma, choriocarcinoma, choroid plexus carcinoma, clear cell adenocarcinoma, colon cancer, colorectal cancer, comedocarcinoma, cortisol-producing carcinoma, cylindrical cell carcinoma, dedifferentiated liposarcoma, ductal adenocarcinoma of the prostate, ductal carcinoma, ductal carcinoma in situ, duodenal cancer, eccrine carcinoma, embryonal carcinoma, endometrial carcinoma, endometrial stromal carcinoma, epithelioid sarcoma, esophageal cancer, Ewing’s sarcoma, exophytic carcinoma, fibroblastic sarcoma, fibrocarcinoma, fibrolamellar carcinoma, fibrosarcoma, follicular thyroid carcinoma, gallbladder cancer, gastric adenocarcinoma, giant cell carcinoma, giant cell sarcoma, giant cell tumor of bone, glioma, glioblastoma or glioblastoma multiforme, granulose cell carcinoma, head & neck cancer, hemangioma, hemangiosarcoma, hepatoblastoma, hepatocellular carcinoma, Hürthle cell carcinoma, ileal cancer, infiltrating lobular carcinoma, inflammatory carcinoma of the breast, intraductal carcinoma, intraepidermal carcinoma, jejuna cancer, Kaposi’s sarcoma, Krukenberg’s tumor, Kulchitsky cell carcinoma, Kupffer cell sarcoma, large cell carcinoma, larynx cancer, lentigo maligna melanoma, liposarcoma, liver cancer, lobular carcinoma, lobular carcinoma in situ, lung cancer, lymphoepithelioma, lymphoepithelioma, lymphosarcoma, malignant melanoma, medullary carcinoma, medullary thyroid carcinoma, medulloblastoma, meningeal carcinoma, Merkel cell carcinoma, micropapillary carcinoma, mixed cell sarcoma, mucinous carcinoma, mucoepidermoid carcinoma, mucosal melanoma, myxoid liposarcoma, myxosarcoma, nasopharyngeal carcinoma, nephroblastoma, neuroblastoma, nodular melanoma, non-clear cell renal cancer, non-small cell lung cancer, oat cell carcinoma, ocular melanoma, oral cancer, osteoid carcinoma, osteosarcoma, ovarian cancer, Paget’s carcinoma, pancreatic cancer, pancreatoblastoma, papillary adenocarcinoma, papillary carcinoma, papillary thyroid carcinoma, pelvic cancer, periampullary carcinoma, phyllodes tumor, pituitary cancer, pleomorphic liposarcoma, pleuropulmonary blastoma, primary intraosseous carcinoma, prostate cancer, rectal cancer, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, round cell liposarcoma, scar cancer, schistosomal bladder cancer, schneiderian carcinoma, sebaceous carcinoma, signet-ring cell carcinoma, skin cancer, small cell lung cancer, small cell osteosarcoma, soft tissue sarcoma, splindle cell carcinoma, spindle cell sarcoma, squamous cell carcinoma, stomach cancer, superficial spreading melanoma, synovial sarcoma, telangiectatic sarcoma, terminal duct carcinoma, testicular cancer, thyroid cancer, transitional cell carcinoma, tubular carcinoma, tumorigenic melanoma, undifferentiated carcinoma, urachal adenocarcinoma, urinary bladder cancer, uterine cancer, uterine corpus carcinoma, uveal melanoma, aginal cancer, cerrucous carcinoma, villous carcinoma, well-differentiated liposarcoma, Wilm’s tubor or yolk sac tumor. Preferred cancers according to the invention are glioblastoma, triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer, more preferred cancer is glioblastoma. The terms “anticancer agent”, “anticancer drug”, “chemotherapeutic agent” or “cytotoxic agent”, as used herein, refer to a chemical agent used to treat cancer, administered in regimens of one or more cycles, alone or combined with one or more agents over a period of days to weeks. Such agents are toxic to cells with high proliferative rates, such as cancer cells. Examples of anticancer agents include, but are not limited to: - alkylating agents, such as for example cyclophosphamide, mechlorethamine, chlorambucil, melphalan, dacarbazine, dacarbazine, nitrosoureas or temozolomide; - antimetabolites, including antifolates, such as for example methotrexate, pemetrexed, pralatrexate or trimetrexate; pyrimidine analogues, such as for example azacitidine, capecitabine, cytarabine, decitabine, floxurinine, fluorouracil, gemcitabine or trifluridine; and purine analogues such as for example azathioprine, cladribine, fludarabine, mercaptopurine or tioguanine (formerly thioguanine); - anti-microtubule agents, including taxanes, such as for example paclitaxel, docetaxel, abraxane or taxotere; and vinca alkaloids, such as for example vinblastine, vincristine, vindesine or vinorelbine; - cytotoxic antibiotics, including anthracyclines, such as for example daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone or valrubicin; and peptide antibiotics, such as for example bleomycin or actinomycin; - topoisomerase inhibitors, including inhibitors of topoisomerase I, such as for example irinotecan or topotecan; and inhibitors of topoisomerase II, such as for example etoposide, teniposide or tafluposide; - epothilones; - histone deacetylase inhibitors, such as for example vorinostat, romidepsin, panobinostat or quisinostat; - kinase inhibitors, such as for example afatinib, avitinib, bortezomib, carfilzomib, dabrafenib, erlotinib, gefitinib, imatinib, osimertinib, vemurafenib or vismodegib; - platinum-based agents, such as for example carboplatin, cisplatin or oxaliplatin; - proteasome inhibitors, such as for example bortezomib, carfilzomib or ixazomib; - retinoids, such as tretinoin, alitretinoin or bexarotene. Preferred anticancer agents according to the invention are alkylating agents. Preferred anticancer agent according to the invention is temozolomide. The present invention will be better understood with reference to the following examples and figures. These examples are intended to be representative of specific embodiments of the invention, and are not intended as limiting the scope of the invention. FIGURES Figure 1: In vitro inhibition of IRE1 activity by the compounds of the invention. Measure of IRE1 activity in vitro in the presence of increasing concentrations of MKC (A) and compounds 34 (Z4A), 35 (Z4B), 36 (Z4C), 37 (Z4D) and 38 (Z4E) (B-F). FRET signals were measured upon fluorescent probe cleavage over a 25 minutes incubation. The IC50 calculated from the fitting step are shown in the figure. Symbols and error bars represent mean values ± SD. Figure 2: Compound 33 (Z4) and compound 28 (Z4P) inhibit IRE1 activity in cellular models. (A) Protein levels of IRE1 and phospho-IRE1 in U87 cells treated with compound 33 (Z4) and compound 28 (Z4P) over 24 hours. Fold change of protein expression between IRE1 and phospho-IRE1 is represented in bar chart form normalized to untreated U87 cells. (B) Quantification of XBP1s mRNA levels upon treatment with 1 μg/mL Tunicamycin over 24hrs in the presence or absence of pre-treatments with 25 µM compound 33 (Z4) or compound 28 (Z4P) normalized to untreated U87 cells. (C) Quantification of SPARC mRNA levels in the presence of 5μg/mL Actinomycin D and presence or absence of 10 μg/mL Tunicamycin and 9 µg/mL compound 33 (Z4) or compound 28 (Z4P) over 4 hours in U87 cells. (D) Cell viability of U87 cells as function of TMZ concentration, in the presence of increasing concentrations of compound 33 (Z4) and compound 28 (Z4P). The non-linear regression was calculated using the GraphPad Prism software. Symbols and error bars represent mean values ± SD. Figure 3: Compounds of the invention-mediated enhancement of TMZ toxicity in U87 cells. Cell viability assay carried out using U87 cells treated with increasing concentrations of compounds 34 (Z4A), 35 (Z4B), 36 (Z4C), 37 (Z4D), 38 (Z4E) and 28 (Z4P) upon TMZ treatment. The non-linear regression was calculated using the GraphPad Prism software. Symbols and error bars represent mean values ± SD. Figure 4: In vivo effects of compound 28 (Z4P). (A) Weight tracking of animals treated with 3 different concentrations of compound 28 (Z4P) over 7 days. (B) Kaplan-Meier representation of mouse survival under this regiment (black: control; red: compound 28 (Z4P); blue: TMZ; yellow: compound 28 (Z4P) + TMZ). (C) Quantitative analysis of bioluminescence imaging signal over 35 days of experiment for the control (PBS) and compound 28 (Z4P) groups. Symbols and error bars represent mean values ± SEM (number of animals are indicated in the Methods section of Biological evaluation). Figure 5: compound 28 (Z4P) crosses the BBB, delays relapse in vivo under TMZ treatment. (A) Treatment timeline for compound 28 (Z4P) in GB model over 34 days. Period of tumor monitoring is expressed as dotted marks. (B) Quantification of XBP1s mRNA levels from in vivo tumors upon treatment with compound 28 (Z4P) against the control. (C) Treatment timeline for combination of compound 28 (Z4P) (top arrow) and TMZ (shaded area) in GB model over 186 days. Period of tumor monitoring is expressed in as dotted marks. (D) Quantitative analysis of bioluminescence imaging signal over 186 days of experiment for the groups treated with TMZ alone or in combination with Z4P (COMBO). Symbols and error bars represent mean values ± SEM. EXAMPLES ABBREVIATIONS anh.: anhydrous; CAN: acetonitrile; DCC: N,N'-dicyclohexylcarbodiimide; DCM: dichloromethane; DIAD: diisopropyl azodicarboxylate; DiPE: diisopropylethylether; DIPEA: diisopropylethylamine; DMAP: 4-dimethylaminopyridine; DMSO: dimethylsufoxide; DPPA: diphenylphosphoryl azide; equiv.: equivalent; ESI: electrospray ionization; HPLC: high-performance liquid chromatography; HRMS: high-resolution mass spectrometry; MS: mass spectrometry; NMR: nuclear magnetic resonance; PE: petroleum ether; ppm: parts per million; r.t. or rt: room temperature; SCC: Silica-gel column chromatography; TFA: trifluoroacetic acid; THF: tetrahydrofurane; TLC: thin-layer chromatography. SYNTHESIS General information - Commercially available reagents and solvents were purchased from Enamine, Fluorochem, Sigma-Aldrich, Alfa Aesar, Acros and were used as received. All reactions under inert atmosphere were performed under an argon atmosphere. Petroleum ether (PE) refers to petroleum ether with 40−60°C. Thin-layer chromatography (TLC) was performed on TLC silica gel 60 F254 aluminum plates. Compounds were visualized by exposure to UV light (254 nm) or by dipping the plates into potassium permanganate or phosphomolybdic acid solution followed by heating. Silica-gel column chromatography (SCC) was carried out using silica gel (particle size 40−63 µm) with isocratic or step gradient elution as indicated. Automated flash column chromatography was carried out on an Interchim PuriFlash 450 using FlashPure Ecoflex silica cartridges (irregular particle size 50 µm). NMR spectra were acquired on 300/400/500 MHz spectrometers and were referenced to the residual solvent. All chemical shifts are reported in parts per million (ppm). Abbreviations used in the description of resonances are s (singlet), d (doublet), t (triplet), q (quartet), sept (septet), a (apparent), bs (broad singlet), and m (multiplet). Coupling constants (J) are quoted to the nearest 0.1 Hz. HPLC-MS analyses were performed on a Shimadzu Prominence system couple to an Advion ESI mass spectrometer using a Thermoscientific Hypersil Gold aQ column chromatography (5µ, 250 × 4.6 mm) [method: binary gradient, solvent A = H2O + 0.1% TFA, solvent B = MeCN + 0.1% TFA, flow = 1 mL/min, 20 to 100% A/B]. Typical procedure 1 – Synthesis of N-phthalimide derivatives [J. Med. Chem., 2007, 50, 1076] In a dry flask, under argon, equipped with a magnetic stirring bar was added PPh3 (1.5 equiv.) and phthalimide (1.0 equiv.) in dry THF (10 mL / 3 mmol). The mixture was cooled at 0°C and the alcohol derivative (1.0 equiv.) was added. After 10 minutes at 0°C, DIAD (1.5 equiv.) was added dropwise to keep temperature under 5°C. After 15 minutes, the ice bath was removed et the mixture was stirred at room temperature for few hours. The reaction was monitored by 1H NMR. When the alcohol was fully consumed, THF was evaporated under vacuo. The residue was dissolved in Et2O and stocked overnight in a fridge. Triphenylphosphine’s oxide will precipitate. The mixture was filtered, powder was washed with cold Et2O and the resulting solution was evaporated under vacuo. The crude was purified by Column Chromatography on Silica (SCC). Each product was confirmed by 1H NMR compared with library. Typical procedure 2 – Sonogashira’s Reaction [Chem. Commun., 2013, 49, 4157] In a dry flask, under argon, equipped with a magnetic stirring bar: Alkyne derivative (1 equiv.), 4-iodoanisole (1 equiv.), CuI (4% mol) and PdCl2(PPh3)2 (2% mol). Dry acetonitrile (3 mL / 0.5 mmol) was added under argon and the solution was degassed with argon (10 – 15 minutes). After degassing, dry NEt3 (5.0 equiv.) was added and the solution was stirred at room temperature for few hours. The reaction was monitored by TLC. When both of N-phthalimide and 4-iodoanisole were consumed, solvents were evaporated and the crude was triturated in DiPE (Diisopropylether) and filtered to give product with good purity or was purified using SCC. Typical procedure 3: Deprotection of N-phthalimide Derivates using Hydrazine [Org. Lett., 2020, 22, 194-198] In a dry flask, equipped with a magnetic stirring bar: N-phthalimide derivative (1 equiv.) was dissolved in EtOH (18 mL / 2 mmol) and hydrazine hydrate (10 equiv.) was added dropwise to the solution. The resulting mixture was heated to reflux for a minimum of 4 hours. The reaction was monitored by TLC. The formed pasty precipitate was then filtered through a plug of Celite, washed with cold EtOH, evaporated under reduced pressure (T < 35°C). Small amount of DCM was added and a secondary product precipitate. Filtration and the filtrate was concentrated under reduced pressure and purified using SCC, DCM/MeOH to afford desired pure product. Typical procedure 4: Complete reduction of triple bond into simple bond [J. Med. Chem., 2010, 53, 8679] In a dry flask, equipped with a magnetic stirring bar: Alkyne substrate (1 equiv.) was dissolved in EtOH. Then 10% Pd/C was added (1/4 of alkyne substrate’s mass) and the reaction mixture was stirring under hydrogen pressure (1 atm) at room temperature. The reaction was monitored by TLC. The mixture was then filtered through a Celite pad, the catalyst was rinsed three times with EtOH. The resulting filtrate was then evaporated in vacuo to obtain alkane product without purification. Typical procedure 5: Urea formation using 2-isocyanato-1,4-dimethylbenzene In a dry reaction flask was added the amine derivative (1 equiv.), under argon, in dry DCM. 2-isocyanato-1,4-dimethylbenzene (1 equiv.) was dissolved in dry DCM and added dropwise to the reaction tube. The mixture was stirred overnight at room temperature. The formed precipitate was filtered and washed with cold DCM to afford pure desired product in major cases. Typical procedure 5 Bis: Urea formation using triphosgene [Org. Lett., 2004, 6, 3111- 3114] In a dry reaction flask was added an amine derivative (1 equiv.) and NEt3 (2 equiv.), under argon in dry DCM. The mixture was cooled à 0°C and diluted triphosgene (0.5 equiv.) in DCM was added dropwise. The mixture was allowed to return at room temperature and was heated at 40°C for 6h. After completion, the solution was cooled to room temperature and was concentrated under vacuo. Freshly dry DCM was added to the crude and a second amide derivative (1 equiv.) was added. The mixture was stirred overnight, and the reaction was monitored by TLC or 1H NMR. The solvent was removed and crude was purified using SCC, DCM/MeOH to afford pure product. A fraction (10-20 mg) was purified by semi-preparative HPLC to afford sufficient quantity of pure compound for characterization and biological assays Typical procedure 6: Deprotection using BBr3 [Bioorg. Med. Chem. 2016, p. 2318] In a dry flask, equipped with a magnetic stirring bar: Urea derivative (1 equiv.) was dissolved in DCM (2 mL / 0.1 mmol). The solution was cooled at -78°C and BBr3 (1M in DCM – 4.0 equiv.) was added dropwise. The mixture was put under vigorous stirring with slow return to room temperature until complete consumption of the starting material. The reaction was monitored by TLC. The mixture was quenched by adding of ice (# 2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10-20mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound for characterization and biological assays. Typical procedure 7: Acyl azide formation from carboxylic derivative In a dry flask, equipped with a magnetic stirring bar: Carboxylic derivative (1 equiv.) was dissolved in dry toluene (2 mL / 100 mg) and NEt3 (1.05 equiv.) was added at room temperature. After 15 minutes, DPPA (1.05 equiv.) was slowly added. The mixture was stirred until complete consumption of starting material, and monitored by TLC. Water was added and the solution was extracted twice by EtOAc. Combined organic layer was washed with brine, dried by MgSO4 and concentrated under vacuo. The crude material was used without purification or was purified using SCC, Cyclohexane/EtOAc to afford pure product. Typical procedure 8: Urea formation from acyl azide derivative through from Curtius rearrangment In a dry flask, equipped with a magnetic stirring bar: Acyl azide derivative (1 equiv.) was dissolved in dry toluene (2 mL / 100 mg) and was heated at reflux overnight. The mixture was allowed to return at room temperature and 2,5-dimethylaniline (1 equiv.) was added. The mixture was stirred overnight at room temperature. The precipitate was filtered and washed with toluene to afford desired compound or water was added and the mixture was extracted twice by EtOAc. Combined organic layer was washed with brine, dried by MgSO4 and concentrated under vacuo. The crude material was used without purification or was purified using SCC, DCM/MeOH to afford pure product. Typical procedure 9: Ullmann reaction for piperidine [G. Tang et al., Bioorg. Med. Chem. Lett., 2010, 20, 6020-6023] In an oven dry Schlenck, under argon, was added iodine derivative (1 equiv.), piperidine derivative (1.2 equiv.), K2CO3 (2.0 equiv.), CuI (10% mol) and L-proline (20% mol) in dry DMSO. The mixture was heated at 90°C overnight, followed by TLC upon complete consumption of iodine derivative. The solution was allowed to return at room temperature and was partitioned between water and EtOAc. The organic layer was separated and the aqueous layer was extracted with EtOAc. The combined organic layers were washed with brine, dried with MgSO4 and concentrated under vacuo. The crude was purified using column chromatography on silica (from 100% cyclohexane to 80/20 cyclohexane/EtOAc in all cases). Typical procedure 10: BOC deprotection using HCl solution In an oven dry flask under argon, was added carbamate derivates in a solution of HCl in dioxane (4M, 2 mL / 100 mg of carbamate). The mixture was stirred at room temperature for 2 hours until complete consumption of starting material. The solvent was evaporated to give corresponding chlorinated salts of desired products in quantitative yields. Typical procedure 11: Suzuki reaction In an oven dry flask under argon, was added bromide derivative (1 equiv.), aryl boronic acid (1.2 equiv.), 2M aqueous solution of Na2CO3 (1.2 equiv.) and catalytic amount of Pd(PPh3)4 (5% mol) in Toluene:MeOH / 9:1 (6.5 mL / 300 mg of bromide derivative). The reaction mixture was stirred at reflux overnight. The solution was allowed to return at room temperature, was diluted with water, extracted three times with EtOAc. The combined organic layers were washed with brine, dried with MgSO4 and concentrated under vacuo. The crude was purified using column chromatography on silica, Cyclohexane/EtOAc to afford pure product. Typical procedure 12: Reductive amination of 4-(4-hydroxyphenyl)butan-2-one derivatives [Adapted from ACS Chem. Neurosci., 2017, 8, 486-500]
Figure imgf000141_0001
To a 500 mL round bottom flask filled with methanol (150 mL) was added the ketone starting material (28.05 mmol, 1 equiv.) and ammonium acetate (12.97 g, 168.3 mmol, 6 equiv.). The reaction mixture was left to stir at room temperature for 30 min before lowering the temperature to 0°C with an ice bath. Sodium cyanoborohydride (2.64 g, 42.07 mmol, 1.5 equiv.) was then added portion wise. After addition, the ice batch was removed and the reaction left to stir overnight at room temperature. TLC monitoring indicated full consumption of the starting material and the reaction was subsequently quenched at 0°C by addition of HCl 1M (200 mL). After 15 min of stirring, the reaction mixture was concentrated by rotary evaporation to remove most of the methanol and then extracted with diethyl ether (2 × 100 mL). The aqueous layer is isolated and basified by addition of concentrated NaOH until pH ≥ 10, and then sodium chloride is added until saturation, followed by extraction with DCM (3 × 100 mL). The combined organic layers were dried over anh. MgSO4, filtered, evaporated to dryness by rotary-evaporation and the residue dried under high-vacuum to yield the crude product (80 to 85% yield on average). Crude product was usually pure enough to be used without further purification, but if needed, purification can be performed by silica gel column chromatography with gradient elution (100:0-90:10 DCM/MeOH). Typical procedure 13 – Urea synthesis using 1-(3-isocyanatobutyl)-4-methoxybenzene intermediate STEP 1 4-(4-methoxyphenyl)butan-2-amine - To a solution of anhydrous dichloromethane under inert atmosphere with NEt3 (0.282 g, 2.79 mmol, 3 equiv.) was added 4-(4- methoxyphenyl)butan-2-amine (0.5 g, 2.79 mmol, 1 equiv.), the mixture was cooled to 0-5 °C with an ice bath. Triphosgene was added (0.414 g, 1.395 mmol, 0.5 equiv.), and the mixture was allowed to return to room temperature. The reaction was monitored by TLC and stopped upon complete consumption of the amine derivative (~ 5h). The reaction mixture was concentrated in vacuo, then filtered with Et2O (3 × 30 mL), the organic layer was concentrated in vacuo to yield to pure isocyanate product (425 mg, 74 %) with sufficient purity to be used directly in the next step without further purification. 1H NMR (300 MHz, CDCl3, 20°C), δ = 7.08 (dd, J = 3 Hz, CHAr × 2), 6.82 (dd, J = 3 Hz, CHAr × 2), 3.77 (s, O-CH3), 3.53 (m, CH), 2.62 (m, CH2), 1.76 (m, CH2), 1.29 (dd, J = 6 Hz, CH3) ppm. STEP 2
Figure imgf000142_0001
To a solution of anhydrous dichloromethane (6 mL) in an oven dried 25 mL round bottom flask under argon atmosphere and at room temperature was added 1-(3-isocyanatobutyl)-4- methoxybenzene (1 equiv.) and the corresponding aniline derivative (1 equiv.). The reaction mixture was stirred at room temperature overnight, monitored by TLC and stopped upon complete consumption of the aniline derivative. The treatment is specific for each compound: please refer to the protocol of each analogue. Typical procedure 14 – Urea synthesis using isocyanatobenzene derivatives
Figure imgf000143_0001
To a solution of anhydrous dichloromethane (6 mL) in an oven dried 25 mL round bottom flask under argon atmosphere and at room temperature was added 4-(4- methoxyphenyl)butan-2-amine (1 equiv.) and the corresponding isocyanatobenzene derivative (1 equiv.). The reaction mixture was stirred at room temperature, overnight, monitored by TLC and stopped upon complete consumption of the amine derivative. The treatment is specific for each compound: please refer to the protocol of each analogue. Typical procedure 15 – Saponification of ester group.
Figure imgf000143_0002
To a solution of acetyl protected compound (1.0 equiv.) in THF (3 mL / 0.1 mmol) at room temperature was added LiOH (4.0 equiv.) in H2O (1.5 mL / 0.1 mmol). The reaction was stirred at room temperature (Monitored by TLC). The reaction mixture was then quenched with H2O, and the biphasic reaction mixture was extracted three times with EtOAc. The combined organic layers were dried over MgSO4 and concentrated to afford crude product, which was purified by SCC and/or a fraction (10 - 20 mg) of the crude was purified by semi- preparative HPLC / preparative-TLC to afford sufficient quantity of pure compound for characterization and biological assays. Typical procedure 16 – Deprotection of benzyl protecting group.
Figure imgf000143_0003
To a solution of benzyl protected compound (1.0 equiv.) in MeOH (10 mL / 0.2 mmol) under argon, 10% Pd/C was added (1/4 of alkyne substrate’s mass). The flask was evacuated and filled with hydrogen gas by balloon. The mixture was stirred at room temperature for 2 h. The Pd/C was removed by vacuum filtration through Celite and the Celite layer was washed with MeOH (3×10 mL). The filtrate was concentrated to yield to the crude product which was purified by SCC and/or a fraction (10 - 20 mg) of the crude was purified by semi- preparative HPLC / prep-TLC to afford sufficient quantity of pure compound for characterization and biological assays. Scheme 1. General synthetic scheme for Examples 1-3.
Figure imgf000144_0001
Figure imgf000144_0002
1-(2,5-dimethylphenyl)-3-(4-hydroxybenzyl)urea 2,5-dimethylaniline (1.624 mmol, 1.0 equiv., 0.2 mL) and 4-(aminomethyl)phenol as primary amine (1.624 mmol, 1.0 equiv., 200 mg) were reacted according to Typical procedure 5 Bis. Crude was purified with SCC (100% DCM to 95/5 DCM/MeOH) to afford the desired compound 1, 104 mg, yield: 22.8%. A fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound for characterization and biological assays.
Figure imgf000144_0003
1H NMR (300 MHz, DMSO-d6, 20°C), δ = 9.29 (s, 1H), 7.74 – 7.58 (m, 2H), 7.11 (m, 2H), 6.97 (d, 1H), 6.90 (t, 1H), 6.78 – 6.63 (m, 3H), 4.16 (d, 2H), 2.21 (s, 3H), 2.11 (s, 3H) ppm. 13C NMR (75 MHz, DMSO-d6, 20°C), δ = 156.26, 155.28, 138.01, 134.85, 130.24, 129.79, 128.61, 123.45, 122.40, 120.91, 115.05, 42.46, 20.94, 17.46 ppm. HRMS (M+Na)+: - Calculated: 309.0999 - Found: 309.1005. EXAMPLE 2: 1-(2,5-dimethylbenzyl)-3-(4-hydroxybenzyl)urea (2,5-dimethylphenyl)methanamine (1.624 mmol, 1.0 equiv., 0.23 mL) and 4- (aminomethyl)phenol as primary amine (1.624 mmol, 1.0 equiv., 200 mg) were reacted according to Typical procedure 5 Bis. Crude was purified with SCC (100% DCM to 98/2 DCM/MeOH) to afford compound 2, 75 mg, yield: 16.2%. A fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound for characterization and biological assays.
Figure imgf000145_0001
1H NMR (300 MHz, DMSO-d6, 20°C), δ = 9.23 (s, 1H), 7.10 – 7.01 (m, 2H), 7.06 – 6.97 (m, 3H), 6.98 – 6.90 (m, 1H), 6.76 – 6.64 (m, 2H), 6.21 (dt, 2H), 4.16 (d, 2H), 4.11 (d, J = 5.9 Hz, 2H), 2.24 (s, 3H), 2.20 (s, 3H). 13C NMR (75MHz, DMSO-d6, 20°C), δ = 157.97, 156.11, 138.15, 134.45, 132.17, 131.01, 129.82, 128.33, 127.86, 127.11, 114.96, 42.56, 41.04, 30.71, 20.74, 18.09. HRMS (M+Na)+: Calculated: 307.1417 - Found: 307.1416. EXAMPLE 3: 1-(2,5-dimethylbenzyl)-3-(4-hydroxyphenethyl)urea 2,5-dimethylphenyl)methanamine (1.458 mmol, 1.0 equiv., 0.21 mL) and 4-(2- aminoethyl)phenol as primary amine (1.458 mmol, 1.0 equiv., 200 mg) were reacted according to Typical procedure 5 Bis. Crude was purified with SCC (100% DCM to 98/2 DCM/MeOH) to afford compound 3, 107 mg, yield: 24.6%. A fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound for characterization and biological assays.
Figure imgf000146_0001
1H NMR (300 MHz, DMSO-d6, 20°C), δ = 9.16 (s, 1H), 7.05 – 6.90 (m, 6H), 6.72 – 6.64 (m, 2H), 6.19 (t, 1H), 5.82 (t, 1H), 4.13 (d, 2H), 3.24 – 3.14 (m, 2H), 2.57 (t, 2H), 2.24 (s, 3H), 2.20 (s, 3H) ppm. 13C NMR (75 MHz, DMSO-d6, 20°C), δ = 157.83, 155.55, 138.12, 134.39, 132.18, 129.77, 129.70, 129.45, 127.98, 127.08, 115.05, 41.23, 40.96, 35.29, 20.69, 18.04 ppm. HRMS (M+H)+: Calculated: 299.1754 - Found: 299.1756. Scheme 2. General synthetic scheme for Examples 4-5.
Figure imgf000146_0002
EXAMPLE 4: 1-(2,5-dimethylphenyl)-3-(3-(4-hydroxyphenyl)propyl)urea STEP 1 2-(prop-2-yn-1-yl)isoindoline-1,3-dione (A1) - According to Typical procedure 1 using prop-2-yn-1-ol as alcohol (0.107 mol, 1.0 equiv., 6.2 mL). Crude was purified with SCC (100% DCM) to afford: A1, 11.7 g, yield: 59%.
Figure imgf000147_0001
1H NMR (300 MHz, CDCl3, 20°C), δ = 7.94 – 7.82 (m, 2H), 7.80 – 7.68 (m, 2H), 4.45 (d, J = 2.5 Hz, 2H), 2.22 (t, J = 2.5 Hz, 1H) ppm. 13C NMR (75 MHz, CDCl3, 20°C), δ = 167.11, 134.36, 132.13, 123.72, 76.74, 71.62, 27.13 ppm. STEP 2 2-(3-(4-methoxyphenyl)prop-2-yn-1-yl)isoindoline-1,3-dione (A2) - According to Typical procedure 2 using 2-(prop-2-yn-1-yl)isoindoline-1,3-dione (A1) as N-phthalimide derivative (5.40 mmol, 1.0 equiv., 1.0 g). The mixture was stirring overnight. Solvent were removed and crude was triturated with cold EtOAc, filtrated and dried. The resulting powder was sonicated in distilled water, filtrated and dried to afford: A2, 1.2 g, yield: 76.3%. A2 was used in next step without further purification.
Figure imgf000147_0002
1H NMR (400 MHz, DMSO-d6, 20°C), δ = 7.98 – 7.83 (m, 4H), 7.40 – 7.31 (m, 2H), 6.95 – 6.87 (m, 2H), 4.60 (s, 2H), 3.75 (s, 3H) ppm. 13C NMR (101 MHz, DMSO-d6, 20°C), δ = 166.82, 159.52, 134.70, 133.08, 131.49, 123.35, 114.25, 113.51, 82.43, 82.04, 55.21, 27.48 ppm. STEP 3 3-(4-methoxyphenyl)prop-2-yn-1-amine (A3) - According to Typical procedure 3 using 2- (3-(4-methoxyphenyl)prop-2-yn-1-yl)isoindoline-1,3-dione as N-phthalimide derivative (A2) (3.43 mmol, 1.0 equiv., 1.0 g) with Hydrazine Hydrate (34.33 mmol, 10.0 equiv., 1.08 mL). Crude was purified with SCC (100% DCM to 98/2 DCM/MeOH) to afford: A3, 0.45 g, yield: 81%.
Figure imgf000148_0001
1H NMR (400 MHz, DMSO-d6, 20°C), δ = 7.36 – 7.28 (m, 2H, 1, 3), 6.97 – 6.87 (m, 2H, 4, 6), 3.76 (s, 3H, 8), 3.47 (s, 2H, 11) ppm. 13C NMR (101 MHz, DMSO-d6, 20°C), δ = 158.96, 132.59, 114.98, 114.19, 90.47, 81.11, 55.16, 31.32 ppm. STEP 4 3-(4-methoxyphenyl)propan-1-amine (A4) - According to Typical procedure 4 using 3-(4- mmethoxyphenyl)prop-2-yn-1-amine (A3) as alkyne derivative (1.613 mmol, 1.0 equiv., 260 mg). Treatment afford: A4, 244 mg, yield: 91.6%. Crude was used in next step without purification.
Figure imgf000148_0002
1H NMR (400 MHz, CDCl3, 20°C), δ = 7.14 – 7.06 (m, 2H), 6.87 – 6.79 (m, 2H), 3.79 (s, 3H), 2.76 – 2.68 (m, 2H), 2.60 (dd, 2H), 1.74 (dtd, 2H) ppm. 13C NMR (101 MHz, CDCl3, 20°C), δ = 157.90, 134.37, 129.38, 113.93, 55.41, 41.92, 35.81, 32.48 ppm. STEP 5 1-(2,5-dimethylphenyl)-3-(3-(4-methoxyphenyl)propyl)urea (A5) - According to Typical procedure 5 Bis using 2,5-dimethylaniline (0.303 mmol, 1.0 equiv., 37 µL) and 4-(4- methoxyphenyl)but-3-yn-2-amine (A4) as primary amine (0.303 mmol, 1.0 equiv., 50 mg). Evaporation, trituration with DiPE, filtration and crude was purified with SCC (100% DCM to 99/1 DCM/MeOH) to afford: A5, 59 mg, yield: 62.4%.
Figure imgf000149_0001
1H NMR (400 MHz, DMSO-d6, 20°C), δ = 7.66 – 7.61 (m, 1H), 7.50 (s, 1H), 7.17 – 7.09 (m, 2H), 6.98 (d, 1H), 6.89 – 6.81 (m, 2H), 6.71 – 6.64 (m, 1H), 6.53 (t, 1H), 3.72 (s, 3H), 3.07 (q, 2H), 2.59 – 2.51 (m, 2H), 2.21 (s, 3H), 2.12 (s, 3H), 1.69 (p, 2H) ppm. 13C NMR (101 MHz, DMSO-d6, 20°C), δ = 157.39, 155.36, 138.02, 134.85, 133.51, 129.79, 129.17, 123.52, 122.41, 121.05, 113.72, 54.95, 38.60, 31.72, 31.58, 20.94, 17.45 ppm. STEP 6 1-(2,5-dimethylphenyl)-3-(3-(4-hydroxyphenyl)propyl)urea (4) - According to Typical procedure 6 using 1-(2,5-dimethylphenyl)-3-(3-(4-methoxyphenyl)propyl)urea (A5) as methoxy-protected urea (0.08 mmol, 1.0 equiv., 25 mg). The mixture was quenched by adding of ice (# 2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 4 for characterization and biological assays.
Figure imgf000149_0002
1H NMR (300 MHz, Acetone-d6, 20°C), δ = 8.10 (s, 1H, OH 19), 7.74 (d, J = 1.8 Hz,1H , CH 8), 7.15 (s, 1H, NH 9), 7.08 – 6.97 (m, 2H, CH 17), 6.97 (d, J = 7.6 Hz, 1H, CH 4), 6.74 (m, 2H, CH 16), 6.71 (dd, J = 7.6, 1.8 Hz, 1H, CH 3), 6.08 (t, J = 5.8 Hz, 1H, NH 11), 3.22 (td, J = 7.0, 5.8 Hz, 2H, CH212), 2.56 (t, J = 7.0 Hz, 2H, CH214), 2.24 (s, 3H, CH31), 2.15 (s, 3H, CH36), 1.77 (quint., J = 7.0 Hz, 2H, CH213) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ = 156.44 (CO 10), 156.37 (COH 18), 139.06 (C 15), 136.33 (C 5), 133.49 (C 2), 130.73 (CH 4), 130.08 (CH 17), 125.20 (C 7), 123.90 (CH 3), 122.87 (CH 8), 115.97 (CH 16), 40.09 (CH212), 33.26 (CH213), 32.93 (CH214), 21.31 (CH31), 17.68 (CH36) ppm. HRMS (M+Na)+ : Calculated: 321.15735 - Found: 321.1574. MS (ESI+): m/z (%) 299.1 (100) [M+H+]. EXAMPLE 5: Synthesis of 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)-2- methylbutan-2-yl)urea STEP 1 4-(4-methoxyphenyl)-2-methylbut-3-yn-2-amine (B1) - According to Typical procedure 2 using 2-methylbut-3-yn-2-amine as amine derivative (6.015 mmol, 1.0 equiv., 0.5 g, 0.6 mL) and distilled THF as solvent. The mixture was stirring overnight. Solvent were removed and a saturated aqueous solution of NH4Cl was added, extraction with Et2O (3 times). Organic phase was washed with brine and dried with MgSO4. The solution was concentrated in vacuo and crude was purified with SCC (PE/EtOAc/NEt3 - 2/1/0.01) to afford: B1, 0.9 g, yield: 71.9%.
Figure imgf000150_0001
1H NMR (300 MHz, CDCl3, 20°C), δ = 7.37 – 7.26 (m, 2H), 6.86 – 6.75 (m, 2H,), 3.79 (s, 3H), 1.48 (s, 6H) ppm. 13C NMR (75 MHz, CDCl3, 20°C), δ = 159.39, 133.00, 115.66, 113.97, 95.65, 79.91, 55.38, 45.82, 32.07 ppm. STEP 2 4-(4-methoxyphenyl)-2-methylbutan-2-amine (B2) - According to Typical procedure 4 using 4-(4-methoxyphenyl)2-methylbut3-yn-2-amine (B1) as N-phthalimide derivative (0.528 mmol, 1.0 equiv., 100 mg). Crude was used without purification: B2, 96 mg, yield: 94%.
Figure imgf000151_0001
1H NMR (400 MHz, CDCl3, 20°C), δ = 7.15 – 7.07 (m, 2H), 6.87 – 6.78 (m, 2H), 3.78 (s, 3H), 2.64 – 2.55 (m, 2H), 1.69 – 1.60 (m, 2H), 1.16 (s, 6H) ppm. 13C NMR (101 MHz, CDCl3, 20°C), δ = 157.88, 134.97, 129.28, 114.01, 55.43, 49.70, 47.41, 30.54, 30.29 ppm. STEP 3 1-(2,5-dimethylphenyl)-3-(4-(4-methoxyphenyl)-2-methylbutan-2-yl)urea (B3) - According to Typical procedure 5 Bis using 2,5-dimethylaniline (0.259 mmol, 1.0 equiv., 32 µL) and 4-(4-methoxyphenyl)-2-methylbutan-2-amine (B2) as primary amine (0.259 mmol, 1.0 equiv., 50 mg). Evaporation, trituration with DCM, filtration to afford: B3, 59 mg, yield: 67%.
Figure imgf000151_0002
1H NMR (400 MHz, CDCl3, 20°C), δ = 7.72 – 7.66 (m, 1H), 7.43 (s, 1H), 7.14 – 7.05 (m, 2H), 6.97 (d, 1H), 6.87 – 6.78 (m, 2H), 6.65 (ddd, 1H), 6.40 (s, 1H), 3.70 (s, 3H), 2.21 (s, 3H), 2.12 (s, 3H), 1.95 – 1.86 (m, 2H), 1.30 (s, 7H) ppm. 13C NMR (101 MHz, CDCl3, 20°C), δ = 157.26, 154.47, 138.18, 134.83, 134.36, 129.77, 129.03, 123.05, 122.08, 120.62, 113.69, 54.96, 51.83, 42.30, 29.19, 27.32, 20.98, 17.54 ppm. STEP 4 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)-2-methylbutan-2-yl)urea (5) - According to Typical procedure 6 using 1-(2,5-dimethylphenyl)-3-(4-(4-methoxyphenyl)-2- methylbutan-2-yl)urea (B3) as methoxy-protected urea (0.073 mmol, 1.0 equiv., 25 mg). The mixture was quenched by adding of ice (# 2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 5 for characterization and biological assays.
Figure imgf000152_0001
1H NMR (300 MHz, Acetone-d6, 20°C), δ = 8.06 (s, 1H, OH 20), 7.79 (d, J = 1.8 Hz, 1H, CH 8), 7.15 (s, 1H, NH 9), 7.01 (m, 2H, CH 17), 6.96 (d, J = 7.6 Hz, 1H, CH 4), 7.73 (m, 2H, CH 18), 6.69 (dd, J = 7.6, 1.8 Hz, 1H, CH 3), 5.93 (s, 1H, NH 11), 2.55 (m, 2H, CH2 14/15), 2.24 (s, 3H, CH31), 2.15 (s, 3H, CH36), 2.00 (m, 2H, CH214/15), 1.36 (s, 6H, CH3 13) ppm. 13C NMR (101 MHz, CDCl3, 20°C), δ = 156.19 (COH 19), 155.55 (CO 10), 139.19 (C 5), 136.28 (C 2), 134.34 (C 16), 130.68 (CH 4), 130.00 (CH217), 124.80 (C 7), 123.60 (CH 3), 122.52 (CH 8), 115.91 (CH218), 53.15 (C 12), 43.71 (CH214), 30.51 (CH215), 27.92 (CH3 13), 21.35 (CH31), 17.80 (CH36) ppm. HRMS (M+Na)+: Calculated: 349.18865 - Found: 349.1888. HPLC purity @ λ = 254 nm: 95%. MS (ESI+): m/z (%) 327.2 (100) [M+H+]. Scheme 3. General synthetic scheme for Examples 6-8. EXAMPLE 6: 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)but-3-yn-2-yl)urea STEP 1 2-(but-3-yn-2-yl)isoindoline-1,3-dione (C1) - According to Typical procedure 1 using but- 3-yn-2-ol as alcohol (0.107 mol, 1.0 equiv., 8.5 mL). Crude was purified by SCC - Mobile phase: 100% Cyclohexane to Cyclohexane:EtOAc 9:1. Fraction was collected and evaporated to give: crude was purified with SCC (100% cyclohexane to 9/1 cyclohexane:EtOAc) to afford: C1, 13.8 g, yield: 65.7%.
Figure imgf000153_0001
1H NMR (400 MHz, CDCl3, 20°C), δ = 7.85 (dd, 2H), 7.72 (dd, 2H), 5.20 (qd, 1H), 2.34 (d, 1H), 1.70 (d, 3H) ppm. 13C NMR (101 MHz, CDCl3, 20°C), δ = 166.99, 134.25, 131.98, 123.56, 81.22, 71.32, 36.98, 20.17 ppm. STEP 2 2-(4-(4-hydroxyphenyl)but-3-yn-2-yl)isoindoline-1,3-dione (C2) - According to Typical procedure 2 using 2-(but-3-yn-2-yl)isoindoline-1,3-dione (C1) as N-phthalimide derivative (1.255 mmol, 1.0 equiv., 250 mg). The mixture was stirring overnight. The solution was diluted with water and extracted twice with EtOAc, washed with brine and dried with MgSO4. Organic layer was filtered and concentrated under vacuo. Crude was triturated with DiPE and filtered to afford: C2, 350 mg, yield: 95,7%. C2 was used in next step without further purification. 1H NMR (300 MHz, CDCl3, 20°C), δ = 7.86 (dd, 2H), 7.73 (dd, 2H), 7.35 – 7.27 (m, 2H), 6.80 – 6.71 (m, 2H), 5.41 (q, 1H), 1.78 (d, J = 7.1 Hz, 3H) ppm. STEP 3 4-(4-hydroxyphenyl)but-3-yn-2-amine (C3) - According to Typical procedure 3 using 2-(4- (4-hydroxyphenyl)but-3-yn-2-yl)isoindoline-1,3-dione (C2) as N-phthalimide derivative (1.202 mmol, 1.0 equiv., 350 mg) with Hydrazine Hydrate (12.015 mmol, 10.0 equiv., 0.38 mL). Crude was purified with SCC (100% DCM to 98/2 DCM/MeOH) to afford: C3, 61 mg, yield: 31%.
Figure imgf000154_0001
1H NMR (400 MHz, DMSO-d6, 20°C), δ = 7.23 – 7.14 (m, 2H), 6.76 – 6.68 (m, 2H), 3.81 (q, J = 6.7 Hz, 1H), 1.29 (d, J = 6.7 Hz, 3H) ppm. 13C NMR (400 MHz, DMSO-d6, 20°C), δ = 157.41, 132.64, 115.53, 113.02, 92.47, 81.08, 69.78, 38.62, 24.47 ppm. STEP 4 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)but-3-yn-2-yl)urea (6) - According to Typical procedure 5 using 2-isocyanato-1,4-dimethylbenzene (1 equiv.) and 4-(3- aminobut-1-yn-1-yl)phenol (C3) (1 equiv.). Solvent were evaporated, the crude was triturated with DiPE and filtered to afford compound 6. A fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound for characterization and biological assays. 1H NMR (300 MHz, Acetone-d6, 20°C), δ = 8.78 (s, 1H, OH 20), 7.83 – 7.76 (d, J = 2.1 Hz, 1H, CH 8), 7.25 (m, 2H, CH 18), 7.21 (s, 1H, NH 9), 6.98 (d, J = 7.7 Hz, 1H, CH 4), 6.84 – 6.76 (m, 2H, CH 17), 6.77 – 6.68 (dd, J = 7.7, 2.1 Hz, 1H, CH 3), 6.43 (d, J = 8.0 Hz, 1H, NH 11), 4.88 (dq, J = 8.0, 6.8 Hz, 1H, CH 12), 2.29 – 2.19 (s, 3H, CH36), 2.17 (s, 3H, CH3 1), 1.46 (d, J = 6.8 Hz, 3H, CH313) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ = 158.59 (COH 19), 155.24 (CO 10), 138.78 (C 16), 136.40 (C 5), 133.88 (CH 18), 130.76 (CH 4), 124.98 (C 2), 124.00 (CH 3), 122.53 (CH 8), 116.34 (CH 17), 114.69 (C 7), 89.58 (CC 14/15), 82.42 (CC 14/15), 38.70 (CH 12), 23.53 (CH313), 21.32 (CH31), 17.65 (CH36) ppm. HRMS (M+Na)+: Calculated: 331.1417 - Found: 331.1415. HPLC purity @ λ = 254 nm: 96%. MS (ESI+): m/z (%) 309.1 (100) [M+H+]. EXAMPLE 7: 1-(2,5-dimethylphenyl)-3-(3-(4-hydroxyphenyl)prop-2-yn-1-yl)urea STEP 1: 1-(2,5-dimethylphenyl)-3-(prop-2-yn-1-yl)urea (D1) - According to Typical procedure 5 using 2-isocyanato-1,4-dimethylbenzene (2.27 mmol, 1 equiv., 334 mg) and prop-2-yn-1- amine as primary amine (2.27 mmol, 1.0 equiv., 144 µL). Solvent was evaporated and the crude was triturated with DiPE and filtered to afford: D1, 345 mg, yield: 75%.
Figure imgf000155_0001
1H NMR (400 MHz, CDCl3, 20°C), δ = 7.19 – 7.14 (m, 1H), 7.13 (d, 1H), 6.98 (dd, 1H), 5.95 (s, 1H), 4.70 (s, 1H), 4.04 (dd, 2H), 2.32 (s, 3H), 2.24 (s, 3H), 2.21 (t, 1H) ppm. 13C NMR (101 MHz, CDCl3, 20°C), δ = 199.31, 155.95, 137.27, 135.27, 131.17, 127.72, 126.97, 80.47, 77.48, 77.16, 76.84, 71.42, 30.24, 21.06, 17.55 ppm. STEP 2 1-(2,5-dimethylphenyl)-3-(3-(4-hydroxyphenyl)prop-2-yn-1-yl)urea (7) - According to Typical procedure 2 using 1-(2,5-dimethylphenyl)-3-(prop-2-yn-1-yl)urea (D1) (0.494 mmol, 1.0 equiv., 100 mg) as alkyne derivative and 4-iodophenol (0.494 mmol, 1.0 equiv., 109 mg) as iodine derivative. The mixture was stirred overnight. The solution was diluted with water and extracted twice with EtOAc, washed with brine and dried with MgSO4. Organic layer was filtered and concentrated under vacuo. Crude was purified with SCC (100% DCM to 98/2 DCM/MeOH) to afford compound 7, 95 mg. A fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound for characterization and biological assays.
Figure imgf000156_0001
1H NMR (300 MHz, Acetone-d6, 20°C), δ = 8.71 (s, 1H, OH 19), 7.76 (d, J = 2.0 Hz, 1H, CH 8), 7.28 (s, 1H, NH 9), 7.26 (m, 2H, CH 17), 6.99 (d, J = 7.9 Hz, 1H, CH 4), 6.86 – 6.75 (m, 2H, CH 16), 6.78 – 6.68 (m, J = 7.9, 2.0 Hz, 1H, CH 3), 6.34 (t, J = 5.5 Hz, 1H, NH 11), 4.21 (d, J = 5.5 Hz, 2H, CH212), 2.25 (s, 3H, CH31), 2.18 (s, 3H, CH36) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ = 158.58 (COH 18), 155.88 (CO 10), 138.72 (C 15), 136.42 (C 5), 133.91 (CH 17), 130.79 (CH 4), 125.35 (C 2), 124.21 (CH 3), 122.89 (CH 8), 116.35 (CH 16), 114.77 (C 7), 85.60 (CC 13/14), 83.00 (CC 13/14), 30.71 (CH2 12), 21.30 (CH31), 17.65 (CH36) ppm. HPLC purity @ λ = 254 nm: 95%. MS (ESI+): m/z (%) 295.3 (100) [M+H+]. EXAMPLE 8: 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)-2-methylbut-3-yn-2- yl)urea STEP 1 1-(2,5-dimethylphenyl)-3-(2-methylbut-3-yn-2-yl)urea (E1) - According to Typical procedure 5 using 2-isocyanato-1,4-dimethylbenzene (1.205 mmol, 1.0 equiv., 178 mg) and 2-methylbut-3-yn-2-amine as primary amine (1.203 mmol, 1.0 equiv., 119 µL). Solvent were evaporated, the crude was triturated with DiPE and filtered to afford: E1, 208 mg, yield: 75%.
Figure imgf000157_0001
1H NMR (400 MHz, CDCl3, 20°C), δ = 7.40 – 7.35 (m, 1H), 7.08 (d, 1H), 6.93 – 6.86 (m, 1H), 6.42 (s, 1H), 4.74 (s, 1H), 2.43 (s, 1H), 2.31 (s, 3H), 2.23 (s, 3H), 1.63 (s, 6H) ppm. 13C NMR (101 MHz, CDCl3, 20°C), δ = 155.16, 136.92, 136.13, 130.75, 128.00, 126.23, 125.09, 87.51, 70.40, 47.34, 30.10, 21.17, 17.81 ppm. STEP 2 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)-2-methylbut-3-yn-2-yl)urea (8) - According to Typical procedure 2 using 1-(2,5-dimethylphenyl)-3-(2-methylbut-3-yn-2-yl)urea (E1) (0.434 mmol, 1.0 equiv., 100 mg) as alkyne derivative and 4-iodophenol (0.434 mmol, 1.0 equiv., 96 mg) as iodine derivative. The mixture was stirred overnight. The solution was diluted with water and extracted twice with EtOAc, washed with brine and dried with MgSO4. Organic layer was filtered and concentrated under vacuo. Crude was purified with SCC (100% DCM to 98/2 DCM/MeOH) to afford compound 8, 94 mg, yield: 67%. A fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound for characterization and biological assays. 1H NMR (300 MHz, Acetone-d6, 20°C), δ = 8.65 (s, 1H, OH 20), 7.87 – 7.80 (d, J = 2.0 Hz, 1H, CH 8), 7.30 – 7.19 (m, 2H, CH 18), 7.16 (s, 1H, NH 9), 6.97 (d, J = 7.4 Hz, 1H, CH 4), 6.85 – 6.74 (m, 2H, CH 17), 6.75 – 6.65 (dd, J = 7.4, 2.0 Hz, 1H, CH 3), 6.28 (s, 1H, NH 11), 2.25 (s, 3H, CH31), 2.15 (s, 3H, CH36), 1.71 (s, 6H, CH313) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ = 158.42 (COH 19), 155.01 (CO 10), 139.01 (C 16), 136.38 (C 5), 133.89 (CH 18), 130.72 (CH 4), 124.61 (C 2), 123.73 (CH 3), 122.31 (CH 8), 116.29 (CH 17), 115.12 (C 7), 92.89 (CC 14/15), 81.42 (CC 14/15), 48.52 (C 12), 30.19 (CH313), 21.35 (CH31), 17.72 (CH36) ppm. HRMS (M+Na)+: Calculated: 348.16825 - Found: 348.1683. HPLC purity @ λ = 254 nm: 99%. MS (ESI+): m/z (%) 323.0 (100) [M+H+]. EXAMPLE 9: 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)-1H-indol-3-yl)urea
STEP 1 methyl 1-(4-methoxyphenyl)-1H-indole-3-carboxylate (F1)- In an oven dry round bottom flask, under argon, were added methyl 1H-indole-3-carboxylate (1.71 mmol, 1.0 equiv., 300 mg), 4-iodoanisole (2.23 mmol, 1.3 equiv., 521 mg), CuI (10% mol, 33 mg) K3PO4 (3.423 mmol, 2.0 equiv., 693 mg) and trans-1,2-diaminocyclohexane (10% mol, 20 µL) in 1,4- dioxane (4 mL). The mixture was stirred at reflux overnight. After completion of the reaction, the mixture was allowed to return at room temperature. Quenched with water (5 mL) and extracted with EtOAc (2 × 5 mL). The organic layer was filtered through celite and concentrated under vacuo. The obtained residue was triturated with DiPE and filtered to afford: F1, 410 mg, yield: 85%.
Figure imgf000159_0001
1H NMR (400 MHz, MeOD, 20°C), δ = 8.23 – 8.13 (m, 1H), 8.09 (s, 1H), 7.54 – 7.45 (m, 2H), 7.43 (ddt, 1H), 7.35 – 7.24 (m, 2H), 7.21 – 7.13 (m, 2H), 3.95 (s, 3H), 3.92 (s, 3H) ppm. 13C NMR (101 MHz, MeOD, 20°C), δ = 167.27, 160.91, 138.62, 135.91, 132.52, 127.99, 127.47, 124.45, 123.34, 122.42, 116.08, 112.03, 109.18, 56.12, 51.56 ppm. STEP 2 1-(4-methoxyphenyl)-1H-indole-3-carboxylic acid (F2) - To a solution of methyl 1-(4- methoxyphenyl)-1H-indole-3-carboxylate F1 (0.35 mmol, 1.0 equiv., 100 mg) in THF (1 mL) and MeOH (0.5 mL) at room temperature was added 1N aqueous NaOH solution (0.9 mmol, 2.5 equiv., 0.9 mL). The resulting solution was stirred to 50°C overnight. After completion of the reaction, the mixture was allowed to return at room temperature and was acidified using 1N aqueous HCl. The aqueous layer was extracted with EtOAc (3 × 3 mL), and the combined organic layer was dried over anhydrous MgSO4, filtered and solvents were evaporated to afford: F2, 95 mg, yield: <99%.
Figure imgf000160_0001
1H NMR (400 MHz, CDCl3, 20°C), δ = 8.34 – 8.27 (m, 1H), 8.07 (s, 1H), 7.47 – 7.39 (m, 3H), 7.39 – 7.25 (m, 2H), 7.11 – 7.03 (m, 2H), 3.90 (s, 3H) ppm. 13C NMR (101 MHz, CDCl3, 20°C), δ = 169.96, 159.45, 137.55, 135.82, 131.40, 127.05, 126.54, 123.61, 122.79, 122.05, 115.12, 111.21, 107.98, 55.81 ppm. STEP 3 1-(4-methoxyphenyl)-1H-indole-3-carbonyl azide (F3) - In an oven dry flask under argon, was added 1-(4-methoxyphenyl)-1H-indole-3-carboxylic acid F2 (0.37 mmol, 1.0 equiv., 100 mg) in dry toluene (2 mL) at room temperature. Triethylamine (0.39 mmol, 1.05 equiv., 55 µL) was added and the solution was stirred 15 minutes and DPPA (0.39 mmol, 1.05 equiv., 84 µL) was added too. After 5h of stirring at room temperature, the solvent was removed and the mixture was purified on column chromatography in silica to afford: F3, 95 mg, yield: 87%. 1H NMR (300 MHz, CDCl3, 20°C), δ = 8.31 (m, 1H), 7.97 (s, 1H), 7.43 – 7.38 (m, 2H), 7.36 – 7.29 (m, 2H), 7.26 (s, 1H), 7.09 – 7.03 (m, 2H), 3.90 (s, 3H) ppm. 13C NMR (75 MHz, CDCl3, 20°C), δ = 168.29, 159.59, 137.80, 135.75, 131.00, 126.60, 126.51, 124.06, 123.26, 122.01, 115.13, 111.34, 110.21, 55.81 ppm. STEP 4 1-(2,5-dimethylphenyl)-3-(1-(4-methoxyphenyl)-1H-indol-3-yl)urea (F5) - In an oven dry flask under argon, was added the isolated 1-(4-methoxyphenyl)-1H-indole-3-carbonyl azide (F3) (0.36 mmol, 1.0 equiv., 95 mg) was dissolved in toluene (2 mL) and heated at reflux overnight to form the isocyanate compound by Curtius rearrangement F4 (not isolated). The mixture was allowed to return at room temperature and 2,5-dimethylaniline (0.36 mmol, 1.0 equiv., 44 µL) was added and stirred overnight. The mixture was filtered and the white powder was washed with cold toluene to afford: F5, 85 mg, yield: 61%.
Figure imgf000161_0001
1H NMR (300 MHz, DMSO-d6, 20°C), δ = 9.04 (s, 1H), 7.94 (s, 1H), 7.83 – 7.76 (m, 1H), 7.74 (s, 1H), 7.73 – 7.63 (m, 1H), 7.54 – 7.41 (m, 3H), 7.30 – 7.07 (m, 4H), 7.05 (d, 1H), 6.79 – 6.70 (m, 1H), 3.83 (s, 3H), 2.24 (d, 6H) ppm. 13C NMR (75 MHz, DMSO-d6, 20°C), δ = 157.42, 152.64, 137.49, 135.06, 133.09, 132.21, 129.94, 125.04, 123.63, 122.88, 122.75, 121.74, 120.91, 119.23, 117.58, 117.21, 116.31, 114.95, 110.16, 55.41, 20.94, 17.49 ppm. STEP 5 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)-1H-indol-3-yl)urea (9) - According to Typical procedure 6 using 1-(2,5-dimethylphenyl)-3-(1-(4-methoxyphenyl)-1H-indol-3- yl)urea (F5) (65 µmol, 1.0 equiv., 25 mg) in dry DCM (2 mL). The mixture was quenched by adding of ice (# 2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 9 for characterization and biological assays.
Figure imgf000162_0001
1H NMR (500 MHz, Acetone, 20°C), δ = 8.60 (s, 1H, OH 24), 8.55 (s, 1H, NH 9), 7.93 – 7.89 (d, J = 1.5 Hz, 1H, CH 8), 7.84 (s, 1H, NH 11), 7.78 – 7.70 (d, J = 8.2 Hz, 1H, CH 15), 7.65 (s, 1H, CH 13), 7.46 (d, J = 8.2 Hz, 1H, CH 18), 7.44 – 7.38 (m, 2H, CH 21), 7.20 (ddd, J = 8.2, 7.0, 1.1 Hz, 1H, CH 17), 7.10 (ddd, J = 8.2, 7.0, 1.1 Hz, 1H, CH 16), 7.09 – 7.02 (m, 2H, CH 22), 7.04 (d, J = 7.6 Hz 1H, CH 4), 6.77 (dd, J = 7.6, 1.5 Hz 1H, CH 3), 2.30 (s, 3H, CH31), 2.25 (s, 3H, CH36) ppm. 13C NMR (126 MHz, Acetone, 20°C), δ = 209.16 (CO 10), 156.31 (COH 23), 136.00 (C 2), 134.54 (C 19), 131.87 (C 20), 129.88 (CH 4), 125.67 (CH 21), 123.56 (C 5), 123.13 (CH 3), 122.34 (CH 17), 121.64 (CH 8), 119.13 (CH 16), 117.67 (CH 15), 116.24 (CH 22), 110.04 (CH 18), 20.43 (CH31), 16.91 (CH36) ppm. HPLC purity @ λ = 254 nm: 100%. MS (ESI+): m/z (%) 371.8 (100) [M+H+]. EXAMPLE 10: N-(2,5-dimethylphenyl)-4-(4-hydroxyphenyl)piperazine-1- carboxamide rt, overnight 4-(piperazin-1-yl)phenol
Figure imgf000163_0001
N-(2,5-dimethylphenyl)-4-(4-hydroxyphenyl)piperazine-1-carboxamide (10) - According to Typical procedure 5 using 2-isocyanato-1,4-dimethylbenzene (0.281 mmol, 1 equiv., 41 mg) and 4-(piperazin-1-yl)phenol (0.281 mmol, 1.0 equiv., 50 mg) in dry DCM (3 mL). The precipitate was filtered, washed with cold DCM to afford the compound 10, 69 mg. A fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 10 for characterization and biological assays.
Figure imgf000163_0002
1H NMR (300 MHz, Acetone-d6, 20°C), δ = 7.93 (s, 1H, OH 17), 7.35 (s, 1H, NH 9), 7.25 (d, J = 1.9 Hz, 1H, CH 8), 7.03 (d, J = 7.7 Hz, 1H, CH 4), 6.94 – 6.83 (m, 2H, CH 15), 6.87 – 6.77 (dd, J = 7.7, 1.9 Hz, 1H, CH 3), 6.81 – 6.70 (m, 2H, CH 14), 3.70 – 3.61 (m, 4H, CH2 12), 3.09 – 2.98 (m, 4H, CH211), 2.28 – 2.21 (m, 3H, CH31), 2.20 (s, 3H, CH36) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ = 156.34 (CO 10), 152.59 (COH 16), 145.94 (C 13), 138.80 (C 5), 136.04 (C 2), 130.76 (CH 4), 129.35 (C 7), 126.08 (CH 8), 125.63 (CH 3), 119.71 (CH 15), 116.44 (CH 14), 51.83 (CH212), 45.29 (CH211), 21.04 (CH31), 17.81 (CH36) ppm. HRMS (M+Na)+: Calculated: 348.16825 - Found: 348.1683. HPLC purity @ λ = 254 nm: 100%. MS (ESI+): m/z (%) 326.1 (32) [M+H+]. EXAMPLE 11: 1-(2,5-dimethylphenyl)-3-(3'-hydroxy-[1,1'-biphenyl]-4-yl)urea STEP 1 1-(2,5-dimethylphenyl)-3-(3'-methoxy-[1,1'-biphenyl]-4-yl)urea (G1) - According to Typical procedure 5 using 2-isocyanato-1,4-dimethylbenzene (1 mmol, 1.0 equiv., 148 mg) and 4'-methoxy-[1,1'-biphenyl]-4-amine (1 mmol, 1.0 equiv., 200 mg) in dry DCM (6 mL). The solvent was evaporated and the residue was triturated with DiPE and filtered to afford: G1, 357 mg, yield 103%, which was used directly in next step without further purification.
Figure imgf000164_0001
MS (ESI+): m/z (%) 347.0 (100) [M+H+]. STEP 2 1-(2,5-dimethylphenyl)-3-(3'-hydroxy-[1,1'-biphenyl]-4-yl)urea (11) - According to Typical procedure 6 using 1-(2,5-dimethylphenyl)-3-(3'-methoxy-[1,1'-biphenyl]-4-yl)urea G1 (72 µmol, 1.0 equiv., 25 mg) in dry DCM (2 mL). The mixture was quenched by adding of ice (# 2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 11 for characterization and biological assays.
Figure imgf000164_0002
1H NMR (300 MHz, Acetone-d6, 20°C), δ = 8.51 (s, 1H, OH 22), 8.42 (s, 1H, NH 11), 7.82 – 7.75 (m, 1H, CH 7), 7.66 – 7.59 (m, 2H, CH213), 7.57 – 7.51 (m, 2H, CH214), 7.49 (s, 1H, NH 9), 7.24 (t, 1H, dd, 1H, CH 18), 7.09 (d dd, 1H, CH 19), 7.09 (t, 1H, CH 21), 7.04 (d, 1H, CH 3), 6.79 (m, 1H, CH 4), 6.79 (dd, 1H, CH 17), 2.29 (s, 3H, CH36), 2.23 (s, 3H, CH31) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ = 158.77 (CO 10), 153.55 (COH 20), 143.05 (C 16), 140.65 (C 15), 138.27 (C 2), 136.49 (C 5), 135.42 (C 12), 130.88 (CH 3), 130.66 (CH 18), 127.92 (CH2 14), 125.89 (C 8), 124.67 (CH 4/17), 123.27 (CH 7), 119.50 (CH2 13), 118.50 (CH 19/21), 114.64 (CH 19/21), 114.13 (CH 4/17), 21.30 (CH3 6), 17.69 (CH31) ppm. HPLC purity @ λ = 254 nm: 100%. MS (ESI+): m/z (%) 333.0 (100) [M+H+]. EXAMPLE 12: 1-(2,5-dimethylphenyl)-3-(4'-hydroxy-[1,1'-biphenyl]-4-yl)urea STEP 1 1-(2,5-dimethylphenyl)-3-(4'-methoxy-[1,1'-biphenyl]-4-yl)urea (H1) - According to Typical procedure 5 using 2-isocyanato-1,4-dimethylbenzene (1 mmol, 1.0 equiv., 148 mg) and 4'-methoxy-[1,1'-biphenyl]-4-amine (1 mmol, 1.0 equiv., 200 mg) in dry DCM (6 mL). The solvent was evaporated and the residue was triturated with DiPE and filtered to afford: H1, 304 mg, yield 87%.
Figure imgf000165_0001
1H NMR (400 MHz, DMSO-d6, 20°C), δ = 9.05 (s, 1H), 7.86 (s, 1H), 7.69 (s, 1H), 7.61 – 7.48 (m, 6H), 7.05 (d, J = 7.6 Hz, 1H), 7.02 – 6.96 (m, 2H), 6.77 (d, J = 7.6 Hz, 1H), 3.79 (s, 3H), 2.25 (s, 3H), 2.20 (s, 3H) ppm. 13C NMR (101 MHz, DMSO-d6, 20°C), δ = 158.43, 152.60, 138.75, 137.16, 135.07, 133.20, 132.32, 129.98, 127.14, 126.47, 124.37, 123.33, 121.61, 118.36, 114.31, 55.13, 20.92, 17.45 ppm. STEP 2 1-(2,5-dimethylphenyl)-3-(4'-hydroxy-[1,1'-biphenyl]-4-yl)urea (12)- According to Typical procedure 6 using 1-(2,5-dimethylphenyl)-3-(4'-methoxy-[1,1'-biphenyl]-4-yl)urea H1 (72 µmol, 1.0 equiv., 25 mg) in dry DCM (2 mL). The mixture was quenched by adding of ice (# 2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 12 for characterization and biological assays.
Figure imgf000166_0001
1H NMR (300 MHz, Acetone-d6, 20°C), δ = 8.50 (s, 1H, OH 20), 8.39 (s, 1H, NH 9), 7.79 (d, J = 1.2 Hz, 1H, CH 7), 7.65 – 7.55 (m, 2H, CH 17), 7.50 (m, 2H, CH 18), 7.49 (s, 1H, NH 11), 7.46 (m, 2H, CH 14), 7.03 (d, J = 7.7 Hz, 1H, CH 3), 6.96 – 6.85 (m, 2H, CH 13), 6.83 – 6.74 (dd, J = 7.7, 1.2 Hz, 1H, CH 4), 2.29 (s, 3H, CH36), 2.23 (s, 3H, CH31) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ = 157.59 (CO 10), 139.85 (COH 19), 138.38 (C 8), 136.47 (C 5), 135.53 (C 16), 132.99 (C 12), 130.86 (CH 3), 128.32 (CH 14), 127.35 (CH 18), 125.78 (C 2), 124.56 (CH 4), 123.19 (CH 7), 119.59 (CH 17), 116.52 (CH 13), 103.18 (C 15), 21.31 (CH36), 17.73 (CH31) ppm. HPLC purity @ λ = 254 nm: 97.5%. MS (ESI+): m/z (%) 333.0 (100) [M+H+]. EXAMPLE 13: 1-(2,5-dimethylphenyl)-3-(4'-hydroxy-[1,1'-biphenyl]-3-yl)urea STEP 1 1-(2,5-dimethylphenyl)-3-(4'-methoxy-[1,1'-biphenyl]-3-yl)urea (I1) - According to Typical procedure 5 using 2-isocyanato-1,4-dimethylbenzene (1 mmol, 1.0 equiv., 148 mg) and 4'- methoxy-[1,1'-biphenyl]-3-amine (1 mmol, 1.0 equiv., 200 mg) in dry DCM (6 mL). The solvent was evaporated and the residue was triturated with DiPE and filtered to afford: I1, 276 mg, yield 79%.
Figure imgf000167_0001
1H NMR (400 MHz, DMSO-d6, 20°C), δ = 9.08 (s, 1H), 7.87 (s, 1H), 7.77 (m, 1H), 7.70 (s, 1H), 7.56 (d, 2H), 7.33 (m, 2H), 7.20 (m, 1H), 7.04 (m, 2+1H), 6.77 (d, 1H), 3.80 (s, 3H), 2.25 (s, 3H), 2.20 (s, 3H) ppm. 13C NMR (101 MHz, DMSO-d6, 20°C), δ = 158.91, 152.68, 140.49, 140.41, 137.14, 135.06, 132.67, 129.96, 129.30, 127.67, 124.34, 123.32, 121.60, 119.68, 116.40, 115.77, 114.34, 55.16, 20.90, 17.44 ppm. STEP 2 1-(2,5-dimethylphenyl)-3-(4'-hydroxy-[1,1'-biphenyl]-3-yl)urea (13) - According to Typical procedure 6 using 1-(2,5-dimethylphenyl)-3-(4'-methoxy-[1,1'-biphenyl]-3-yl)urea I1 (72 µmol, 1.0 equiv., 25 mg) in dry DCM (2 mL). The mixture was quenched by adding of ice (# 2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 13 for characterization and biological assays.
Figure imgf000167_0002
1H NMR (300 MHz, Acetone-d6, 20°C), δ = 8.48 (s, 1H, OH 22), 8.47 (s, 1H, NH 9), 7.83 (t, J = 2.0 Hz, 1H, CH 17), 7.83 – 7.76 (d, J = 1.4 Hz, 1H, CH 8), 7.54 – 7.46 (m, 2H, CH 19), 7.49 (s, 1H, NH 11), 7.44 (ddd, J = 7.8, 2.0, 1.1 Hz, 1H, CH 13), 7.30 (t, J = 7.8 Hz, 1H, CH 14), 7.20 (ddd, J = 7.8, 2.0, 1.1 Hz, 1H, CH 15), 7.03 (d, J = 7.7 Hz, 1H, CH 4), 6.98 – 6.87 (m, 2H, CH 20), 6.84 – 6.74 (dd, J = 7.7, 1.4 Hz, 1H, CH 3), 2.29 (s, 3H, CH3 1), 2.23 (s, 3H, CH36) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ = 158.08 (CO 10), 153.65 (COH 21), 142.46 (C 12), 141.60 (C 12), 138.31 (C 5), 136.50 (C 2), 133.26 (C 18), 130.87 (CH 4), 129.95 (CH 14), 128.81 (CH 19), 125.80 (C 7), 124.62 (CH 3), 123.23 (CH 8), 120.88 (CH 15), 117.40 (CH 13), 117.26 (CH 17), 116.53 (CH 20), 21.30 (CH31), 17.69 (CH36) ppm. HPLC purity @ λ = 254 nm: 100%. MS (ESI+): m/z (%) 333.0 (100) [M+H+]. EXAMPLE 14: 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)cyclohexyl)urea
Figure imgf000168_0001
STEP 1 4-(4-Methoxyphenyl)cyclohexanone (J1) - In an oven dry flask under argon, was added 4- (4-hydroxyphenyl)cyclohexanone (5.26 mmol, 1.0 equiv., 1 g) with CS2CO3 (7.88 mmol, 1.5 equiv., 2.57 g) in dry acetone (13 mL). Iodomethane (excess, 15.0 equiv.) was added dropwise and the mixture was refluxed for 1h. After completion, monitored using TLC, solvent was removed and water was added (25 mL), the aqueous phase was extracted by DCM (3 x 25 mL). The combined organic layer was dried with Na2SO4, filtered and evaporated under reduced pressure. The solid residue was purified using chromatography to afford: J1, 0.95 g, yield: 88%.
Figure imgf000169_0001
1H NMR (300 MHz, MeOD, 20°C), δ = 7.19 (m, 2H), 6.85 (m, 2H), 3.76 (s, 3H), 3.01 (tt, 1H), 2.61 (td, 2H), 2.39 (ddt, 2H), 2.24 – 2.08 (m, 1H), 2.08 – 1.79 (m, 3H), 1.78 – 1.49 (m, 1H) ppm. STEP 2 4-(4-methoxyphenyl)cyclohexan-1-amine (J2) - According to Typical procedure 2 using 4- (4-Methoxyphenyl)cyclohexanone J1 (1.22 mmol, 1.0 equiv., 250 mg). After 36h of reaction, treatment afford: J2, 210 mg, yield: 83.6%. Amine derivative was used without purification. STEP 3 1-(2,5-dimethylphenyl)-3-(4-(4-methoxyphenyl)cyclohexyl)urea (J3) - According to Typical procedure 5 using 2-isocyanato-1,4-dimethylbenzene (0.925 mmol, 1.0 equiv., 136 mg) and 4-(4-methoxyphenyl)cyclohexan-1-amine J2 (1 mmol, 1.0 equiv., 200 mg) in dry DCM (5 mL). The solvent was evaporated and the residue was triturated washed with cold DCM and filtered to afford: J3, 151 mg, yield: 66%.
Figure imgf000169_0002
1H NMR (300 MHz, DMSO-d6, 20°C), δ = 7.70 (s, 1H), 7.45 (s, 1H), 7.19 – 7.12 (m, 2H), 6.97 (d, 1H), 6.87 – 6.81 (m, 2H), 6.66 (dd, 1H), 6.49 (d, 1H), 3.71 (s, 3H), 3.56 – 3.40 (m, 1H), 2.44 (dt, 1H), 2.21 (s, 3H), 2.12 (s, 3H), 2.00 (dd, 2H), 1.79 (d, 2H), 1.50 (qd, 2H), 1.35 – 1.18 (m, 2H) ppm. 13C NMR (75 MHz, DMSO-d6, 20°C), δ = 157.46, 154.64, 138.80, 138.13, 134.87, 129.82, 127.54, 123.10, 122.20, 120.60, 113.67, 54.97, 48.08, 42.03, 33.49, 33.06, 21.01, 17.49 ppm. STEP 4 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)cyclohexyl)urea (14) - According to Typical procedure 6 using 1-(2,5-dimethylphenyl)-3-(4-(4-methoxyphenyl)cyclohexyl)urea J3 (71 µmol, 1.0 equiv., 25 mg) in dry DCM (2 mL). The mixture was quenched by adding of ice (# 2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 14 for characterization and biological assays.
Figure imgf000170_0001
1H NMR (400 MHz, DMSO-d6, 20°C), δ = 9.10 (s, 1H, OH 20), 7.68 (d, 1H, J ≤ 2 Hz, CH 7), 7.43 (s, 1H, NH 9), 7.02 (m, 2H, CH 17), 6.97 (d, 1H, J = 8.3 Hz, CH 3), 6.66 (m, 2H, CH 18), 6.66 (dd, 1H, J = 8.3, ≤ 2 Hz, CH 4), 6.46 (d, J = 7.6 Hz, 1H, NH 11), 3.45 (tdt, J (approx. value) = 12.0 (eq-eq), 7.6 (NH), 3.5 (eq-ax) Hz, 1H, CH 12), 2.38 (tt, J (approx. value) = 12.0 (eq-eq), 3.5 (eq-ax) Hz, 1H, CH 15), 2.20 (s, 3H, CH36), 2.11 (s, 3H, CH31), 1.98 (dq, J (approx. value) = 12.0 (gem), 3.5 (ax-eq / ax-ax), 2H, CHax 13), 1.77 (dq, J (approx. value) = 12.0 (gem), 3.5 (ax-eq / ax-ax), 2H, CHax 14), 1.47 (tdd, J (approx. value) = 12.0 (eq-eq), 12.0 (gem), 3.5 (eq-ax) Hz, 2H, CHeq 14), 1.25 (tdd, J (approx. value) = 12.0 (eq-eq), 12.0 (gem), 3.5 (eq-ax) Hz, 2H, CHeq 13) ppm. 13C NMR (101 MHz, DMSO-d6, 20°C), δ = 155.39 (CO 10), 154.68 (COH 20), 138.12 (CH 2), 137.04 (CH 16), 134.88 (CH 5), 129.83 (CH 3), 127.43 (CH 17), 123.19 (CH 8), 122.26 (CH 4), 120.69 (CH 7), 114.99 (CH 18), 48.15 (CH 12), 42.06 (CH 15), 33.54 (CH 13), 33.15 (CH 14), 21.01 (CH36), 17.48 (CH31) ppm. HPLC purity @ λ = 254 nm: 100%. MS (ESI+): m/z (%) 338.9 (100) [M+H+]. EXAMPLE 15: 1-(2,5-dimethylphenyl)-3-(1-(3-hydroxyphenyl)piperidin-3-yl)urea STEP 1 tert-butyl (1-(3-methoxyphenyl)piperidin-3-yl)carbamate (K1) - According to Typical procedure 9 using 1-iodo-3-methoxybenzene as iodine derivative (1.068 mmol, 1.0 equiv., 250 mg) and tert-butyl piperidin-3-ylcarbamate as piperidine derivative (1.282 mmol, 1.2 equiv., 257 mg). The crude was purified using column chromatography on silica (from 100% cyclohexane to 80/20 cyclohexane/EtOAc to afford: K1, 194 mg, yield: 49%.
Figure imgf000171_0001
1H NMR (400 MHz, MeOD, 20°C), δ = 7.11 (t, J = 8.2 Hz, 1H), 6.56 (dd, J = 8.2, 2.3 Hz, 1H), 6.50 (d, J = 2.4 Hz, 1H), 6.40 (dd, J = 8.2, 2.3 Hz, 1H), 3.75 (s, 3H), 3.68 – 3.55 (m, 2H), 3.42 (dt, 1H), 2.75 (ddd, 1H), 2.59 (dd,1H), 1.90 (dt, 1H), 1.80 (dp, 1H), 1.67 (dtt, 1H), 1.45 (s, 9+1H) ppm. 13C NMR (101 MHz, MeOD, 20°C), δ = 162.07, 157.80, 154.32, 130.84, 130.70, 110.75, 105.97, 104.30, 80.11, 56.59, 55.55, 51.03, 49.64, 49.43, 49.21, 49.00, 48.79, 48.57, 48.36, 31.44, 28.78, 24.71 ppm. STEP 2 1-(3-methoxyphenyl)piperidin-3-amine dihydrochloride (K2) - According to Typical procedure 10 using tert-butyl (1-(3-methoxyphenyl)piperidin-3-yl)carbamate K1 (0.163 mmol, 1.0 equiv., 50 mg). Evaporation of solvent afford K2 in quantitative yield. 1H NMR (400 MHz, MeOD, 20°C), δ = 7.48 – 7.40 (m, 1H), 7.22 (t, 1H), 7.22 – 7.16 (m, 1H), 7.01 – 6.96 (m, 1H), 3.88 (tt, 1H), 3.86 (s, 3H), 3.81 – 3.71 (m, 1H), 3.59 (td, 3H), 2.20 (m, 3H), 1.87 (m, 1H) ppm. STEP 3 1-(2,5-dimethylphenyl)-3-(1-(3-methoxyphenyl)piperidin-3-yl)urea (K3) - According to Typical procedure 5 using 2-isocyanato-1,4-dimethylbenzene (0.143 mmol, 1.0 equiv., 21 mg) and 1-(3-methoxyphenyl)piperidin-3-amine dihydrochloride K2 (0.143 mmol, 1.0 equiv., 40 mg) in dry DCM (2 mL) following by triethylamine (0.287 mmol, 2.0 equiv., 40 µL). The mixture was stirred at room temperature overnight. The precipitate was filtered, washed with cold DCM to afford: K3, 42 mg, yield: 83%.
Figure imgf000172_0001
1H NMR (300 MHz, DMSO-d6, 20°C), δ = 7.72 (s, 1H), 7.62 (s, 1H), 7.10 (t, J = 8.2 Hz, 1H), 6.97 (d, J = 7.6 Hz, 1H), 6.74 (d, J = 7.5 Hz, 1H), 6.71 – 6.64 (m, 1H), 6.54 (ddd, J = 8.3, 2.4, 0.8 Hz, 1H), 6.46 (t, J = 2.3 Hz, 1H), 6.34 (ddd, J = 8.1, 2.4, 0.8 Hz, 1H), 3.71 (s, 3+1H), 3.49 (dd, 1H), 3.00 (m, 2H), 2.81 (dd, 1H), 2.21 (s, 3H), 2.12 (s, 3H), 1.85 – 1.70 (m, 1H), 1.65 – 1.38 (m, 1H), 1.15 (t, 2H) ppm. 13C NMR (75 MHz, DMSO-d6, 20°C), δ = 160.24, 154.76, 152.45, 138.01, 134.90, 129.84, 123.16, 122.31, 120.66, 108.62, 103.88, 101.92, 54.39, 48.87, 45.65, 45.09, 29.95, 28.03, 22.66, 21.02, 17.55 ppm. STEP 4 1-(2,5-dimethylphenyl)-3-(1-(3-hydroxyphenyl)piperidin-3-yl)urea (15) - According to Typical procedure 6 using 1-(2,5-dimethylphenyl)-3-(1-(3-methoxyphenyl)piperidin-3- yl)urea K3 (71 µmol, 1.0 equiv., 25 mg) in dry DCM (2 mL). The mixture was quenched by adding of ice (# 2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 15 for characterization and biological assays.
Figure imgf000173_0001
1H NMR (300 MHz, Acetone-d6, 20°C), δ = 8.08 (s, 1H, OH 24), 7.84 (d, J = 1.8 Hz, 1H, CH 8), 7.28 (s, 1H, NH 9), 7.03 (d, J = 8.0 Hz, 1H, CH 4), 6.98 (t, J = 8.0 Hz, 1H, CH 20), 6.75 – 6.66 (dd, J = 8.0, 2.2 Hz, 1H, CH 3), 6.44 – 6.39 (ddd, J = 8.0, 2.2, 0.8 Hz, 1H, CH 21), 6.42 (t, J = 0.8 Hz, 1H, CH 23), 6.28 (ddd, J = 8.0, 2.2, 0.8 Hz, 1H, CH 19), 6.20 (d, 1H, NH 11), 3.94 (m, 1H, CH 12), 3.57 – 3.46 (m, 1H, CH piperidine), 3.28 – 3.14 (m, 1H, CH piperidine), 2.99 (m, 1H, CH piperidine), 2.91 – 2.82 (m, 2H, CH piperidine), 2.25 (s, 3H, CH31/6), 2.14 (s, 3H, CH31/6), 1.91 – 1.46 (m, 3H, CH piperidine) ppm. 13C NMR (300 MHz, Acetone-d6, 20°C), δ = 159.12 (COH 22), 155.70 (C 18), 154.20 (CO 10), 139.07 (C 7), 136.34 (C 2), 130.73 (CH 4), 130.48 (CH 20), 124.69 (C 5), 123.72 (CH 3), 122.41 (CH 8), 108.77 (CH 21), 107.21 (CH 19), 104.44 (CH 23), 56.26 (CH2 piperidine), 50.22 (CH2 piperidine), 46.70 (CH 12), 30.92 (CH2 piperidine), 23.77 (CH2 piperidine), 21.36 (CH31), 17.70 (CH36) ppm. HPLC purity @ λ = 254 nm: 100%. MS (ESI+): m/z (%) 340.0 (100) [M+H+]. HRMS (M+Na)+: Calculated: 362.1839 - Found: 362.1840. EXAMPLE 16: 3-(2,5-dimethylphenyl)-1-(4-(4-hydroxyphenyl)butan-2-yl)-1- methylurea (B3-1)
Figure imgf000174_0001
STEP 1 4-(4-(benzyloxy)phenyl)-N-methylbutan-2-amine (L1) - To a 100 mL round bottom flask filled with methanol (30 mL) was added 4-(4-(benzyloxy)phenyl)butan-2-one (500 mg, 1.966 mmol, 1 equiv.) and aqueous methylamine (40 wt. % in water, 680 µL, 7.864 mmol, 4 equiv). After 30 minutes stirring at room temperature, sodium cyanoborohydride (148.2 mg, 2.359 mmol, 1.2 equiv.) was added and stirring was continued for 18h. The reaction was quenched by addition of saturated NaHCO3 (20 mL) and the reaction mixture was extracted with DCM (3 × 20 mL). The organic phase was dried over anh. MgSO4, filtered, and evaporated to dryness by rotary-evaporation. The residue was retaken in EtOAc and extracted with 0.1 M HCl solution (3 × 10 mL). The isolated aqueous acidic phase was then basified by addition of 1 M NaOH until pH ~10 and then extracted with EtOAc (3 × 10 mL). The organic phase was dried over anh. MgSO4, filtered, evaporated to dryness by rotary- evaporation, and dried under high-vacuum to yield 178 mg of crude product L1 with sufficient purity to be used in the next step without further purification. Yield: 34%.
Figure imgf000174_0002
1H NMR (300 MHz, Acetone-d6, 20°C): δ = 7.52 – 7.26 (m, 6H), 7.19 – 7.07 (m, 2H), 6.96 – 6.86 (m, 2H), 5.08 (s, 2H), 2.64 – 2.46 (m, 3H), 2.33 (s, 3H), 1.78 – 1.43 (m, 2H), 1.04 (d, J = 6.3 Hz, 3H) ppm. STEP 2 1-(4-(4-(benzyloxy)phenyl)butan-2-yl)-3-(2,5-dimethylphenyl)-1-methylurea (L2) - To a 10 mL round bottom flask equipped with a condenser, filled with anh. DCM (6 mL) and under argon atmosphere was added 4-(4-(benzyloxy)phenyl)-N-methylbutan-2-amine L1 (178 mg, 0.661 mmol, 1 equiv.) and triethylamine (101.3 µL, 0.727 mmol, 1.1 equiv.). After 20 minutes stirring at room temperature, 4-nitrophenyl (2,5-dimethylphenyl)carbamate (208 mg, 0.727 mmol, 1.1 equiv.) was added and the reaction was heated to 35°C and left stirring for 72 h. The reaction mixture was washed with 0.1 M NaOH (2 × 10 mL) and the organic phase dried over anh. MgSO4, filtered, and evaporated to dryness by rotary-evaporation, then concentrated to dryness and the residue dried under high-vacuum to yield 220 mg of crude. Purification by silica gel column chromatography (25 g Silica) with solid loading and isocratic elution (80:20 PE/Acetone) yielded pure desired product L2, 145 mg, yield: 46%, Rf (PE/Acetone 80:20): 0.265.
Figure imgf000175_0001
1H NMR (300 MHz, Acetone-d6, 20°C): δ = 7.51 – 7.24 (m, 6H), 7.21 – 7.09 (m, 2H), 7.03 (d, J = 7.9 Hz, 2H), 6.97 – 6.87 (m, 2H), 6.87 – 6.74 (m, 1H), 5.06 (s, 2H), 4.57 – 4.40 (m, 1H), 2.54 (t, J = 7.9 Hz, 2H), 2.29 – 2.11 (m, 6H), 1.92 – 1.58 (m, 2H), 1.13 (d, J = 6.7 Hz, 3H) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C) δ = 157.90, 156.76, 139.12, 138.54, 135.93, 135.30, 130.61, 130.08 (2C), 129.19 (2C), 128.76, 128.65, 128.46, 128.29 (2C), 125.60, 125.19, 115.46 (2C), 70.31, 50.01, 37.16, 32.76, 30.60, 27.64, 21.16, 18.65, 17.82 ppm. STEP 3 3-(2,5-dimethylphenyl)-1-(4-(4-hydroxyphenyl)butan-2-yl)-1-methylurea (16) - To a 10 mL round bottom flask filled with MeOH (5 mL) was added 1-(4-(4-(benzyloxy)phenyl)butan- 2-yl)-3-(2,5-dimethylphenyl)-1-methylurea L2 (145 mg, 0.303 mmol, 1 equiv.) and Palladium on charcoal 10 wt.% (22 mg, 15 wt. %). The reaction mixture was then put under H2 atmosphere, left stirring and monitored by TLC. After 3h, TLC showed complete deprotection and the reaction mixture was filtered over a celite pad to remove the Pd/C, rinsed with methanol (10 mL), to afford 122 mg of crude product. Purification by silica gel column chromatography (12g Silica) with solid loading and isocratic elution (85:15 PE/Acetone) yielded pure desired compound 16, 98 mg, yield: 99%, Rf (PE/Acetone 70:30): 0.67.
Figure imgf000176_0001
1H NMR (300 MHz, Acetone-d6, 20°C): δ = 8.09 (s, 1H), 7.37 (d, J = 1.8 Hz, 1H), 7.10 – 6.98 (m, 3H), 6.94 (s, 1H), 6.83 – 6.76 (m, 1H), 6.75 – 6.69 (m, 2H), 4.56 – 4.38 (m, 1H), 2.92 (s, 3H), 2.51 (t, 2H), 2.28 – 2.16 (m, 6H), 1.92 – 1.59 (m, 2H), 1.14 (d, J = 6.7 Hz, 3H) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C) δ = 156.78, 156.35, 139.18, 136.02, 133.77, 130.65, 130.07 (3C), 128.66, 125.53, 125.17, 115.95 (2C), 50.15, 37.38, 32.85, 27.69, 21.12, 18.65, 17.76 ppm. EXAMPLE 17: 2-(2,5-dimethylphenyl)-N-(4-(4-hydroxyphenyl)butan-2-yl)acetamide
Figure imgf000176_0002
STEP 1 2-(2,5-dimethylphenyl)-N-(4-(4-methoxyphenyl)butan-2-yl)acetamide (M1) - To a 25 mL round bottom flask filled with dry DCM (12 mL) and under argon atmosphere was added 2- (2,5-dimethylphenyl)acetic acid (80 mg, 0.487 mmol, 1 equiv.), 1,3- diisopropylcarbodiimide (115 µL, 0.731 mmol, 1.5 equiv.), 1-hydroxybenzotriazole (112 mg, 0.731 mmol, 1.5 equiv.) and DIPEA (127 µL, 0.731 mmol, 1.5 equiv.). After 30 minutes stirring at room temperature, 4-(4-methoxyphenyl)butan-2-amine (105 mg, 0.585 mmol, 1.2 equiv.) was added and stirring was continued for 18h. The reaction mixture was evaporated to dryness by rotary evaporation and the residue dissolved in ethyl acetate (15 mL) and filtered on a frit funnel (por. 4). Filtrate was washed with 0.1M citric acid (15 mL), 0.1M sodium hydroxide and water (15 mL) and the organic layer was dried over anh. MgSO4, filtered, evaporated to dryness by rotary-evaporation and the residue dried under high- vacuum to yield 185 mg of crude. Purification by silica gel column chromatography (12g Silica, ratio 1:65) with solid loading and gradient elution (95:5 to 85:25 PE/EtOAc) yielded pure desired product M1, 124 mg, yield: 78%, Rf (PE/EtOAc 80:20): 0.23.
Figure imgf000177_0001
1H NMR (300 MHz, Acetone-d6, 20°C): δ = 7.10-7.00 (m, 4H), 6.94 (dd, 1H), 6.81 (dt, 2H), 3.92 (sept, 1H), 3.74 (s, 3H), 3.46 (s, 2H), 2.53 (m, 2H), 2.27 (s, 3H), 2.24 (s, 3H), 1.68 (m, 2H) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C) δ = 170.13, 163.08, 162.43, 160.41, 158.83, 135.77, 134.93, 134.53, 131.68, 130.78, 130.06, 128.13, 114.50, 55.38, 45.32, 41.82, 39.66, 32.24, 21.28, 20.96, 19.38 ppm. STEP 2 2-(2,5-dimethylphenyl)-N-(4-(4-hydroxyphenyl)butan-2-yl)acetamide (17) – 2-(2,5- dimethylphenyl)-N-(4-(4-methoxyphenyl)butan-2-yl)acetamide M1 (0.384 mmol, 1.0 equiv., 124 mg) was reacted according to Typical procedure 6. Purification of the crude residue (169 mg) by silica gel flash chromatography (4g column) with liquid deposition and gradient elution 100:0 to 92:8 DCM/MeOH) yielded the pure desired compound 17, 61 mg, yield: 51%, Rf (DCM/MeOH 96:4): 0.27. 1H NMR (300 MHz, Acetone-d6, 20°C): δ = 8.18 (s, 1H), 7.10 – 6.89 (m, 6H), 6.87 (s, 1H), 6.77 – 6.66 (m, 2H), 4.04 – 3.84 (m, 1H), 3.47 (s, 2H), 2.61 – 2.39 (m, 2H), 2.27 (s, 3H), 2.24 (s, 3H), 1.76 – 1.57 (m, 2H), 1.10 (d, J = 6.6 Hz, 3H) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C) δ = 170.13, 158.83, 135.77, 134.93, 134.53, 131.68, 130.78, 130.06, 128.13, 114.50, 55.38, 45.32, 41.82, 39.66, 32.24, 21.28, 20.96, 19.38 ppm. EXAMPLE 18: 2,5-dimethylphenyl (4-(4-hydroxyphenyl)butan-2-yl)carbamate
Figure imgf000178_0001
step 2
Figure imgf000178_0002
step 3
Figure imgf000178_0003
STEP 1 4-nitrophenyl (4-(4-methoxyphenyl)butan-2-yl)carbamate (N1) – To a solution of 4-(4- methoxyphenyl)butan-2-amine (2.568 g, 14.326 mmol, 1 equiv.) and DIPEA (4.99 mL, 28.611 mmol, 2 equiv.) in anh. DCM (60 mL) in a 250 mL round bottom flask under argon atmosphere and maintained at 0 °C with an ice bath was added a solution of 4-nitrophenyl chloroformate (2.887 g, 28.611 mmol, 2 equiv.) in anh. DCM (40 mL). 30 minutes after the end of the addition, the ice bath was removed and the reaction allowed to warm up to room temperature and left stirring for 18h. TLC monitoring indicated complete consumption of the amine and the reaction mixture was transferred into a 500 mL separatory funnel to be washed with saturated NaHCO3 (120 mL) and brine (2 × 100 mL). The organic layer was dried over anh. MgSO4, filtered, evaporated to dryness by rotary-evaporation and the residue dried under high-vacuum to yield 5.13 g of crude as a yellow oil that solidify upon sitting. Purification of the crude residue by silica gel flash chromatography with liquid deposition and gradient elution (50:50 to 0:100 PE/DCM) yielded the pure desired product N1, 2.237 g, yield: 55%, Rf (PE/DCM 25:75): 0.43.
Figure imgf000179_0001
1H NMR (300 MHz, CDCl3, 20°C): δ = 8.30 – 8.18 (dt, 2H), 7.37 – 7.26 (dt, 2H), 7.17 – 7.06 (dt, 2H), 6.89 – 6.78 (dt, 2H), 4.96 (d, J = 8.6 Hz, 1H), 3.92 – 3.72 (m, 1H), 3.78 (s, 3H), 2.66 (t, J = 7.9 Hz, 2H), 1.95 – 1.72 (m, 2H), 1.28 (d, J = 6.6 Hz, 3H) ppm. 13C NMR (75 MHz, Chloroform-d1, 20°C) δ = 158.06, 156.12, 152.52, 144.78, 133.41, 129.33, 125.24, 122.05, 114.06, 77.36, 55.39, 47.64, 38.91, 31.59, 21.22 ppm. STEP 2 2,5-dimethylphenyl (4-(4-methoxyphenyl)butan-2-yl)carbamate (N2) – To a solution of 2,5- dimethylphenol (37.4 mg, 0.305 mmol, 1 equiv.) in anh. THF (4 mL) in a 10 mL round bottom flask under argon and maintained at 0°C with an ice bath was added NaH (0.367 mmol, 1.2 equiv., 60 wt% in oil). The reaction was left stirring at 0°C until gas emission stopped. A solution of 4-nitrophenyl (4-(4-methoxyphenyl)butan-2-yl)carbamate N1 (126 mg, 0.367 mmol, 1.2 equiv.) in anh. THF (3 mL) was then added and the reaction mixture immediately turned bright yellow to orange. The ice bath was then removed and the reaction monitored by TLC, which indicated complete conversion after 3h. The reaction mixture was evaporated to dryness by rotary evaporation and the residue resuspended in DIPE (7 mL), sonicated and filtered on a fritted funnel (por. 4). The filtrate was evaporated to dryness by rotary evaporation to afford 115 mg of crude product N2, used in the next step without further purification. Rf (PE/Acetone 70:30): 0.48. 1H NMR (300 MHz, Acetone-d6, 20°C): δ = 7.16 (d, J = 8.4 Hz, 2H), 7.08 (d, J = 7.7 Hz, 1H), 6.91 (d, J = 7.7 Hz, 1H), 6.88 – 6.81 (m, 3H), 6.68 (d, J = 8.6 Hz, 1H), 3.75 (s, 3H), 2.77 – 2.56 (m, 2H), 2.27 (s, 3H), 2.14 (s, 3H), 1.94 – 1.72 (m, 2H), 1.23 (d, J = 6.6 Hz, 3H) ppm. STEP 3 2,5-dimethylphenyl (4-(4-hydroxyphenyl)butan-2-yl)carbamate (18) - 2,5-dimethylphenyl (4-(4-methoxyphenyl)butan-2-yl)carbamate N2 (115 mg, 0.334 mmol, 1.0 equiv.) was reacted according to Typical procedure 6 and yielded 121 mg of crude product as a yellowish oil. Purification of the crude residue by silica gel flash chromatography (4g FlashPure ID HP Silica column) with liquid deposition and gradient elution (100:0 to 30:70 PE/Acetone) yielded the pure desired compound 18, 32 mg, yield: 31%.
Figure imgf000180_0001
1H NMR (300 MHz, Acetone-d6, 20°C): δ = 8.05 (s, 1H), 7.07 (dd, J = 8.0, 4.6 Hz, 4H), 6.95 – 6.80 (m, 2H), 6.83 – 6.69 (m, 2H), 6.65 (d, J = 8.6 Hz, 1H), 3.72 (h, J = 6.9 Hz, 1H), 2.75 – 2.52 (m, 2H), 2.27 (s, 3H), 2.13 (s, 3H), 1.92 – 1.66 (m, 2H), 1.23 (d, J = 6.6 Hz, 3H) ppm. 13C NMR (75 MHz, Chloroform-d1, 20°C) δ = 156.36, 156.26, 154.70, 150.86, 137.15, 133.62, 131.22, 130.13, 128.34, 126.60, 123.84, 115.98, 115.89, 47.69, 39.77, 32.34, 21.46, 20.81, 15.85, 1.10 ppm. HPLC purity @ λ = 210 nm: 96.5%. MS (ESI+): m/z (%) 314.1 (100) [M+H+]. EXAMPLE 19: 4-(4-hydroxyphenyl)butan-2-yl 2-(2,5-dimethylphenyl)acetate
Figure imgf000181_0001
STEP 1 4-(4-methoxyphenyl)butan-2-yl 2-(2,5-dimethylphenyl)acetate (O1) – To a solution of 2- (2,5-dimethylphenyl)acetic acid (80 mg, 0.487 mmol, 1 equiv.), N,N'- dicyclohexylcarbodiimide (110.6 mg, 0.536 mmol 1.1 equiv.) and 4-Dimethylaminopyridine (9 mg, 0.073 mmol, 0.15 equiv.) in anh. DCM (8 mL) in a 25 mL round bottom flask under argon was added 4-(4-methoxyphenyl)butan-2-ol (96.6 mg, 0.536 mmol, 1.1 equiv.). The reaction was left stirring at room temperature and TLC monitoring indicated completion of the reaction after 4.5h. After completion, N,N'-dicyclohexylurea was filtered off on a fritted funnel (por. 4) and the filtrate washed with water (15 mL), 0.1 M citric acid (15 mL), 0.1M NaOH (15 mL) and water (15 mL). The organic layer was dried over anh. MgSO4, filtered, evaporated to dryness by rotary-evaporation and the residue dried under high-vacuum to yield 206 mg of crude. Purification by silica gel flash chromatography (10.3 g silica, ratio 1:50) with liquid deposition and gradient elution (100:0 to 96:4 PE/EtOAc) yielded the pure desired product O1, 147 mg, yield: 97%, Rf PE/EtOAc 95:5): 0.39.
Figure imgf000181_0002
1H NMR (300 MHz, Acetone-d6, 20°C): δ = 6.91 – 6.71 (m, 5H), 6.66 – 6.54 (m, 2H), 4.70 – 4.53 (m, 1H), 3.53 (s, 3H), 3.39 (d, J = 3.1 Hz, 2H), 2.40 – 2.16 (m, 2H), 2.09 – 1.94 (m, 5H), 1.68 – 1.47 (m, 2H), 0.99 (d, J = 6.3 Hz, 3H) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C) δ = 171.39, 158.91, 135.94, 134.48, 134.35, 134.27, 131.79, 130.87, 130.05, 128.54, 114.55, 71.00, 55.39, 39.91, 38.69, 31.27, 20.90, 20.23, 19.23 ppm. STEP 2 4-(4-hydroxyphenyl)butan-2-yl 2-(2,5-dimethylphenyl)acetate (19) - 4-(4- methoxyphenyl)butan-2-yl 2-(2,5-dimethylphenyl)acetate O1 (147 mg, 0.450 mmol, 1.0 equiv.) was reacted according to Typical procedure 6 and yielded 174 mg of crude product. Purification of the crude residue by silica gel flash chromatography (4g FlashPure ID HP Silica column) with liquid deposition and gradient elution (100:0 to 20:80 PE/DCM) yielded the pure desired compound 19, 23 mg, yield: 16%.
Figure imgf000182_0001
1H NMR (300 MHz, Acetone-d6, 20°C): δ = 7.35 – 7.18 (m, 2H), 7.16 – 6.96 (m, 5H), 4.26 – 4.07 (m, 1H), 3.89 (s, 2H), 2.94 – 2.66 (m, 2H), 2.33 (s, 3H), 2.28 (q, J = 0.7 Hz, 3H), 2.15 – 2.02 (m, 2H), 1.72 (d, J = 6.6 Hz, 3H) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C) δ = 170.62, 150.27, 139.47, 136.12, 134.63, 133.57, 131.91, 130.99, 130.17, 128.84, 122.45, 52.19, 43.48, 39.52, 33.93, 26.78, 20.89, 19.19 ppm. HPLC purity @ λ = 254 nm: 99.4%. EXAMPLE 20: 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)thiourea STEP 1 1-(2,5-dimethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)thiourea (P1) – Due to its toxicity, 2,5-dimethylphenylisothyanate (60 mg, 0.368 mmol, 1 equiv.) was weighted under the fume hood in an oven dry 5 mL round bottom flask, rapidly put under argon atmosphere and to which was added anh. DCM (2 mL). A solution of 4-(4-methoxyphenyl)butan-2- amine (69 g, 0.386 mmol, 1.05 equiv.) in anh. DCM (1 mL) was then added and the reaction left stirring at room temperature. TLC monitoring indicated complete transformation after 1.5h. The reaction mixture was evaporated to dryness by rotary evaporation and the residue P1 (126 mg) used as is in the next step without further purification. 1H NMR (300 MHz, Acetone-d6, 20°C): δ = 8.34 (s, 1H), 7.19 – 6.97 (m, 5H), 6.87 – 6.77 (m, 2H), 4.60 (bs, 1H), 3.74 (s, 3H), 2.60 (m, 2H), 2.28 (s, 3H), 2.23 (s, 3H), 1.89 – 1.63 (m, 2H), 1.17 (d, J = 6.6 Hz, 3H) ppm. STEP 2 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)thiourea (20) - 1-(2,5- dimethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)thiourea P1 (126 mg, 0.368 mmol, 1.0 equiv.) was reacted according to Typical procedure 6 and yielded 160 mg of crude product. Purification of the crude residue by silica gel flash chromatography with solid loading on celite and isocratic elution (75:25 PE/Acetone) yielded the pure compound 20, 87 mg, yield: 72%. 1H NMR (300 MHz, Acetone-d6, 20°C): δ = 8.30 (s, 1H), 8.03 (s, 1H), 7.15 (d, J = 7.6 Hz, 1H), 7.08 – 6.97 (m, 4H), 6.78 – 6.67 (m, 2H), 6.50 (s, 1H), 4.59 (s, 1H), 2.64 – 2.48 (m, 2H), 2.32 – 2.15 (m, 5H), 2.09 (s, 2H), 1.90 – 1.62 (m, 2H), 1.29 (s, 1H), 1.17 (d, J = 6.6 Hz, 3H) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C) δ = 182.05, 156.29, 137.20, 136.98, 133.70, 133.32, 131.66, 130.05 (2C), 129.39, 128.88, 115.93 (2C), 51.42, 39.45, 32.46, 20.83, 20.71, 17.58 ppm. HPLC purity @ λ = 254 nm: 99.3%. MS (ESI+): m/z (%) 329.2 (100) [M+H+]. EXAMPLE 21: 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(o-tolyl)urea
Figure imgf000184_0001
STEP 1 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(o-tolyl)urea (Q1) - According to typical procedure 13 part 2 using 1-(3-isocyanatobutyl)-4-methoxybenzene (80 mg, 0.390 mmol, 1 equiv.) and o-toluidine (41.77 mg, 0.390 mmol, 1 equiv.). The crude product was purified by SCC with solid deposition and gradient elution (98:2 to 95:5 dichloromethane/methanol), yielded the pure desired product Q1: 40 mg, 33%. 1H NMR (300 MHz, CDCl3, 20°C), δ = 7.43 (d, J = 6 Hz, CHAr), 7.18 (t, J = 6 Hz, CHAr × 2), 7.09 (d, J = 9 Hz, CHAr), 7.05 (d, J = 9 Hz, CHAr × 2), 6.79 (dd, J = 9 Hz, CHAr × 2), 6.37 (s, NH), 3.91 (q, J = 6 Hz, CH), 3.75 (s, CH3), 2.57 (t, J = 6 Hz, CH2), 2.24 (s, CH3), 1.66 (m, CH2), 1.13 (d, J = 6 Hz, CH3) ppm. STEP 2 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(o-tolyl)urea (21) - According to typical procedure 6 using 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(o-tolyl)urea Q1 (40 mg, 0.128 mmol, 1 equiv.) and BBr3 (192.46 mg, 0.768 mmol, 6 equiv.). The crude product was purified by SCC with solid deposition and gradient elution (95:5 to 8:2 petroleum ether/acetone), yielded the pure desired compound 21: 32 mg, 32%. 1H NMR (300 MHz, Acetone-d6, 20°C), δ = 8.09 (s, OH), 7.93 (d, NH), 7.2 (s, CHAr), 7.09 (t, CHAr × 2), 7.01 (dd, CHAr × 2), 6.85 (t, CHAr), 6.73 (dd, CHAr × 2), 5.96 (d, J = 9 Hz, NH), 3.87 (m, CH), 2.59 (m, CH2), 2.2 (s, CH3), 1.7 (m, CH2), 1.17 (d, J = 6 Hz, CH3) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ = 156.33, 155.88, 139.42, 133.74, 130.87, 130.05, 127.83, 127.02, 122.97, 121.83, 115.95, 45.99, 40.34, 32.30, 21.84, 18.10 ppm. EXAMPLE 22: 1-(3,5-dimethyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea
Figure imgf000185_0001
STEP 1 1-(3,5-dimethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea (R1) - According to typical procedure 13 part 2 using 4-(4-methoxyphenyl)butan-2-amine (80 mg, 0,446 mmol, 1 equiv.) and isocyanato-3,5-dimethylbenzene (59.7 mg, 0.446 mmol, 1 equiv.). The crude product was purified by SCC (95:5 to 8:2 petroleum ether/acetone), yielded the pure desired product R1: 123 mg, 88%. 1H NMR (300 MHz, CDCl3, 20°C), δ = 7.22 (s, NH), 7.02 (d, J = 9 Hz, CHAr × 2), 6.90 (s, CHAr × 2), 6.77 (d, J = 9 Hz, CHAr × 2), 6.63 (s, CHAr), 5.29 (s, NH), 3.89 (m, CH), 3.74 (s, CH3), 2.56 (m, CH2), 2.20 (s, CH3), 1.65 (m, CH2), 1.13 (d, J = 6 Hz, CH3) ppm. STEP 2 1-(3,5-dimethyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea (22) - According to typical procedure 6 using 1-(3,5-dimethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea R1 (123 mg, 0.377 mmol, 1 equiv.) and BBr3 (566.36 mg, 2.261 mmol, 6 equiv.). The crude product was purified by SCC (99:1 to 95:5 dichloromethane/methanol), yielded the pure desired compound 22: 98 mg, 52%. 1H NMR (300 MHz, CDCl3, 20°C), δ = 7.28 (s, NH), 6.88 (d, J = 9 Hz, CHAr × 2), 6.78 (m, CHAr × 2), 6.76 (m, CHAr × 3), 6.44 (s, NH), 3.91 (m, J = 6 Hz, CH), 2.58 (t, J = 6 Hz, CH2), 2.29 (s, CH3 × 2), 1.44 (m, CH2), 1.16 (d, J = 6 Hz, CH3) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ = 156.33, 155.83, 141.51, 138.71, 133.72, 130.04, 123.83, 116.80, 115.96, 45.95, 40.32, 32.30, 21.83, 21.50 ppm. HPLC purity @ λ = 254 nm: 100 %. MS (ESI+): m/z (%) 313.0 (100) [M+H+]. EXAMPLE 23: 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(3-isopropylphenyl)urea
STEP 1 1-(4-(4-(benzyloxy)phenyl)butan-2-yl)-3-(3-isopropylphenyl)urea (S1) - According to Typical procedure 13 part 2 using 1-(benzyloxy)-4-(3-isocyantobutyl)benzene (95 mg, 0.336 mmol, 1 equiv.) and 3-isopropylaniline (50 mg, 0.370 mmol, 1.1 equiv.) were reacted. The crude product was purified by SCC with solid deposition (95:5 to 85:15 petroleum ether/ethyl acetate), yielded the pure desired product S1: 47 mg, 34%. 1H NMR (300 MHz, Acetone–d6, 20°C), δ = 7.80 (s, NH), 7.47 (m, CHAr × 2), 7.38 (m, CHAr × 3), 7.32 (m, CHAr × 2), 7.13 (m, CHAr × 3), 6.91 (m, CHAr × 2), 6.81 (m, CHAr) 5.07 (s, CH2), 3.88 (m, CH), 2.83 (m, CH), 2.63 (m, CH2), 1.72 (m, CH2), 1.20 (d, J = 6 Hz, CH3) ppm. STEP 2 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(3-isopropylphenyl)urea (23) - A mixture of 1-(4-(4- (benzyloxy)phenyl)butan-2-yl)-3-(3-isopropylphenyl)urea S1 (47 mg, 0.113 mmol, 1 equiv.) and Pd/C (10 wt.%) was placed in MeOH (10 mL), the flask was evacuated and filled with hydrogen gas by balloon. The mixture was stirred at room temperature for 2 h. The Pd/C was removed by vacuum filtration through celite and the celite layer was washed with MeOH (10 mL × 3). The mixture was concentrated to yield to the crude product which was purified by SCC with solid deposition and gradient elution (95:5 to 8:2 petroleum ether/acetone), yielded the pure desired compound 23: 32 mg, 87%. 1H NMR (300 MHz, Acetone–d6, 20°C), δ = 8.17 (s, OH), 8.03 (s, NH), 7.38 (m, CHAr), 7.32 (m, CHAr), 7.14 (t, J = 9 Hz, CHAr), 7.02 (d, J = 9 Hz, CHAr × 2), 6.80 (d, J = 6 Hz, CHAr), 6.74 (dd, J = 3 Hz, CHAr × 2), 5.88 (d, J = 9 Hz, NH), 3.87 (m, CH), 2.85 (m, CH), 2.58 (m, CH2), 1.71 (m, CH2), 1.20 (d, J = 6 Hz, CH3), 1.17 (d, J = 6 Hz, CH3) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ = 155.40, 154.81, 149.18, 140.94, 132.90, 129.29, 129.17, 128.44, 119.17, 115.99, 115.55, 115.04, 113.76, 45.02, 44.91, 39.46, 34.04, 31.42, 23.42, 20.97 ppm. EXAMPLE 24: 1-(benzo[d]thiazol-6-yl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea
Figure imgf000188_0001
Figure imgf000188_0002
Figure imgf000188_0003
Figure imgf000188_0004
Figure imgf000188_0005
STEP 1 6-isocyanatobenzo[d]thiazole (T1) - To a solution of anhydrous dichloromethane (6 mL) under inert atmosphere with NEt3 (161.69 mg, 1.598 mmol, 3 equiv.) was added the benzo[d]thiazol-6-amine (80 mg, 0.533 mmol, 1 equiv.), the mixture was cooled to 0-5°C with an ice bath. Triphosgene was added (79 mg, 0.266 mmol, 0.5 equiv.), and the mixture was allowed to return at room temperature. The reaction was monitored by TLC and stopped upon complete consumption of the amine derivative (overnight). The reaction mixture was concentrated in vacuo, then filtered with Et2O (3 × 15 mL). The organic layer was concentrated in vacuo to yield to pure isocyanate product T1 (62 mg, 66%) with sufficient purity to be used directly in step 3 without further purification. STEP 2 4-(3-aminobutyl)phenyl acetate (T2) – Synthesized according to typical procedure: Reductive amination of 4-(4-hydroxyphenyl)butan-2-one derivatives – To a solution of anhydrous dichloromethane (8 mL) and TFA (trifluoroacetic acid – 8 mL) was added acetic anhydride (810 mg, 7.93 mmol, 4 equiv.) and 4-(4-hydroxyphenyl)butan-2-aminium chloride (400 mg, 1.98 mmol, 1 equiv.) at room temperature. The resulting mixture was stirred for 4h, monitored by TLC and stopped upon complete consumption, then the solution was concentrated by rotary evaporation to afford crude product, which was purified by SCC (95:5 to 50/50 dichloromethane/methanol), yielded the pure desired compound T2: 388 mg, 80%. 1H NMR (300 MHz, CDCl3, 20°C), δ = 7.78 (s, NH3+), 7.09 (d, J = 9 Hz, CHAr × 2), 6.92 (d, J = 9 Hz, CHAr × 2), 3.14 (m, CH), 2.57 (m, J = 9 Hz, CH2), 2.23 (s, CH3), 1.92 (bs, CH2), 1.70 (bs, CH2), 1.21 (d, J = 6 Hz, CH3) ppm. STEP 3 4-(3-(3-benzo[d]thiazol-6-yl)ureido)butyl)phenyl acetate (T3) - To a solution of tetrahydrofuran (6 mL) in an oven dried 25 mL round bottom flask under argon atmosphere and at room temperature was added 6-isocyanatobenzo[d]thiazole T1 (62 mg, 0.352 mmol, 1 equiv.), NEt3 (42.7 mg, 0.422 mmol, 1.2 equiv.) and 4-(3-aminobutyl)phenyl acetate T2 (80 mg, 0.387 mmol, 1.1 equiv.). The reaction mixture was stirred at room temperature, overnight, monitored by TLC and stopped upon complete consumption of the amine derivative. The resulting mixture was evaporated by rotary evaporation and the crude product was purified by SCC (97:3 dichloromethane/methanol), yielded the pure desired product T3: 55 mg, 40%. 1H NMR (300 MHz, Acetone–d6, 20°C), δ = 9.15 (s, CH), 8.40 (d, J = 3 Hz, CHAr), 8.11 (s, NH), 7.91 (d, J = 9 Hz, CHAr), 7.54 (dd, J = 3 Hz, CHAr), 7.24 (d, J = 9 Hz, CHAr × 2), 7.00 (d, J = 9 Hz, CHAr × 2), 5.81 (d, J = 9 Hz, NH), 3.95 (m, CH), 2.71 (m, CH2), 2.23 (s, CH3), 1.78 (m, CH2), 1.20 (d, J = 9 Hz, CH3) ppm. STEP 4 1-(benzo[d]thiazol-6-yl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea (24) - To a solution of 4- (3-(3-benzo[d]thiazol-6-yl)ureido)butyl)phenyl acetate T3 (55 mg, 0.143 mmol, 1 equiv.) in tetrahydrofuran (6 mL) at room temperature was added LiOH (13.74 mg, 0.574, 4 equiv.) in H2O (2 mL). The reaction was stirred for 24h. The reaction mixture was then quenched with H2O (4 mL), and the biphasic reaction mixture was extracted with ethyl acetate (3 × 10 mL). The combined organic layers were dried over MgSO4 and concentrated to afford crude product, which was purified by SCC (99:1 to 95:5 dichloromethane/methanol), yielded the pure desired compound 24: 31 mg, 63%. 1H NMR (300 MHz, Acetone–d6, 20°C), δ = 9.16 (s, CHAr), 8.40 (s, OH), 8.16 (s, NH), 8.13 (s, CHAr), 7.89 (d, J = 6 Hz, CHAr), 7.54 (dd, J = 3 Hz, CHAr), 7.02 (d, J = 9 Hz, CHAr x 2), 6.73 (d, J = 9 Hz, CHAr x 2), 5.81 (d, J = 9 Hz, NH), 3.91 (s, J = 6 Hz, CH), 2.60 (m, CH2), 1.72 (m, CH2), 1.18 (d, J = 6 Hz, CH3) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ = 156.33, 155.86, 155.74, 155.42, 140.65, 133.75, 130.08, 127.10, 122.51, 118.42, 115.97, 112.59, 46.05, 40.29, 32.31, 21.82 ppm. HPLC purity @ λ = 254 nm: 98 %. MS (ESI+): m/z (%) 341.9 (100) [M+H+]. EXAMPLE 25: 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(quinolin-7-yl)urea U1
Figure imgf000191_0001
Figure imgf000191_0002
U3 25 STEP 1 7-isocyanatoquinoline (U1) - To a solution of anhydrous dichloromethane (6 mL) under inert atmosphere with NEt3 (309.9 mg, 0.613 mmol, 5 equiv.) was added the quinolin-7-amine (133 mg, 0.613 mmol, 1 equiv.), the mixture was cooled to 0-5°C with an ice bath. Triphosgene was added (181.8 mg, 0.613 mmol, 1 equiv.), and the mixture was allowed to return at room temperature. The reaction was monitored by TLC upon complete consumption of the amine derivative (overnight). The reaction mixture was concentrated in vacuo, then filtered with Et2O (3 × 20 mL). The organic layer was concentrated in vacuo to yield to pure isocyanate product U1 (80 mg, 68%) with sufficient purity to be used directly in step 3 without further purification. STEP 2 4-(3-aminobutyl)phenyl acetate (U2) - Synthesized according to typical procedure : Reductive amination of 4-(4-hydroxyphenyl)butan-2-one derivatives – To a solution of anhydrous dichloromethane (8 mL) and TFA (trifluoroacetic acid – 8 mL) was added acetic anhydride (810 mg, 7.93 mmol, 4 equiv.) and 4-(4-hydroxyphenyl)butan-2-aminium chloride (400 mg, 1.98 mmol, 1 equiv.) at room temperature. The resulting mixture was stirred for 4h, monitored by TLC upon complete consumption, then the solution was concentrated by rotary evaporation to afford crude product, which was purified by SCC (95:5 to 50/50 dichloromethane/methanol), yielded the pure desired product U2: 388 mg, 80%. 1H NMR (300 MHz, CDCl3, 20°C), δ = 7.78 (s, NH3+), 7.09 (d, J = 9 Hz, CHAr × 2), 6.92 (d, J = 9 Hz, CHAr × 2), 3.14 (m, CH), 2.57 (m, J = 9 Hz, CH2), 2.23 (s, CH3), 1.92 (bs, CH2), 1.70 (bs, CH2), 1.21 (d, J = 6 Hz, CH3) ppm. STEP 3 4-(3-(3-(quinolin-7-yl)ureido)butyl)phenyl acetate (U3) - To a solution of tetrahydrofuran (6 mL) in an oven dried 25 mL round bottom flask under argon atmosphere and at room temperature was added 7-isocyanatoquinoline U1 (80 mg, 0.47 mmol, 1 equiv.) NEt3 (95.14 mg, 0.94 mmol, 2 equiv.) and 4-(3-aminobutyl)phenyl acetate U2 (97.44 mg, 0.47 mmol, 1 equiv.). The reaction mixture was stirred at room temperature, overnight, monitored by TLC upon complete consumption of the amine derivative. The resulting mixture was evaporated by rotary evaporation and the crude product was purified by SCC (95:5 to 7:3 dichloromethane/acetone), yielded the pure desired product U3: 23 mg, 13%. 1H NMR (300 MHz, Acetone–d6, 20°C), δ = 7.94 (m, CHAr), 7.39 (m, CHAr × 2), 7.22 (m, CHAr × 3), 7.17 (m, CHAr), 6.99 (d, J = 9 Hz, CHAr × 2), 6.85 (dd, J = 3 Hz, CHAr), 5.63 (d, J = 6 Hz, NH), 3.92 (m, CH), 2.70 (m, CH2), 2.23 (s, CH3), 1.76 (m, CH2), 1.20 (d, J = 9 Hz, CH3) ppm. STEP 4 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(quinolin-7-yl)urea (25) - To a solution of 4-(3-(3- (quinolin-7-yl)ureido)butyl)phenyl acetate U3 (23 mg, 0.061 mmol, 1 equiv.) in tetrahydrofuran (6 mL) at room temperature was added LiOH (5.838 mg, 0.244 mmol, 4 equiv.) in H2O (2 mL). The reaction was stirred for 24 h. The reaction mixture was then quenched with H2O (4 mL), and the biphasic reaction mixture was extracted with ethyl acetate (3 × 10 mL). The combined organic layers were dried over MgSO4 and concentrated to afford crude product, which was purified by SCC (9:1 to 7:3 petroleum ether/acetone), yielded the pure desired compound 25: 18 mg, 88%. 1H NMR (300 MHz, Acetone–d6, 20°C), δ = 8.15 (s, NH), 8.09 (s, CHAr), 7.31 (dd, J = 3 Hz, CHAr), 7.02 (d, J = 6 Hz, CHAr × 2), 6.73 (m, CHAr × 3), 6.67 (d, J = 6 Hz, NH), 5.90 (s, CH2), 3.82 (m, CH), 2.58 (m, CH2), 1.70 (m, CH2), 1.14 (d, J = 6 Hz, CH3) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ = 156.39, 155.53, 142.88, 136.17, 133.77, 130.13, 129.02, 121.28, 120.88, 116.01, 115.01, 46.11, 40.29, 32.35, 21.83 ppm. HPLC purity @ λ = 254 nm: 99 %. MS (ESI+): m/z (%) 335.9 (100) [M+H+]. EXAMPLE 26: 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(2-methylnaphtalen-1-yl)urea
Figure imgf000193_0001
STEP 1 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(2-methylnaphtalen-1-yl)urea (V1) - According to typical procedure 14 using 1-(3-isocyanatobutyl)-4-methoxybenzene (50 mg, 0.244 mmol, 1 equiv.) and 2-methylnaphtalen-1-amine (38.2 mg, 0.244 mmol, 1 equiv.). The crude product was purified by SCC with solid deposition (7:3 petroleum ether/acetone), yielded the pure desired product V1: 40 mg, 45%. 1H NMR (300 MHz, CDCl3, 20°C), δ = 8.08 (d, J = 9 Hz, CHAr), 7.82 (m, CHAr × 2), 7.51 (m, CHAr × 2), 7.41 (d, J = 6 Hz, CHAr), 6.96 (d, J = 9 Hz, CHAr × 2), 7.76 (d, J = 6 Hz, CHAr × 2), 6.06 (s, NH), 3.99 (m, CH), 3,76 (s, CH3), 2.50 (s, CH3), 2.45 (m, CH2), 1.52 (m, CH2), 1.03 (d, J = 9 Hz, CH3) ppm. STEP 2 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(2-methylnaphtalen-1-yl)urea (26) - According to typical procedure 6 using 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(2-methylnaphtalen-1- yl)urea (40 mg, 0.110 mmol, 1 equiv.) and BBr3 (110.58 mg, 0.441 mmol, 4 equiv.). The crude product was purified by SCC with solid deposition and gradient elution (99:1 to 95:5 dichloromethane/methanol), yielded the pure desired compound 26: 37 mg, 96%. 1H NMR (300 MHz, Acetone-d6, 20°C), δ = 8.24 (s, NH), 8.04 (d, J = 9 Hz, NH), 7.80 (d, J = 6 Hz, CHAr), 7.60 (t, J = 6 Hz, CHAr × 2), 7.42 (m, CHAr x 2), 7.29 (d, J = 9 Hz, CHAr), 6.99 (d, J = 9 Hz, CHAr × 2), 6.73 (d, J = 9 Hz, CHAr × 2), 5.88 (s, OH), 3.87 (m, CH), 2.55 (m, J = 6 Hz, CH2), 2.40 (s, CH3), 1.64 (m, CH2), 1.11 (d, J = 6 Hz, CH3) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ = 156.95, 156.30, 133.92, 133.04, 130.10, 129.74, 128.68, 126.87, 125.85, 124.00, 115.94, 46.18, 40.39, 32.39, 22.00, 18.80 ppm. HPLC purity @ λ = 254 nm: 100%. MS (ESI+): m/z (%) 348.9 (100) [M+H+]. EXAMPLE 27: 1-(2-(tert-butyl)phenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea
Figure imgf000194_0001
STEP 1 1-(4-(4-(benzyloxy)phenyl)butan-2-yl)-3-(2-(tert-butyl)phenyl)urea (W1) - According to typical procedure 14 using 1-(benzyloxy)-4-(3-isocyanatobutyl)benzene (70 mg, 0.249 mmol, 1 equiv.) and 2-(tert-butyl)aniline (40.84 mg, 0.273 mmol, 1.1 equiv.). The crude product was purified by SCC with solid deposition (8:2 petroleum ether/acetone), yielded the pure desired product W1: 88 mg, 82%. 1H NMR (300 MHz, Acetone-d6, 20°C), δ = 7.47 (m, CHAr × 2), 7.37 (m, CHAr × 5), 7.12 (m, CHAr × 4), 6.92 (m, CHAr × 2), 6.83 (m, CHAr), 5.82 (d, J = 9 Hz, NH), 5.07 (s, CH2), 3.89 (m, CH), 2.63 (m, CH2), 1.71 (m, CH2), 1.41 (s, CH3 × 3), 1.14 (d, J = 6 Hz, CH3) ppm. STEP 2 1-(2-(tert-butyl)phenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea (27) - A mixture of 1-(4-(4- (benzyloxy)phenyl)butan-2-yl)-3-(2-(tert-butyl)phenyl)urea W1 (88 mg, 0.204 mmol, 1 equiv.) and Pd/C (10 wt%) was placed in MeOH (10 mL), the flask was evacuated and filled with hydrogen gas by balloon. The mixture was stirred at room temperature for 2 h. The Pd/C was removed by vacuum filtration through celite and the celite layer was washed with MeOH (10 mL × 3). The mixture was concentrated to yield to the crude product which was purified by SCC with solid deposition and gradient elution (99:1 to 95:5 dichloromethane/methanol), yielded the pure desired compound 27: 65 mg, 93%. 1H NMR (300 MHz, CDCl3, 20°C), δ = 7.6 (s, NH), 7.16 (m, CHAr × 2), 7.07 (m, CHAr), 6.88 (d, J = 9 Hz, CHAr × 2), 6.70 (d, J = 9 Hz, CHAr × 2), 6.06 (s, NH), 3.88 (m, CH), 2.44 (m, J = 6 Hz, CH2), 1.56 (m, CH2), 1.32 (s, CH3 × 3), 1.02 (d, J = 6 Hz, CH3) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ = 155.94, 155.46, 144.17, 137.15, 132.90, 129.89, 129.15, 126.23, 125.14, 115.08, 45.29, 39.51, 34.52, 31.47, 30.18, 21.11 ppm. HPLC purity @ λ = 254 nm: 100 %. MS (ESI+): m/z (%) 341.0 (100) [M+H+]. EXAMPLE 28: 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea 4-(4-methoxyphenyl)butan-2-amine (W1) – To a 250 mL round bottom flask filled with methanol (150 mL) was added 4-(4-methoxyphenyl)butan-2-one (5 g, 28.05 mmol, 1 equiv.) and ammonium acetate (12.97 g, 168.3 mmol, 6 equiv.) and the reaction mixture was left to stir at room temperature for 30 min. Temperature is then lowered to 0°C with an ice bath and sodium cyanoborohydride (2.64 g, 42.07 mmol, 1.5 equiv.) was added portion wise. The ice batch was then removed and the reaction left to stir overnight at room temperature. TLC monitoring indicated full consumption of the starting material and the reaction was subsequently quenched at 0°C by addition of HCl 1M (200 mL). After 15 min of stirring, the reaction mixture was concentrated by rotary evaporation to remove most of the methanol and then extracted with diethyl ether (2 × 100 mL). The aqueous layer is isolated and basified by addition of concentrated NaOH until pH ≥ 10, and then sodium chloride is added until saturation, followed by extraction with DCM (3 × 100 mL). The combined organic layers were dried over anh. MgSO4, filtered, evaporated to dryness by rotary-evaporation and the residue dried under high-vacuum to yield 4.2 g of crude product W1 (83% yield, 80 to 85% yield on average). Crude product W1 was pure enough to be used without further purification, but if needed, purification can be performed by silica gel column chromatography with gradient elution (100:0-90:10 DCM/MeOH).
Figure imgf000196_0001
1H NMR (300.16 MHz, CDCl3, 20°C): δ = 7.10 (dt, 2H, CH 7,8); 6.82 (dt, 2H, CH 9,10); 3.78 (s, 3H, CH312); 2.90 (h, 1H, CH 2); 2.60 (qdd, 2H, CH25); 1.62 (m, 2H, CH24); 1.10 (d, 3H, CH31) ppm. 13C NMR (75.47 MHz, CDCl3, 20°C): δ = 157.86 (C 11); 134.51 (C 6); 129.32 (CH 9,10); 113.93 (CH 7,8); 55.40 (CH312); 46.65 (CH 2); 42.24 (CH24); 32.05 (CH25); 24.21 (CH3 1) ppm. 4-nitrophenyl (2,5-dimethylphenyl)carbamate (W2) – To a 250 mL round bottom flask filled with dry EtOAc (100 mL) and under argon atmosphere was added successively the 4- nitrophenyl chloroformate (2.0 g, 9.9 mmol, 1.1 equiv.) potassium carbonate (1.37 g, 9.9 mmol, 1.1 equiv.) and 2,5-dimethylaniline (1.12 mL, 9.0 mmol, 1 equiv.). The reaction is left stirring at room temperature and followed by TLC. After 2h, the reaction mixture is diluted with EtOAc (100 mL), transferred in a separatory funnel and washed successively with 1M citric acid (100 mL), sat. sodium carbonate (100 mL), and brine (100 mL). The organic layer is dried over anh. MgSO4, filtered, evaporated to dryness by rotary-evaporation and the residue dried under high-vacuum to afford 1.9 g of W2 (74% yield) as a pale beige solid, used without further purification.
Figure imgf000197_0001
1H NMR (300.16 MHz, CDCl3, 20°C): δ = 8.29 (dt, 2H, CH 14,15); 7.64 (bs, 1H, NH 9); 7.41 (dt, 2H, CH 12,13); 7.11 (d, 1H, CH 3); 6.93 (d, 1H, CH 4); 6.78 (bs, 1H, CH 7); 2.32 (d, 6H, CH31,6) ppm. 1-(2,5-dimethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea (W3) – To a 25 mL round bottom flask filled with dry DCM (12 mL) and under argon atmosphere was added 4- nitrophenyl (2,5-dimethylphenyl)carbamate W2 (639 mg, 2.231 mmol, 1 equiv.) and triethylamine (311 µL, 2.231 mmol, 1 equiv.), followed by the 4-(4-methoxyphenyl)butan- 2-amine W1 (400 mg, 2.231 mmol, 1 equiv.). The reaction was left stirring at room temperature and TLC monitoring showed full conversion after 1h. The reaction mixture was then concentrated to dryness by rotary-evaporation and the solid residue triturated and sonicated in diisopropyl ether (10 mL), then filtered. The solid was collected and resuspended in cold water (10 mL) to be sonicated, followed by filtration (repeat twice). Finally, the solid was collected and dried under high-vacuum to afford 564 mg of W3 (77% yield) as a pale white solid, used without further purification.
Figure imgf000198_0001
1H NMR (300.16 MHz, DMSO-d6, 20°C): δ = 7.81 (s, 1H, NH 9); 7.67 (s, 1H, CH 8); 7.12 (dt, 2H, CH 17,18); 6.95 (d, 2H, CH 3,4); 6.83 (dt, 2H, CH 19, 20); 6.64 (d, 1H, NH 11); 3.70 (s, 3H, CH322); 3.55 (m, 1H, CH 12); 2.56 (m, 2H, CH215); 2.20 (s, 3H, CH31); 2.14 (s, 3H, CH36); 1.64 (sept, 2H, CH214); 1.08 (d, 3H, CH313) ppm. HPLC purity @ λ = 210 nm: 98.9%. MS (ESI+): m/z (%) 327.3 (100) [M+H+]. 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea (28, Z4P) – To a 100 mL round bottom flask filled with dry DCM (30 mL), under argon atmosphere was added 1- (2,5-dimethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea W3 (300 mg, 0.92 mmol, 1 equiv.). After cooling to 0°C, boron tribromide (1M in DCM, 4.6 mL, 5 equiv.) was added slowly over the course of a few minutes. The reaction mixture was left stirring for 1h at 0°C, and then warmed to room temperature and left stirring overnight. TLC monitoring showed full consumption of the starting material and the reaction was quenched by addition of ice (20 g) and stirred for 30 minutes before being transferred to a separatory funnel. Layers were separated and the aqueous layer extracted with DCM (2 × 40 mL) and EtOAc (2 × 40 mL). The combined organic layers were dried over anh. MgSO4, filtered, evaporated to dryness by rotary-evaporation and the residue dried under high-vacuum to yield 258 mg of crude.50 mg of crude product was purified by silica gel column chromatography (3 g Silica, ratio 1:60) with solid loading and isocratic elution (75:25 PE/Acetone) to afford 47 mg of compound 28 (Z4P) as a white powder.
Figure imgf000199_0001
1H NMR (300.16 MHz, Acetone-d6, 20°C): δ = 8.10 (bs, 1H, OH 22); 7.80 (s, 1H, NH 9); 7.09 (s, 1H, CH 8); 7.03 (dt, 2H, CH 17,18); 6.97 (d, 1H, CH 4); 6.74 (dt, 2H, CH 19,20); 6.69 (d, 1H, CH 3); 5.92 (d, 1H, NH 11); 3.87 (sept, 1H, CH 12); 2.60 (m, 2H, CH215); 2.24 (s, 3H, CH31); 2.15 (s, 3H, CH36); 1.71 (m, 2H, CH214); 1.16 (d, 3H, CH313) ppm. 13C NMR (75.47 MHz, (CD3)2CO, 20°C): δ = 156.3 (C 21); 155.9 (CO 10); 139.2 (C 16); 136.3 (C 2); 133.8 (C 7); 130.7 (CH 4); 130.1 (CH 17,18); 124.8 (C 5); 123.7 (CH 3); 122.5 (C 8); 115.92 (CH 19,20); 46.0 (CH 12); 40.4 (CH214); 32.3 (CH215); 21.8 (CH313); 21.3 (CH31); 17.7 (CH36) ppm. HPLC purity @ λ = 254 nm: 100%. MS (ESI+): m/z (%) 313.2 (100) [M+H+]. Chiral HPLC separation - The preparative chiral HPLC separation was performed on an Agilent 1260 Infinity unit (pump G1311C, autosampler G1329B, DAD G1365D and fraction collector G1364C), and monitored by Agilent OpenLAB CDS Chemstation LC. Samples were dissolved in a mixture of ethanol and hexane (50/50) and eluted on a Chiralpak ID (250 × 10 mm) column, with hexane / ethanol / dichloromethane (80/10/10) as mobile phase, flow-rate = 5 mL/min, UV detection at 254 nm, yielding two enantiomers (RTA = 7.28 min and RTB = 9.12 min) with ee > 99.5%. Optical rotary power measurements - Absolute stereochemistry of enantiomers A and B were determined by measuring their optical rotary power and compared with a compound 28 enantiomer of known stereochemistry, Z4P-R (in-house synthesis). Measurements were performed on Perkin Elmer polarimeter 341 with sodium lamp (589 nm) and 1-dm path at 20°C using 0.5 mg/mL solutions of enantiomer A, enantiomer B and Z4P-R in acetonitrile. Results are the average of 3 measurements (1s integration time). Enantiomer A: average optical rotation -0.023°, [α]D = –46; Enantiomer B: average optical rotation +0.021°, [α]D = +42; Z4P-R: average optical rotation +0.023°, [α]D = +46. Enantiomer A = compound S-28 & Enantiomer B = compound R-28.
Figure imgf000200_0001
compound S-28 compound R-28 EXAMPLE 29: 1-(2,5-dimethylphenyl)-3-(4-phenylbutan-2-yl)urea
Figure imgf000200_0002
1-(2,5-dimethylphenyl)-3-(4-phenylbutan-2-yl)urea (29) - To a 5 mL round bottom flask filled with dry DCM (2 mL) and under argon atmosphere was added 4-nitrophenyl (2,5- dimethylphenyl)carbamate (96 mg, 0.335 mmol, 1 equiv.) and triethylamine (47 µL, 0.335 mmol, 1 equiv.), followed by the 4-phenylbutan-2-amine (50 mg, 0.335 mmol, 1 equiv.). The reaction was left stirring at room temperature and TLC monitoring showed full conversion after 1h. The reaction mixture was then concentrated to dryness by rotary- evaporation and the solid residue triturated and sonicated in diisopropyl ether (8 mL), filtered and rinced with diisopropyl ether (4 mL). The solid was collected and resuspended in cold water (4 mL) to be sonicated, followed by filtration and rincing with cold water (2 × 2 mL). Finally, the solid was collected and dried under high-vacuum to afford compound 29 with excellent purity, 84 mg, 85% yield. HPLC purity @ λ = 254 nm: 97.4%. MS (ESI+): m/z (%) 297.2 (100) [M+H+]. 1H NMR (300 MHz, Acetone-d6, 20°C), δ (ppm) = 7.81 (d, J = 1.8 Hz, 1H), 7.32 – 7.11 (m, 5H), 7.09 (s, 1H), 6.97 (d, J = 7.6 Hz, 1H), 6.70 (dd, J = 7.5, 1.8 Hz, 1H), 5.93 (d, J = 8.2 Hz, 1H), 3.99 – 3.79 (m, 1H), 2.79 – 2.59 (m, 2H), 2.25 (s, 3H), 2.16 (s, 3H), 1.76 (m, 2H), 1.17 (d, J = 6.6 Hz, 3H) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ (ppm) = 155.81, 143.25, 136.34, 130.70, 129.20 (3C), 129.14 (3C), 126.50, 123.65, 46.03, 40.06, 33.22, 21.82, 21.35, 17.69 ppm. EXAMPLE 30: 1-(2,5-dimethylphenyl)-3-(4-hydroxyphenethyl)urea
Figure imgf000201_0001
1-(2,5-dimethylphenyl)-3-(4-hydroxyphenethyl)urea (30) - 2,5-dimethylaniline (1,458 mmol, 1.0 equiv., 0.18 mL) and 4-(2-aminoethyl)phenol as primary amine (1.458 mmol, 1.0 equiv., 200 mg) were reacted according to Typical procedure 5. Crude was purified with SCC (100% DCM to 98/2 DCM/MeOH) to afford compound 30, 297 mg, yield: 71.6%. A fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound for characterization and biological assays.
Figure imgf000201_0002
1H NMR (300 MHz, DMSO-d6, 20°C), δ = 9.17 (s, 1H), 7.63 (d, 1H), 7.55 (s, 1H), 7.05 – 6.99 (m, 2H, 1), 6.97 (d, 1H), 6.73 – 6.65 (m, 3H), 6.46 (t, 1H), 3.30 – 3.22 (m, 2H), 2.62 (t, 2H), 2.21 (s, 3H), 2.10 (s, 3H) ppm. 13C NMR (75 MHz, DMSO-d6, 20°C), δ = 155.61, 155.29, 137.95, 134.82, 129.77, 129.58, 129.48, 123.65, 122.47, 121.24, 115.10, 40.95, 35.03, 20.91, 17.43 ppm. HRMS (M+H)+: Calculated: 285.1597 - Found: 285.1573. EXAMPLE 31: N-(2,5-dimethylphenyl)-4-(3-hydroxyphenyl)piperazine-1- carboxamide
Figure imgf000202_0001
STEP 1 N-(2,5-dimethylphenyl)-4-(3-methoxyphenyl)piperazine-1-carboxamide (Z1) - According to Typical procedure 5 using 2-isocyanato-1,4-dimethylbenzene (0.219 mmol, 1.0 equiv., 32 mg) and 1-(3-methoxyphenyl)piperazine hydrochloride (0.219 mmol, 1.0 equiv., 50 mg) in dry DCM (3 mL) followed by triethylamine (0.219 mmol, 1.0 equiv., 30 µL). The mixture was stirred at room temperature overnight. The precipitate was filtered and washed with cold DCM to afford: Z1, 47 mg, yield: 63%.
Figure imgf000202_0002
1H NMR (400 MHz, DMSO-d6, 20°C), δ = 8.05 (s, 1H), 7.13 (t, 1H), 7.08 – 6.99 (m, 2H), 6.89 – 6.82 (m, 1H), 6.57 (ddd, 1H), 6.50 (t, 1H), 6.40 (ddd, 1H), 3.73 (s, 3H), 3.56 (t, 4H), 3.15 (t, 4H), 2.23 (s, 3H), 2.11 (s, 3H) ppm. 13C NMR (101 MHz, DMSO-d6, 20°C), δ = 160.20, 155.60, 152.34, 137.62, 134.65, 129.91, 129.83, 129.64, 126.63, 125.25, 108.36, 104.55, 101.89, 54.89, 48.30, 43.74, 20.51, 17.50 ppm. STEP 2 N-(2,5-dimethylphenyl)-4-(3-hydroxyphenyl)piperazine-1-carboxamide (31) - According to Typical procedure 6 using N-(2,5-dimethylphenyl)-4-(3-methoxyphenyl)piperazine-1- carboxamide Z1 (74 µmol, 1.0 equiv., 25 mg) in dry DCM (2 mL). The mixture was quenched by adding of ice (# 2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi- preparative HPLC to afford sufficient quantity of pure compound 31 for characterization and biological assays.
Figure imgf000203_0001
1H NMR (400 MHz, DMSO-d6, 20°C), δ = 9.14 (s, 1H, OH 19), 8.04 (s, 1H, NH 9), 7.02 (t, J = 8.0 Hz, 1H, CH 15), 7.03 (d, J = 2.0 Hz, 1H, CH 8), 7.02 (d, J = 2.0 Hz, 1H, CH 4), 6.89 – 6.82 (dd, J = 8.0, 2.0 Hz, 1H, CH 3), 6.42 (dd, J = 8.0, 2.2 Hz, 1H, CH 16), 6.34 (t, J = 2.2 Hz, 1H, CH 18), 6.24 (dd, J = 8.0, 2.2 Hz, 1H, CH 14), 3.56 (t, J = 5.0 Hz, 4H, CH212), 3.10 (t, J = 5.0 Hz, 4H, CH211), 2.23 (s, 3H, CH31), 2.11 (s, 3H, CH36) ppm. 13C NMR (101 MHz, DMSO-d6, 20°C), δ = 158.10, 155.60, 152.34, 137.62, 134.64, 129.89, 129.82, 129.55, 126.60, 125.23, 106.95, 106.52, 102.80, 48.37, 43.76, 20.51, 17.50 ppm. HRMS (M+Na)+: Calculated: 348.16825 - Found: 348.1683. HPLC purity @ λ = 254 nm: 99%. MS (ESI+): m/z (%) 326.0 (63) [M+H+]. EXAMPLE 32: 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(p-tolyl)urea STEP 1 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(p-tolyl)urea (AA1) - According to typical procedure 13 part 2 using 1-(3-isocyanatobutyl)-4-methoxybenzene (50 mg, 0.244 mmol, 1 equiv.) and p-toluidine (26,04 mg, 0.244 mmol, 1 equiv.). The crude product was purified by SCC with solid deposition (75:25 petroleum ether/acetone), yielded the pure desired product AA1: 40 mg, 52%. 1H NMR (300 MHz, Acetone-d6, 20 °C), δ = 7.67 (s, NH), 7.35 (d, J = 9 Hz, CHAr × 2), 7.13 (d, J = 9 Hz, CHAr × 2), 7.03 (d, J = 9 Hz, CHAr × 2), 6,82 (d, J = 9 Hz, CHAr × 2), 5,59 (d, J = 9 Hz, NH), 3.86 (m, CH), 3.74 (s, CH3), 2.52 (m, CH2), 2.23 (s, CH3), 1.73 (m, CH2), 1.15 (d, J = 6 Hz, CH3) ppm. STEP 2 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(p-tolyl)urea (32) - According to typical procedure 6 using 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(p-tolyl)urea AA1 (40 mg, 0.128 mmol, 1 equiv.) and BBr3 (192.46 mg, 0.768 mmol, 6 equiv.). The crude product was purified by SCC with solid deposition and gradient elution (99:1 to 95:5 dichloromethane/methanol), yielded the pure desired compound 32: 38 mg, 97%. 1H NMR (300 MHz, Acetone-d6, 20°C), δ = 8.1 (s, NH), 7.68 (s, OH), 7.3 (d, J = 9 Hz, CHAr × 2), 7.01 (dd, J = 9 Hz, CHAr × 2), 6.74 (d, J = 9 Hz, CHAr × 2), 5.59 (d, J = 9 Hz, NH), 3.86 (m, J = 9 Hz, CH), 2.58 (m, CH2), 2.23 (s, CH3), 1.68 (m, CH2), 1.15 (d, J = 6 Hz, CH3) ppm. 13C NMR (75 MHz, Acetone-d6, 20°C), δ = 155.42, 154.90, 138.31, 132.88, 130.26, 129.17, 118.15, 115.06, 45.04, 39.44, 31.41, 20.96, 19.78 ppm. EXAMPLE 39: 1-(2,5-dimethylbenzyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea STEP 1 1-(2,5-dimethylbenzyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea (AB1) - According to Typical procedure 5 Bis using 2,5-dimethylphenyl)methanamine (0.75 mmol, 1.0 equiv., 0.1 mL) and 4-(4-methoxyphenyl)butan-2-amine W1 (0.75 mmol, 1.0 equiv., 101 mg). The crude product was purified by SCC (DCM/Et2O – 80:20), afforded 1-(2,5-dimethylbenzyl)- 3-(4-(4-methoxyphenyl)butan-2-yl)urea AB1, 135 mg, yield: 53%, as a white solid. 1H NMR (300MHz, Chloroform-d1, 20°C), δ (ppm): 7.04 – 6.98 (m, 4H), 6.94 – 6.91 (m, 1H), 6.78 – 6.75 (m, 2H), 5.15 (s, 1H), 4.95 (s, 1H), 4.20 (s, 2H), 3.76 – 3.66 (m, 1H), 3.75 (s, 3H), 2.57 – 2.50 (m, 2H), 2.23 (s, 3H), 2.21 (s, 3H) 1.66 – 1.58 (m, 2H), 1.08 (d, 3H). 13C NMR (75MHz, Chloroform-d1, 20°C), δ (ppm): 158.2, 157.8, 136.9, 135.5, 134.2, 133.0, 130.3, 129.2, 128.8, 128.0, 113.8, 55.3, 45.8, 42.5, 39.6, 31.6, 30.9, 21.8, 21.0, 18.5. STEP 2 1-(2,5-dimethylbenzyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea (39) - According to Typical procedure 6 using 1-(2,5-dimethylbenzyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea AB1 (0.159 mmol, 1.0 equiv., 54 mg) as methoxy-protected urea in DCM (3 mL) with BBr3 (1M in DCM, 0.635 mmol, 4.0 equiv., 0.635 mL). The mixture was quenched by adding ice (#2 g / 0.1 mmol), extraction with DCM. Solvents were removed and a purification by SCC (DCM/Et2O – 60:40), afforded 1-(2,5-dimethylbenzyl)-3-(4-(4-hydroxyphenyl)butan-2- yl)urea 15, 38 mg, yield: 73%. A fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound for characterization and biological assays. 1H NMR (300MHz, Chloroform-d1, 20°C), δ (ppm): 8.06 (s, 1H, OH 21), 7.09 (d, J = 1.9 Hz, 1H, CH 1), 7.06 – 7.00 (m, 2H, CH 18), 7.02 (d, J = 7.9 Hz, 1H, CH 5), 6.93 (dd, J = 7.6, 1.9 Hz, 1H, CH 4), 6.75 – 6.70 (m, 2H, CH 19), 5.57 (t, 1H, NH 10), 5.39 – 5.36 (d, J = 8.4 Hz, 1H, NH 12), 4.35 – 4.22 (m, 2H, CH29), 3.88 – 3.74 (m, 1H, CH 13), 2.60 – 2.53 (m, 2H, CH216), 2.25 (s, 3H, CH3), 2.24 (s, 3H, CH3), 1.71 – 1.63 (m, 2H, CH215), 1.11 (d, J = 6.5 Hz, 3H, CH314). 13C NMR (75MHz, Chloroform-d1, 20°C), δ (ppm): 158.5 (COH 20), 156.3 (CO 11), 139.2 (C 8), 135.8 (CH 4), 133.9 (C 2), 133.5 (C 6), 130.8 (CH 5), 130.1 (CH 18), 129.3 (C 17), 128.2 (CH 1), 115.9 (CH 19), 46.1 (CH 13), 42.4(CH29), 40.6 (CH215), 32.3 (CH216), 22.1 (CH33), 21.1 (CH314), 18.5 (CH37). HRMS (ESI+): m/z calcd for C20H26N2O2Na+ (M+Na)+ 349.18865; found 349.18860. HPLC purity @ λ=254 nm: 74% (General conditions, Rt = 17.13 min). MS (ESI+): m/z (%) 327.1 (100) [M+H+]. EXAMPLE 40: 1-(2,5-dimethylphenyl)-3-(3-(3-hydroxyphenyl)prop-2-yn-1-yl)urea 1-(2,5-dimethylphenyl)-3-(3-(3-hydroxyphenyl)prop-2-yn-1-yl)urea (40) - According to Typical procedure 2 using 1-(2,5-dimethylphenyl)-3-(prop-2-yn-1-yl)urea D1 (0.227 mmol, 1.0 equiv., 46 mg) as alkyne derivative and 3-iodophenol (0.227 mmol, 1.0 equiv., 50 mg) as iodine derivative. The mixture was stirred overnight. The solution was diluted with H2O (4 mL) and extracted twice with EtOAc, washed with brine and dried with MgSO4. The organic layer was filtered and concentrated under vacuo. Crude was triturated with DiPE and filtered to afford crude 1-(2,5-dimethylphenyl)-3-(3-(3-hydroxyphenyl)prop-2-yn-1- yl)urea 40, 52 mg. A fraction (10 - 20 mg) of the crude mixture was purified by semi- preparative HPLC to afford sufficient quantity of pure compound, as a white solid, for characterization and biological assays.
Figure imgf000207_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.60 (s, 1H, OH 21), 7.76 (d, J = 2.0 Hz, 1H, CH 8), 7.30 (s, 1H, NH 9), 7.17 (t, J = 8.1 Hz, 1H, CH 17), 7.00 (d, J = 7.6 Hz, 1H, CH 4), 6.89 (ddd, J = 8.2, 2.4, 1.2 Hz , 1H, CH 18), 6.87 (t, J = 2.4 Hz, 1H, CH 20), 6.83 (ddd, J = 8.2, 2.4, 1.2 Hz , 1H, CH 16), 6.74 (dd, J = 7.6, 2.0 Hz, 1H, CH 3), 6.38 (t, J = 5.5 Hz, 1H, NH 11), 4.23 (d, J = 5.5 Hz, 2H, CH212), 2.25 (s, 3H, CH31), 2.18 (s, 3H, CH36). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 156.9 (COH 19), 154.8 (CO 10), 137.8 (C 5), 135.5 (C 2), 129.9 (CH 4), 129.6 (CH 17), 124.5 (C 7), 124.1 (C 15), 123.3 (CH 3), 122.8 (CH 16), 122.0 (CH 8), 118.0 (CH 20), 115.8 (CH 18), 86.5 (CC 14), 81.8 (CC 13), 29.7 (CH212), 20.4 (CH31), 16.8 (CH36). HRMS (ESI+): m/z calcd for C18H18N2O2Na+ (M+Na)+ 317.12605; found 317.12580. HPLC purity @ λ=254 nm: 99% (General conditions, Rt = 18.90 min). MS (ESI+): m/z (%) 295.2 (100) [M+H+]. EXAMPLE 41: 1-(2,5-dimethylphenyl)-3-(4-(3-hydroxyphenyl)but-3-yn-2-yl)urea STEP 1 But-3-yn-2-yl methanesulfonate (AC1) - Triethylamine (21.40 mmol, 1.5 equiv., 2.98 mL) was added in a solution of but-3-yn-2-ol (14.27 mmol, 1.0 equiv., 1.129 mL, 1.0 g) in DCM (10 mL). The resulting solution was cooled to 0°C and mesyl chloride (17.12 mmol, 1.2 equiv., 1.33 mL, 1.96 g) was added dropwise. The mixture was allowed to reach ambient temperature and stirred for 4h. DCM (10 mL) and H2O (10 mL) were added and the organic layer was separated, washed with brine (10 mL), dried and concentrated to afford crude but- 3-yn-2-yl methanesulfonate, 2.17 g, yield: 68%. The product was used in next step without purification.
Figure imgf000208_0001
1H NMR (300MHz, Chloroform-d1, 20°C), δ (ppm): 5.29 (qd, J = 6.7, 2.2 Hz, 1H), 3.12 (s, 3H), 2.70 (d, J = 2.2 Hz, 1H), 1.66 (d, J = 6.7 Hz, 3H). 13C NMR (75MHz, Chloroform-d1, 20°C), δ (ppm): 80.3, 76.4, 67.6, 39.3, 22.6. STEP 2 But-3-yn-2-amine hydrochloride (AC2) - But-3-yn-2-yl methanesulfonate AC1 (14.62 mmol, 1.0 equiv., 2.16 g) was stirred for 18h with aqueous NH3 (5.3 ml) at 25/30°C. The resulting mixture was diluted with DCM (10 mL) and H2O (10 mL), the organic layer was separated and dried over MgSO4. The organic layer was set up to reflux for 2h30, until the vapour above the double condenser no longer gave a positive alkali test. After the addition of 4N HCl in Dioxane, the solution was stirred 24h, filtered and washed with cold Et2O to give but-3-yn-2-amine hydrochloride AC2, 445 mg, yield: 29%, as an impure white solid.
Figure imgf000208_0002
1H-NMR (300MHz, DMSO-d6, 20°C), δ (ppm): 8.63 (s, 3H), 4.24 (s, 1H), 3.66 (d, 1H, J = 2.1 Hz), 1.42 (d, 3H, J = 6.9 Hz). STEP 3 1-(but-3-yn-2-yl)-3-(2,5-dimethylphenyl)urea (AC3) - Impure but-3-yn-2-amine hydrochloride AC2 (0.947 mmol, 1.0 equiv., 100 mg) was added in dry DCM (5 mL). Dry triethylamine (0.974 mmol, 1.0 equiv., 0.133 µL) was added and the resulting solution was filtered under argon using syringe and filter adapter to remove non-soluble powder. According to Typical procedure 5, 2-isocyanato-1,4-dimethylbenzene (0.947 mmol, 1.0 equiv., 139 mg) in dry DCM (2 mL) was added. The resulting precipitate was filtered to afford 1-(but-3-yn-2-yl)-3-(2,5-dimethylphenyl)urea AC3, 105 mg, yield: 51%, as a pale white solid. Mp: 200°C.
Figure imgf000209_0001
1H-NMR (300MHz, DMSO-d6, 20°C), δ (ppm): 7.67 (d, J = 1.8 Hz, 1H), 7.55 (s, 1H), 6.99 (d, J = 7.6 Hz, 1H), 6.94 (d, J = 7.9 Hz, 1H), 6.69 (dd, J = 7.6, 1.8 Hz, 1H), 4.55 – 4.39 (dtd, J = 7.9, 6.9, 2.3 Hz, 1H), 3.22 (d, J = 2.3 Hz, 1H), 2.21 (s, 3H), 2.11 (s, 3H), 1.33 (d, J = 6.9 Hz, 3H). 13C NMR (75MHz, DMSO-d6, 20°C), δ (ppm): 154.2, 137.7, 135.0, 129.9, 123.4, 122.7, 120.8, 85.8, 72.3, 36.5, 22.5, 21.0, 17.5. STEP 4 1-(2,5-dimethylphenyl)-3-(4-(3-hydroxyphenyl)but-3-yn-2-yl)urea (41) - According to Typical procedure 2 using 1-(but-3-yn-2-yl)-3-(2,5-dimethylphenyl)urea AC3 (0.227 mmol, 1.0 equiv., 46 mg) as alkyne derivative and 3-iodophenol (0.227 mmol, 1.0 equiv., 50 mg) as iodine derivative. The mixture was stirred overnight. The solution was diluted with H2O (3 mL) and extracted with EtOAc (2 × 3 mL), washed with brine (3 mL) and dried with MgSO4. Organic layer was filtered and concentrated under vacuo. Crude was triturated with DiPE and filtered to afford crude 1-(2,5-dimethylphenyl)-3-(4-(3-hydroxyphenyl)but- 3-yn-2-yl)urea 41, 55 mg. A fraction (10 - 20 mg) of the crude mixture was purified by semi- preparative HPLC to afford sufficient quantity of pure compound, as a white solid, for characterization and biological assays. 1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.62 (s, 1H, OH 22), 7.80 (d, J = 2.1 Hz, 1H, CH 8), 7.22 (s, 1H, NH 9), 7.17 (t, J = 8.1 Hz, 1H, CH 18), 6.99 (d, J = 7.6 Hz, 1H, CH 4), 6.88 (ddd, J = 8.1, 2.4, 1.1 Hz, 1H, CH 19/17), 6.86 (t, J = 2.4 Hz, 1H, CH 21), 6.83 (ddd, J = 8.1, 2.4, 1.1 Hz, 1H, CH 19/17), 6.73 (d, J = 7.6, 2.1 Hz, 1H, CH 3), 6.46 (d, J = 8.1 Hz, 1H, NH 11), 4.90 (dq, J = 8.1, 6.9 Hz, 1H, CH 12), 2.25 (s, 3H, CH31), 2.17 (s, 3H, CH36), 1.47 (d, J = 6.9 Hz, 3H, CH313). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 158.2 (COH 20), 155.2 (CO 10), 138.7 (C 5), 136.4 (C 2), 130.8 (CH 4), 130.5 (CH 18), 125.0 (C 7), 124.9 (C 16), 124.0 (CH 3), 123.6 (CH 21), 122.5 (CH 8), 118.9 (CH 17), 116.6 (CH 19), 91.3 (CC 14), 82.2 (CC 15), 38.6 (CH 12), 23.3 (CH313), 21.3 (CH31), 17.6 (CH36). HRMS (ESI+): m/z calcd for C19H20N2O2Na+ (M+Na)+ 331.1417; found 331.1421. HPLC purity @ λ=254 nm: 100% (General conditions, Rt = 19.94 min). MS (ESI+): m/z (%) 309.2 (100) [M+H+]. EXAMPLE 42: 1-(2,5-dimethylphenyl)-3-(4-(3-hydroxyphenyl)-2-methylbut-3-yn-2- yl)urea 1-(2,5-dimethylphenyl)-3-(4-(3-hydroxyphenyl)-2-methylbut-3-yn-2-yl)urea (44) - According to Typical procedure 2 using 1-(2,5-dimethylphenyl)-3-(2-methylbut-3-yn-2- yl)urea E1 (0.227 mmol, 1.0 equiv., 52 mg) as alkyne derivative and 3-iodophenol (0.227 mmol, 1.0 equiv., 50 mg) as iodine derivative. The mixture was stirred overnight. The solution was diluted with H2O (4 mL) and extracted twice with EtOAc, washed with brine and dried with MgSO4. The organic layer was filtered and concentrated under vacuo to afford crude 1-(2,5-dimethylphenyl)-3-(4-(3-hydroxyphenyl)-2-methylbut-3-yn-2yl)urea 42, 55 mg, yield: 75%. A fraction (10 - 20 mg) of the crude mixture was purified by semi- preparative HPLC to afford sufficient quantity of pure compound, as a white solid, for characterization and biological assays.
Figure imgf000211_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.50 (s, 1H, OH 22), 7.85 (d, J = 1.8 Hz, 1H, CH 8), 7.17 (s, 1H, NH 9), 7.15 (t, J = 8.0 Hz, 1H, CH 18), 6.97 (d, J = 7.6 Hz, 1H, CH 4), 6.86 (ddd, J = 8.2, 2.5, 1.1 Hz, 1H, CH 19), 6.87 (t, J = 1.8Hz, 1H, CH 21), 6.81 (ddd, J = 8.2, 2.5, 1.1 Hz, 1H, CH 17), 6.70 (d, J = 7.6, 1.8 Hz, 1H, CH 3), 6.35 (s, 1H, NH 11), 2.25 (s, 3H, CH31), 2.15 (s, 3H, CH36), 1.71 (s, 6H, CH313). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 157.2 (COH 20), 157.1 (CO 10), 138.1 (C 5), 135.5 (C 2), 129.8 (CH 4), 129.5 (CH 18), 124.4 (C 16), 123.5 (C 7), 122.8 (CH 3), 122.7 (CH 19), 121.2 (CH 8), 118.1 (CH 21), 115.5 (CH 17), 93.7 (CC 14), 80.2 (CC 15), 47.4 (C 12), 29.1 (CH313), 20.4 (CH31), 16.8 (CH36). HRMS (ESI+): m/z calcd for C20H22N2O2Na+ (M+Na)+ 345.15735; found 345.15760. HPLC purity @ λ=254 nm: 98% (General conditions, Rt = 20.91 min). MS (ESI+): m/z (%) 323.2 (100) [M+H+]. EXAMPLE 43: 1-(2,5-dimethylphenyl)-3-(6-(4-hydroxyphenyl)pyridin-2-yl)urea STEP 1 1-(6-bromopyridin-2-yl)-3-(2,5-dimethylphenyl)urea (AD1) - In a solution of triphosgene (1.45 mmol, 0.5 equiv., 429 mg) in dry DCM (12 mL) under argon at 0°C was slowly added 6-bromopyridin-2-amine (2.89 mmol, 1.0 equiv., 500 mg) followed by triethylamine (5.78 mmol, 2.0 equiv., 806 µL). The mixture was stirred at room temperature for 6 hours minimum. The solvent was then removed under reduced pressure to obtain the isocyanate derivate as a yellowish/green solid. The residue was dissolved in dry toluene (6 mL), under argon, and 2,5-dimethylaniline (2.89 mmol, 1.0 equiv., 358 µL) and triethylamine (5.78 mmol, 2.0 equiv., 806 µL) were successively added. The resulting mixture was heated and maintained to reflux for 36 hours. The solvent was evaporated under reduced pressure and the residue was triturated in DCM and filtered to afford 1-(6-bromopyridin-2-yl)-3-(2,5- dimethylphenyl)urea AD1, 552 mg, yield: 60%, as a white solid. Mp: 212°C.
Figure imgf000212_0001
1H NMR (300MHz, DMSO-d6, 20°C), δ (ppm): 10.02 (s, 1H), 9.26 (s, 1H), 7.80 (d, J = 1.8 Hz, 1H), 7.69 (t, J = 8.0 Hz, 1H), 7.55 (dd, J = 8.0, 0.8 Hz, 1H), 7.23 (dd, J = 8.0, 0.8 Hz, 1H), 7.08 (d, J = 7.7 Hz, 1H), 6.80 (dd, J = 7.7, 1.8 Hz, 1H), 2.26 (s, 6H). 13C NMR (75MHz, DMSO-d6, 20°C), δ (ppm): 153.3, 151.6, 141.4, 137.9, 136.6, 135.2, 130.0, 124.2, 123.8, 121.2, 120.8, 110.7, 21.0, 17.8. STEP 2 1-(2,5-dimethylphenyl)-3-(6-(4-hydroxyphenyl)pyridin-2-yl)urea (43) - In a mixture of Dioxane:H2O (5:1, 0.9 mL : 0.18 mL) were successively added 1-(6-bromopyridin-2-yl)-3- (2,5-dimethylphenyl)urea AD1 (0.156 mmol, 1.0 equiv., 50 mg), (4-hydroxyphenyl)boronic acid (0.156 mmol, 1.0 equiv., 22 mg) and Na2CO3 (0.625 mmol, 4.0 equiv., 66 mg). The mixture was degassed with argon and Pd(PPh3)4 (5 mol%, 9 mg) was added, and the mixture was heated at 70°C for 18 hours. The cooled reaction solution was portioned between EtOAc (3 mL) and H2O (3 mL). The organic layer was separated and washed with brine, dried over MgSO4 and concentrated in vacuo to afford 1-(2,5-dimethylphenyl)-3-(6-(4- hydroxyphenyl)pyridin-2-yl)urea 43, 37 mg. A fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound, as a white solid, for characterization and biological assays. 1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 10.66 (s, 1H, OH 21), 8.87 (s, 1H, NH 9/11), 8.77 (s, 1H, NH 9/11), 7.86 – 7.73 (m, 2H, CH 19), 7.77 (t, J = 7.7, 0.8 Hz, 1H, CH 14), 7.75 (d, J = 2.1 Hz, 1H, CH 8), 7.37 (dd, J = 7.7, 0.8 Hz, 1H, CH 15), 7.25 (dd, J = 7.7, 0.8 Hz, 1H, CH 13), 7.03 (d, J = 7.7 Hz, 1H, CH 4), 7.00 – 6.89 (m, 2H, CH 18), 6.82 (dd, J = 8.0, 2.1 Hz, 1H, CH 3), 2.29 (s, 3H, CH31), 2.12 (s, 3H, CH36). 13C NMR (75 MHz, Acetone-d6, 20°C), δ (ppm): 158.7 (COH 20), 155.3 (C 16), 153.5 (C 12), 152.5 (CO 10), 139.2 (CH 14), 137.0 (C 5), 135.4 (C 2), 130.5 (C 17), 129.9 (CH 4), 128.5 (CH 19), 125.7 (C 7), 124.3 (CH 3), 123.5 (CH 8), 115.5 (CH 18), 113.5 (CH 15), 109.6 (CH 13), 20.4 (CH31), 17.2 (CH36). HRMS (ESI+): m/z calcd for C20H19N3O2Na+ (M+Na)+ 356.13695; found 356.13660. HPLC purity @ λ=254 nm: 98% (General conditions, Rt = 20.94 min). MS (ESI+): m/z (%) 333.2 (100) [M+H+]. EXAMPLE 44: 1-(2,5-dimethylphenyl)-3-(3-(4-hydroxyphenyl)cyclohexyl)urea STEP 1 3-(4-methoxyphenyl)cyclohexan-1-one (AE1) - To a mixture of degassed [Rh(cod)Cl]2 (2 mol%, 15 mg) at room temperature were successively added H2O (6 mL), (4- methoxyphenyl)boronic acid (3.77 mmol, 2.5 equiv., 573 mg), Na2CO3 (3.02 mmol, 2.0 equiv., 320 mg) and cyclohex-2-en-1-one (1.51 mmol, 1.0 equiv., 145 mg, 147 µL). The heterogeneous mixture was heated at 80°C until completion and then cooled to room temperature, extracted with EtOAc (2 × 4 mL) and DCM (2 × 4 mL). The combined organic layers were filtered on a short pad of silica gel and evaporated under reduced pressure. A purification was performed using SCC (PE/EtOAc – 10:1) to afford 3-(4- methoxyphenyl)cyclohexan-1-one AE1, 177 mg, yield: 57%, as a whitish solid. Mp: 84°C. 1H NMR (300MHz, Chloroform-d1, 20°C), δ (ppm): 7.19 – 7.08 (m, 2H), 6.90 – 6.83 (m, 2H), 3.79 (s, 3H), 2.96 (tt, J = 11.4, 4.2 Hz, 1H), 2.64 – 2.25 (m, 4H), 2.19 – 1.99 (m, 2H), 1.90 – 1.71 (m, 2H). STEP 2 3-(4-methoxyphenyl)cyclohexan-1-amine (AE2) - According to Typical procedure 2 using 3-(4-methoxyphenyl)cyclohexan-1-one AE1 (0.734 mmol, 1.0 equiv., 150 mg). After 36h of reaction, treatment afford 3-(4-methoxyphenyl)cyclohexan-1-amine AE2, 86 mg, yield: 57%, as a colorless oil. Amine derivative was used without purification.
Figure imgf000214_0001
STEP 3 1-(2,5-dimethylphenyl)-3-(3-(4-methoxyphenyl)cyclohexyl)urea (AE3) - According to Typical procedure 5 using 2-isocyanato-1,4-dimethylbenzene (0.244 mmol, 1.0 equiv., 36 mg) and 3-(4-methoxyphenyl)cyclohexan-1-amine AE2 (0.244 mmol, 1.0 equiv., 50 mg) in dry DCM (2 mL). The solvent was evaporated and the residue was triturated with cold DCM and filtered to afford 1-(2,5-dimethylphenyl)-3-(3-(4-methoxyphenyl)cyclohexyl)urea AE3, as a supposed mixture of cis/trans diastereoisomers (30/70), 27 mg, yield: 31%. White solid.
Figure imgf000214_0002
HRMS (ESI+): m/z calcd for C22H28N2O2Na+ (M+Na)+ 375.2043; found 375.2045. HPLC purity @ λ=254 nm: 81% + 18% (General conditions, Rt1 = 23.68 min, Rt2 = 23.84 min). MS (ESI+): m/z (%) 353.3 (100) [M+H+]. STEP 4 1-(2,5-dimethylphenyl)-3-(3-(4-hydroxyphenyl)cyclohexyl)urea 44 - According to Typical procedure 6 using 1-(2,5-dimethylphenyl)-3-(3-(4-methoxyphenyl)cyclohexyl)urea AE3 (57 µmol, 1.0 equiv., 20 mg) as methoxy-protected urea in DCM (2 mL) with BBr3 (1M in DCM, 0.228 mmol, 4.0 equiv., 0.228 mL). The mixture was quenched by adding ice (#2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 44, as a white solid, for characterization and biological assays.
Figure imgf000215_0001
HRMS (ESI+): m/z calcd for C21H26N2O2Na+ (M+Na)+ 361.18865; found 361.18850. HPLC purity @ λ=254 nm: 75% + 25% (General conditions, Rt1 = 20.54 min, Rt2 = 20.75 min). MS (ESI+): m/z (%) 339.3 (100) [M+H+]. EXAMPLE 45: 1-(2,5-dimethylphenyl)-3-(1-(3-hydroxyphenyl)piperidin-4-yl)urea STEP 1 tert-butyl (1-(3-methoxyphenyl)piperidin-4-yl)carbamate (AF1) - According to Typical procedure 9 using 1-iodo-3-methoxybenzene (1.07 mmol, 1.0 equiv., 250 mg) as iodine derivative and tert-butyl piperidin-4-ylcarbamate (1.28 mmol, 1.2 equiv., 257 mg) as piperidine derivative. The crude was purified using SCC (Cyclohexane/EtOAc – 100:0 to 80:20) to afford tert-butyl (1-(3-methoxyphenyl)piperidin-4-yl)carbamate AF1, 135 mg, yield: 34%, as a yellowish solid. Mp: 120°C. Rf (Cyclohexane/EtOAc – 80:20): 0.2.
Figure imgf000216_0001
1H NMR (400MHz, Methanol-d1, 20°C), δ (ppm): 7.11 (t, J = 8.2 Hz, 1H), 6.56 (dd, J = 8.2, 2.4 Hz, 1H), 6.49 (d, J = 2.4 Hz, 1H), 6.41 (dd, J = 8.2, 2.4 Hz, 1H), 3.75 (s, 3H), 3.61 (dt, J = 12.5, 4.0 Hz, 2H), 3.47 (tt, J = 10.9, 4.0 Hz, 1H), 2.78 (td, J = 12.5, 2.6 Hz, 2H), 1.94 (dt, J = 11.7, 2.6 Hz, 2H), 1.56 (qd, J = 11.7, 4.0 Hz, 2H), 1.45 (s, 9H). 13C NMR (101MHz, Methanol-d1, 20°C), δ (ppm): 162.1, 157.8, 154.2, 130.7, 110.8, 106.0, 104.3, 80.0, 55.6, 50.2, 33.0, 28.8. One carbon was not observed. STEP 2 1-(3-methoxyphenyl)piperidin-4-amine dihydrochloride (AF2) - According to Typical procedure 10 using tert-butyl (1-(3-methoxyphenyl)piperidin-4-yl)carbamate AF1 (0.212 mmol, 1.0 equiv., 65 mg). Evaporation of solvent afford 1-(3-methoxyphenyl)piperidin-4- amine dihydrochloride AF2 in quantitative yield.
Figure imgf000216_0002
1H NMR (400MHz, Methanol-d1, 20°C), δ (ppm): 7.48 (t, J = 8.2 Hz, 1H), 7.30 (t, J = 2.5 Hz, 1H), 7.26 (dd, J = 8.2, 2.5 Hz, 1H), 7.07 (dd, J = 8.2, 2.5 Hz, 1H), 3.87 (s, 3H), 3.86 – 3.71 (m, 4H), 3.65 (tt, J = 11.2, 5.3 Hz, 1H), 2.42 – 2.22 (m, 4H). STEP 3 1-(2,5-dimethylphenyl)-3-(1-(3-methoxyphenyl)piperidin-4-yl)urea (AF3) - According to Typical procedure 5 using 2-isocyanato-1,4-dimethylbenzene (0.197 mmol, 1.0 equiv., 29 mg) and 1-(3-methoxyphenyl)piperidin-3-amine dihydrochloride AF2 (0.197 mmol, 1.0 equiv., 55 mg) in dry DCM (2 mL) followed by triethylamine (0.394 mmol, 2.0 equiv., 55 µL). The mixture was stirred at room temperature overnight. The precipitate was filtered and washed with cold DCM to afford 1-(2,5-dimethylphenyl)-3-(1-(3-methoxyphenyl)piperidin- 4-yl)urea AF3, 53 mg, yield: 76%, as a white solid.
Figure imgf000217_0001
1H NMR (300MHz, DMSO-d6, 20°C), δ (ppm): 7.87 (s, 1H), 7.70 (d, J = 1.8 Hz, 1H), 7.51 (s, 1H), 7.10 (td, J = 8.2, 2.2 Hz, 1H), 6.97 (d, J = 7.6 Hz, 1H), 6.67 (dd, J = 7.6, 1.8 Hz, 1H), 6.54 (ddd, J = 8.2, 2.6, 2.2 Hz, 1H), 6.46 (t, J = 2.6 Hz, 1H), 6.35 (dt, J = 8.2, 2.6 Hz, 1H), 3.71 (s, 3H), 3.68 – 3.62 (m, 1H), 3.60 – 3.53 (m, 1H), 2.91 – 2.81 (m, 2H), 2.21 (s, 3H), 2.12 (s, 3H), 1.97 – 1.86 (m, 2H), 1.65 – 1.37 (m, 2+1H). 13C NMR (75MHz, DMSO-d6, 20°C), δ (ppm): 160.2, 154.7, 152.4, 138.0, 134.9, 129.8, 129.6, 123.2, 122.3, 120.6, 108.5, 103.9, 101.8, 47.3, 46.1, 31.7, 29.0, 21.0, 17.5. STEP 4 1-(2,5-dimethylphenyl)-3-(1-(3-hydroxyphenyl)piperidin-4-yl)urea (45) - According to Typical procedure 6 using 1-(2,5-dimethylphenyl)-3-(1-(3-methoxyphenyl)piperidin-3- yl)urea AF3 (71 µmol, 1.0 equiv., 25 mg) as methoxy-protected urea in DCM (2 mL) with BBr3 (1M in DCM, 0.284 mmol, 4.0 equiv., 0.284 mL). The mixture was quenched by adding ice (#2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 45, as a whitish solid, for characterization and biological assays. 1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.06 (s, 1H, OH 21), 7.82 (d, J = 2.0 Hz, 1H, CH 8), 7.12 (s, 1H, NH 9), 7.00 (td, J = 8.0; 1.1 Hz, 1H, CH 17), 6.97 (d, J = 7.5 Hz, 1H, CH 4), 6.70 (dd, J = 7.5, 2.0 Hz, 1H, CH 3), 6.44 (ddd, J = 8.0, 2.0, 1.1 Hz, 1H, CH 16/18), 6.43 (td, J = 2.0, 1.1 Hz, 1H, CH 20), 6.27 (ddd, J = 8.0, 2.0, 1.1 Hz, 1H, CH 16/18), 6.05 (d, J = 7.6 Hz, 1H, NH 11), 3.79 – 3.75 (m, 1H, CH 12), 3.63 – 3.53 (m, CH214), 2.89 – 2.79 (m, CH214), 2.25 (s, 3H, CH31), 2.15 (s, 3H, CH36), 2.01 – 1.95 (m, 2H, CH213), 1.56 – 1.50 (m, 2H, CH213). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 159.1 (COH 19), 155.6 (CO 10), 153.9 (C 15), 139.1 (C 5), 136.3 (C 2), 130.7 (CH 17), 130.4 (CH 4), 124.7 (C 7), 123.7 (CH 3), 122.4 (CH 8), 108.6 (CH 16/18), 106.9 (CH 16/18), 104.1 (CH 20), 48.9 (CH214), 47.8 (CH 12), 33.1 (CH213), 21.4 (CH31), 17.7 (CH36). HRMS (ESI+): m/z calcd for C20H25N3O2Na+ (M+Na)+ 362.1839; found 362.1841. HPLC purity @ λ=254 nm: 97% (General conditions, Rt = 14.82 min). MS (ESI+): m/z (%) 340.3 (100) [M+H+]. EXAMPLE 46: 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)piperidin-4-yl)urea STEP 1 tert-butyl (1-(4-methoxyphenyl)piperidin-4-yl)carbamate (AG1) - According to Typical procedure 9 using 1-iodo-4-methoxybenzene (1.67 mmol, 1.0 equiv., 390 mg) as iodine derivative and tert-butyl piperidin-4-ylcarbamate (2.0 mmol, 1.2 equiv., 400 mg) as piperidine derivative. The crude was purified using SCC (Cyclohexane/EtOAc – 100:0 to 80:20) to afford tert-butyl (1-(4-methoxyphenyl)piperidin-4-yl)carbamate AG1, 312 mg, yield: 51%, as a white solid. Mp: 110°C. Rf (Cyclohexane/EtOAc – 80:20): 0.18. 1H NMR (400MHz, Methanol-d1, 20°C), δ (ppm): 7.00 – 6.92 (m, 2H), 6.87 – 6.78 (m, 2H), 3.73 (s, 3H), 3.47 – 3.40 (m, 2+1H), 2.71 (td, J = 12.0, 2.6 Hz, 2H), 1.95 (dd, J = 13.7, 3.9 Hz, 2H), 1.60 (qd, J = 12.0, 3.9 Hz, 2H), 1.45 (s, 9H). 13C NMR (101MHz, Methanol-d1, 20°C), δ (ppm): 157.8, 155.9, 147.1, 120.5, 115.4, 80.0, 55.9, 52.0, 49.7, 33.2, 28.8. STEP 2 4-(4-aminopiperidin-1-yl)phenol dihydrobromide (AG2) - According to Typical procedure 10 using tert-butyl (1-(4-methoxyphenyl)piperidin-4-yl)carbamate AG1 (0.134 mmol, 1.0 equiv., 41 mg). After 2 hours, the solution was quenched with ice, extracted with H2O (2×2 mL) and was evaporated under vacuo. A small amount of MeOH was added and the mixture was concentrated under vacuo to afford 4-(4-aminopiperidin-1-yl)phenol dihydrobromide AG2, 47 mg, yield: quantitative, as light yellowish solid.
Figure imgf000219_0001
1H NMR (300MHz, Methanol-d1, 20°C), δ (ppm): 7.65 – 7.54 (m, 2H), 7.00 – 6.88 (m, 2H), 3.91 – 3.69 (m, 4+1H), 2.44 – 2.20 (m, 4H). 13C NMR (75MHz, Methanol-d1, 20°C), δ (ppm): 160.4, 156.7, 123.5, 117.7, 55.7, 46.4, 29.1. STEP 3 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)piperidin-4-yl)urea (46) - According to Typical procedure 5 using 2-isocyanato-1,4-dimethylbenzene (0.127 mmol, 1.0 equiv., 19 mg) and 4-(4-aminopiperidin-1-yl)phenol dihydrobromide AG2 (0.127 mmol, 1.0 equiv., 45 mg) in dry DCM (4 mL) followed by triethylamine (0.254 mmol, 2.0 equiv., 35 µL). The mixture was stirred at room temperature overnight. Treatment afford 1-(2,5- dimethylphenyl)-3-(1-(4-hydroxyphenyl)piperidin-4-yl)urea 46, 16 mg, yield: 37%. The crude mixture was triturated with acetone to afford pure product, as a white solid, for characterization and biological assays.
Figure imgf000220_0001
1H NMR (300MHz, DMSO-d6, 20°C), δ (ppm): 8.82 (s, 1H, OH 19), 7.70 (d, J = 1.8 Hz, 1H, CH 8), 7.50 (s, 1H, NH 9), 6.97 (d, J = 7.6 Hz, 1H, CH 4), 6.86 – 6.75 (m, 2H, CH 17), 6.66 (dd, J = 7.6, 1.8 Hz, 1H, CH 3), 6.68 – 6.58 (m, 1H, CH 16), 6.60 (d, J = 8.5 Hz, 1H, NH 11), 3.63 – 3.51 (m, 1H, CH 12), 3.36 – 3.25 (m, 1H, CH214), 2.76 – 2.65 (m, 2H, CH2 14), 2.21 (s, 3H, CH31), 2.12 (s, 3H, CH36), 1.97 – 1.86 (m, 2H, CH213), 1.56 – 1.45 (m, 2H, CH213). 13C NMR (75MHz, DMSO-d6, 20°C), δ (ppm): 154.7 (CO 10), 150.9 (COH 18), 144.5 (C 15), 138.1 (C 7), 134.9 (C 2), 129.7 (CH 4), 123.1 (C 5), 122.3 (CH 3), 120.6 (CH 8), 118.5 (CH 17), 115.4 (CH 16), 49.4 (CH214), 45.9 (CH 12), 32.2 (CH213), 21.0 (CH31), 17.5 (CH36). HRMS (ESI+): m/z calcd for C20H25N3O2Na+ (M+Na)+ 362.1839; found 362.1841. HPLC purity @ λ=254 nm: 94% (General conditions, Rt = 15.78 min). MS (ESI+): m/z (%) 340.2 (100) [M+H+]. EXAMPLE 47: 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)piperidin-3-yl)urea STEP 1 tert-butyl (1-(4-methoxyphenyl)piperidin-3-yl)carbamate (AH1) - According to Typical procedure 9 using 1-iodo-4-methoxybenzene (1.752 mmol, 1.0 equiv., 410 mg) as iodine derivative and tert-butyl piperidin-3-ylcarbamate (2.102 mmol, 1.2 equiv., 421 mg) as piperidine derivative. The crude was purified using SCC (Cyclohexane/EtOAc – 100:0 to 80:20) to afford tert-butyl (1-(4-methoxyphenyl)piperidin-3-yl)carbamate AH1, 354 mg, yield: 55%, as a beige solid. Mp: 122°C. Rf (Cyclohexane/EtOAc – 80:20): 0.2.
Figure imgf000221_0001
1H NMR (400MHz, Methanol-d1, 20°C), δ (ppm): 6.98 – 6.90 (m, 2H), 6.87 – 6.78 (m, 2H), 3.73 (s, 3H), 3.65 (t, J = Hz, 1H), 3.43 (dd, J = 11.5, 4.0 Hz, 1H), 3.24 (dt, J = 11.5, 4.0 Hz, 1H), 2.67 (td, J = 11.5, 4.0 Hz, 1H), 2.50 (dd, J = 11.3, 9.1 Hz, 1H), 1.90 – 1.80 (m, 2H), 1.76 – 1.65 (m, 1H), 1.45 (s, 9H), 1.35 (qd, J = 11.5, 4.0 Hz, 1H). 13C NMR (101MHz, Methanol-d1, 20°C), δ (ppm): 157.8, 155.7, 147.3, 120.5, 115.4, 80.1, 58.4, 55.9, 52.8, 31.2, 28.8, 28.0, 24.9. STEP 2 4-(3-aminopiperidin-1-yl)phenol dihydrobromide (AH2) - According to Typical procedure 10 using tert-butyl (1-(4-methoxyphenyl)piperidin-4-yl)carbamate AH1 (0.163 mmol, 1.0 equiv., 50 mg). After 2 hours, the solution was quenched with ice, extracted with H2O (2 × 2 mL) and was evaporated under vacuo. A small amount of MeOH was added and the mixture was concentrated under vacuo to afford 4-(3-aminopiperidin-1-yl)phenol dihydrobromide AI2, 57 mg, yield: quantitative.
Figure imgf000221_0002
1H NMR (300MHz, Methanol-d1, 20°C), δ (ppm): 7.70 – 7.59 (m, 2H), 7.01 – 6.90 (m, 2H), 4.00 (tt, J = 11.5, 4.0 Hz, 1H), 3.82 (dd, J = 11.5, 4.0 Hz, 1H), 3.73 – 3.61 (m, 3H), 2.33 – 2.21 (m, 3H), 1.96 – 1.90 (m, 1H). 13C NMR (75MHz, Methanol-d1, 20°C), δ (ppm): 160.4, 134.7, 123.5, 117.8, 57.4, 57.3, 46.9, 26.9, 22.4. STEP 3 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)piperidin-3-yl)urea (47) - According to Typical procedure 5 using 2-isocyanato-1,4-dimethylbenzene (0.113 mmol, 1.0 equiv., 17 mg) and 4-(3-aminopiperidin-1-yl)phenol dihydrobromide AH2 (0.113 mmol, 1.0 equiv., 40 mg) in dry DCM (4 mL) followed by triethylamine (0.339 mmol, 3.0 equiv., 47 µL). The mixture was stirred at room temperature overnight. Treatment afford 1-(2,5- dimethylphenyl)-3-(1-(4-hydroxyphenyl)piperidin-3-yl)urea 47, 29 mg, yield: 76%. The crude mixture was triturated with acetone to afford sufficient quantity of pure product, as a white solid, for characterization and biological assays.
Figure imgf000222_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 7.85 (s, 1H, OH 21), 7.84 (d, J = Hz, 1H, CH 8), 7.27 (s, 1H, NH 9), 6.97 (d, J = 7.6 Hz, 1H, CH 4), 6.87 – 6.80 (m, 2H, CH 19), 6.75 – 6.69 (m, 2H, CH 18), 6.69 (dd, J = 7.6, 2.0 Hz, 1H, CH 3), 6.22 (d, J = 7.9 Hz, 1H, NH 11), 4.01 – 3.95 (m, 1H, CH 12), 3.33 – 3.24 (m, 1H, CH216), 3.07 – 2.88 (m, 1H, CH214), 2.86 – 2.73 (m, 1H, CH216), 2.25 (s, 3H, CH31), 2.15 (s, 3H, CH36), 1.84 – 1.79 (m, 1H, CH214), 1.83 – 1.78 (m, 1H, CH213), 1.84 – 1.76 (m, 1H, CH215), 1.70 – 1.64 (m, 1H, CH215), 1.58 – 1.51 (m, 1H, CH213). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 155.6 (CO 10), 152.3 (COH 20), 146.8 (C 7), 146.5 (C 17), 139.2 (C 5), 136.4 (C 2), 130.7 (CH 4), 123.7 (CH 3), 122.4 (CH 8), 120.1 (CH 19), 116.4 (CH 18), 58.4 (CH216), 52.4 (CH214), 46.9 (CH 12), 30.7 (CH213), 24.0 (CH215), 21.4 (CH31), 17.7 (CH36). HRMS (ESI+): m/z calcd for C20H25N3O2Na+ (M+Na)+ 362.1839; found 362.1841. HPLC purity @ λ=254 nm: 89% (General conditions, Rt = 15.12 min). MS (ESI+): m/z (%) 340.2 (100) [M+H+]. EXAMPLE 48: 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)-1- methylurea STEP 1 1-(2,5-dimethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)-1-methylurea (AI1) - 1-(2,5- dimethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea 2 (0.613 mmol, 1.0 equiv., 200 mg) was dissolved in dry THF (5 mL) and was cooled at 0°C. NaH (2.452 mmol, 4.0 equiv.) was added and the resulting mixture was stirred during 45 min. The ice bath was then removed and the iodomethane (2.452 mmol, 4.0 equiv., 0.8 mL) was slowly added and the solution was stirred overnight at room temperature. The reaction was quenched with a saturated aqueous solution of NH4Cl (5 mL) and was extracted with EtOAc (12 mL). Organic phase was washed with brine (8 mL), dried and concentrated in vacuo. The crude product was purified with SCC (PE/EtOAc – 90:10 to 60:40) to afford 1-(2,5-dimethylphenyl)-3-(4- (4-methoxyphenyl)butan-2-yl)-1-methylurea AI1, 53 mg, yield: 25%, as a colorless oil.
Figure imgf000223_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 7.19 (d, J = 7.7 Hz, 1H), 7.07 (d, J = 1.9 Hz, 1H), 7.10 – 7.02 (m, 2H), 7.05 (dd, J = 7.7, 1.9 Hz 1H), 6.85 – 6.78 (m, 2H), 4.56 (d, J = 8.5 Hz, 1H), 3.87 – 3.83 (m, 1H), 3.75 (s, 3H), 3.09 (s, 3H), 2.52 – 2.47 (m, 2H), 2.29 (s, 3H), 2.19 (s, 3H), 1.60 – 1.52 (m, 2H), 1.04 (d, J = 6.6 Hz, 3H). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 158.7, 157.2, 142.9, 138.1, 135.2, 134.3, 132.2, 130.1, 130.1, 129.5, 114.5, 55.4, 46.7, 40.1, 36.4, 32.5, 23.0, 21.9, 20.9, 17.2, 14.4. STEP 2 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)-1-methylurea (48) - According to Typical procedure 6 using 1-(2,5-dimethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)- 1-methylurea AI1 (0.156 mmol, 1.0 equiv., 53 mg) as methoxy-protected urea in DCM (4 mL) with BBr3 (1M in DCM, 0.624 mmol, 4.0 equiv., 0.624 mL). The mixture was quenched by adding ice (#2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and the crude product was purified using SCC (DCM/MeOH – 100:0 to 95:5) to afford pure 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)-1-methylurea 48, 43 mg, yield: 85%, as a colorless oil.
Figure imgf000224_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.08 (s, 1H, OH 20), 7.17 (d, J = 7.7 Hz, 1H, CH 4), 7.06 (dd, J = 7.7, 1.9 Hz, 1H, CH 3), 7.02 (d, J = 1.9 Hz, 1H, CH 8), 7.00 – 6.93 (m, 2H, CH 18), 6.75 – 6.65 (m, 2H, CH 17), 4.52 (d, J = 8.5 Hz, 1H, NH 11), 3.82 (dq, J = 8.5, 6.6 Hz, 1H, CH 12), 3.07 (s, 3H, CH311), 2.52 – 2.38 (m, 2H, CH215), 2.31 – 2.25 (m, 3H, CH31), 2.17 (s, 3H, CH36), 1.63 – 1.44 (m, 2H, CH214), 1.01 (d, J = 6.6 Hz, 3H, CH3 13). 13C NMR (75MHz, Acetone-d6, 20°C), δ = 157.2 (COH 19), 156.3 (CO 10), 142.8 (C 7), 138.0 (C 2), 134.2 (C 5), 133.9 (C 16), 132.0 (CH 4), 130.1 (CH 8), 130.0 (CH 17), 129.4 (CH 3), 115.9 (CH 18), 46.7 (CH 12), 40.1 (CH214), 36.3 (CH311), 32.5 (CH215), 21.8 (CH313), 20.8 (CH31), 17.1 (CH36). HRMS (ESI+): m/z calcd for C20H26N2O2Na+ (M+Na)+ 349.18865; found 349.18870. HPLC purity @ λ=254 nm: 95% (General conditions, Rt = 20.77 min). MS (ESI+): m/z (%) 327.3 (100) [M+H+]. EXAMPLE 49: 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)-1,3- dimethylurea STEP 1 4-(4-methoxyphenyl)-N-methylbutan-2-amine (AJ1) - Similarly to Typical procedure 12, 4-(4-methoxyphenyl)butan-2-one (2.244 mmol, 1.0 equiv., 400 mg) and MeNH2 (40% aq., 8.977 mmol, 4.0 equiv., 0.777 mL) were dissolved in MeOH (4 mL) and stirred for 45 minutes. Then, NaBH3CN (3.366 mmol, 1.5 equiv., 212 mg) was added portion wise at 0°C and the resulting mixture was stirred overnight at room temperature. The reaction was quenched by a solution of 1M HCl (15 mL). The MeOH was evaporated and the aqueous phase was washed with Et2O (2×10 mL) and basified with 1M NaOH (16 mL). The solution was extracted with DCM (2×15 mL), combined organic layers were dried and concentrated in vacuo to afford 4-(4-methoxyphenyl)-N-methylbutan-2-amine AJ1, 140 mg, yield: 32%, as a clear oil.
Figure imgf000225_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 7.15 – 7.08 (m, 2H), 6.85 – 6.79 (m, 2H), 3.75 (s, 3H), 2.61 – 2.55 (m, 2H), 2.53 – 2.44 (m, 1H), 2.32 (s, 3H), 1.87 – 1.83 (m, 1H), 1.73 – 1.46 (m, 2H), 1.04 – 1.00 (d, J = 6.3 Hz, 3H). STEP 2 3-(2,5-dimethylphenyl)-1-(4-(4-methoxyphenyl)butan-2-yl)-1-methylurea (AJ2) - According to Typical procedure 5 using 4-(4-methoxyphenyl)-N-methylbutan-2-amine AJ1 (0.724 mmol, 1.0 equiv., 140 mg) and 2-isocyanato-1,4-dimethylbenzene (0.724 mmol, 1.0 equiv., 0.103 mL) were dissolved in dry DCM (4 mL) and stirred overnight at room temperature. The solvent was evaporated in vacuo to afford pure 3-(2,5-dimethylphenyl)-1- (4-(4-methoxyphenyl)butan-2-yl)-1-methylurea AJ2, 253 mg, yield: quantitative; as a clear oil. 1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 7.57 (d, J = 1.9 Hz, 1H), 7.10 – 7.05 (m, 2H), 7.00 (d, J = 7.7 Hz 1H), 6.80 – 6.76 (m, 2H), 6.05 (s, 1H), 4.45 – 4.38 (m, 1H), 3.72 (s, 3H), 2.77 (s, 3H), 2.66 – 2.45 (m, 2H), 2.28 (s, 3H), 2.12 (s, 3H), 1.85 – 1.62 (m, 2H), 1.13 (d, J = 6.8 Hz, 3H). STEP 3 1-(2,5-dimethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)-1,3-dimethylurea (AJ3) - In inert atmosphere, 3-(2,5-dimethylphenyl)-1-(4-(4-methoxyphenyl)butan-2-yl)-1- methylurea AJ2 (0.743 mmol, 1.0 equiv., 253 mg) was dissolved in dry THF (5 mL) and cooled to 0°C. Then, NaH (7.431 mmol, 10.0 equiv.) was added and the resulting mixture was stirred for 45 min. The ice bath was removed and iodomethane (7.431 mmol, 10.0 equiv., 0.46 mL) was slowly added and the reaction was stirred overnight. The solution was quenched with a saturated aqueous solution of NH4Cl sat. (5 mL) and extracted with EtOAc (15 mL), organic layer was washed with brine (8 mL), dried and concentrated in vacuo. The crude product was purified with SCC (PE/EtOAc – 100:0 to 90:10) to afford pure 1-(2,5- dimethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)-1,3-dimethylurea AJ3, 221 mg, yield: 84%, as a colorless oil.
Figure imgf000226_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 7.07 (d, J = 7.7 Hz, 1H), 7.04 – 6.99 (m, 2H), 6.92 (dd, J = 7.7, 1.9 Hz 1H), 6.84 – 6.76 (m, 2H), 6.77 (d, J = 1.9 Hz, 1H), 4.06 – 3.94 (m, 1H), 3.75 (s, 3H), 2.99 (s, 3H), 2.42 – 2.16 (m, 2H), 2.34 (s, 3H), 2.22 (s, 3H), 2.16 (s, 3H), 1.64 – 1.46 (m, 2H), 0.83 (d, J = 6.7 Hz, 3H). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 163.3, 157.7, 145.4, 136.7, 134.0, 131.2, 130.9, 129.1, 127.3, 127.1, 113.7, 55.2, 51.5, 38.9, 36.3, 31.9, 28.4, 20.7, 17.3, 17.2. STEP 4 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)-1,3-dimethylurea (49) - According to Typical procedure 6 using 1-(2,5-dimethylphenyl)-3-(4-(4- methoxyphenyl)butan-2-yl)-1,3-dimethylurea AJ3 (0.623 mmol, 1.0 equiv., 221 mg) as methoxy-protected urea in DCM (14 mL) with BBr3 (1M in DCM, 2.492 mmol, 4.0 equiv., 2.492 mL). The mixture was quenched by adding ice (#2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 49, as whitist oil, for characterization and biological assays.
Figure imgf000227_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ(ppm): 8.13 (s, 1H, OH 20), 7.14 (d, J = 7.7 Hz, 1H, CH 4), 6.96 (dd, J = 7.7, 1.9 Hz, 1H, CH 3), 7.00 – 6.92 (m, 2H, CH 18), 6.78 (d, J = 1.9 Hz, 1H, CH 8), 6.78 – 6.70 (m, 2H, CH 17), 4.02 – 3.90 (m, 1H, CH 12), 2.92 (s, 3H, CH39), 2.37 (s, 3H, CH311), 2.37 – 2.30 (m, 2H, CH215), 2.23 (s, 3H, CH31), 2.17 (s, 3H, CH36), 1.68 – 1.45 (m, 2H, CH214), 0.81 (d, J = 6.7 Hz, 3H, CH313). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 163.7 (CO 10), 156.4 (C 19), 146.8 (C 7), 137.5 (C 2), 133.7 (C 5), 132.1 (CH 4), 132.0 (C 16), 130.0 (CH 17), 128.0 (CH 8), 127.8 (CH 3), 116.0 (CH 18), 52.2 (CH 12), 39.3 (C 9), 37.2 (CH214), 32.7 (CH215), 28.7 (CH3 11), 20.8 (CH31), 17.6 (CH313), 17.4 (CH36). HRMS (ESI+): m/z calcd for C21H28N2O2Na+ (M+Na)+ 363.2043; found 363.2045. HPLC purity @ λ=254 nm: 98% (General conditions, Rt = 22.13 min). MS (ESI+): m/z (%) 341.3 (100) [M+H+]. EXAMPLE 50: 4-(4-hydroxyphenyl)butan-2-yl (2,5-dimethylphenyl)carbamate STEP 1 4-(4-((tert-butyldimethylsilyl)oxy)phenyl)butan-2-one (AK1) - In a round bottom flask, was dissolved 4-(4-hydroxyphenyl)butan-2-one (18.26 mmol, 1.0 equiv., 3.0 g) and imidazole (73.07 mmol, 4.0 equiv., 4.97 g) in dry DCM (73 mL, 0.25M). tBDMSCl (36.54 mmol, 2.0 equiv., 6.31 mL, 5.51 g) was slowly added and the resulting mixture was stirring overnight at room temperature. An aqueous solution of NH4Cl (20 mL) was added and the solution was extracted by DCM (20 mL). Combined organic layer was dried over MgSO4, filtered and concentrated under vacuum. Purification was performed on SCC (Cyclohexane/EtOAc – 100:0 to 60:40) to afford 4-(4-((tert-butyldimethylsilyl)oxy)phenyl)butan-2-ol AK1, 5.37 g, yield: 107%, as a colorless oil. Rf (Cyclohexane/EtOAc – 60:40): 0.65.
Figure imgf000228_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 7.14 – 7.03 (m, 2H), 6.82 – 6.73 (m, 2H), 2.79 – 2.70 (m, 4H), 2.08 (s, 3H), 0.98 (s, 9H), 0.19 (s, 6H). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 207.3, 154.6, 135.2, 130.1, 120.7, 45.5, 30.2, 29.6, 26.1, 18.7, -4.3. STEP 2 4-(4-((tert-butyldimethylsilyl)oxy)phenyl)butan-2-ol AK2 - In a round bottom flask, was dissolved 4-(4-((tert-butyldimethylsilyl)oxy)phenyl)butan-2-one AK1 (2.16 mmol, 1.0 equiv., 600 mg) in dry MeOH (5 mL). The stirring solution was cooled at 0°C. NaBH4 (2.16 mmol, 1.0 equiv., 81 mg) was added portion wise (~20 mg × 4) before return to room temperature. After full conversion (Monitored by TLC, around 3h), the solvent was evaporated and the resulting mixture was redissolved in EtOAc (20 mL), washed with brine (20 mL). The aqueous phase was extracted by EtOAc (10 mL). The combined organic layers were washed with brine (10 mL), dried over MgSO4, filtered and evaporated under vacuo to obtain crude 4-(4-((tert-butyldimethylsilyl)oxy)phenyl)butan-2-ol AK2, 551 mg, as a colorless oil, used directly in next step without purification. Rf (Cyclohexane/EtOAc – 60:40): 0.55.
Figure imgf000229_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 7.12 – 7.06 (m, 2H), 6.80 – 6.74 (m, 2H), 3.72 (qd, J = 6.2, 4.9 Hz, 1H), 3.47 (d, J = 4.9 Hz, 1H), 2.77 – 2.50 (m, 2H), 1.76 – 1.57 (m, 2H), 1.15 (d, J = 6.2 Hz, 3H), 0.99 (s, 9H), 0.19 (s, 6H). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 154.4, 136.4, 130.1, 120.6, 66.9, 42.3, 32.0, 26.1, 24.1, 18.7, -4.3. STEP 3 4-(4-((tert-butyldimethylsilyl)oxy)phenyl)butan-2-yl (2,5-dimethylphenyl) carbamate AK3 - In a round bottom flask, under argon, were successively added 4-(4-((tert- butyldimethylsilyl)oxy)phenyl)butan-2-ol AK2 (0.178 mmol, 1.0 equiv., 50 mg), DiPEA (0.356 mmol, 2.0 equiv., 62 µL) and 4-dimethylaminopyridine (DMAP)(0.073 mmol, 0.4 equiv., 9 mg) in dry DCM (1.8 mL), and 2-isocyanato-1,4-dimethylbenzene (0.356 mmol, 2.0 equiv., 52 mg). The reaction was stirred at room temperature for 1h. The solvent was removed under reduced pressure and the residue was purified by SCC (Cyclohexane/EtOAc – 100:0 to 90:10) to afford 4-(4-((tert-butyldimethylsilyl)oxy)phenyl)butan-2-yl (2,5- dimethylphenyl)carbamate AK3, 48 mg, yield: 63%, as a colorless oil. Rf (Cyclohexane/EtOAc – 70:30): 0.75.
Figure imgf000229_0002
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 7.69 (s, 1H), 7.50 – 7.42 (m, 1H), 7.14 – 7.06 (m, 2H), 7.06 (d, J = 7.7 Hz, 1H), 6.88 – 6.80 (m, 1H), 6.83 – 7.76 (m, 2H), 4.88 – 4.82 (m, 1H), 2.78 – 2.56 (m, 1H), 2.64 – 2.58 (m, 1H), 2.28 (s, 3H), 2.24 (s, 3H), 1.99 – 1.77 (m, 2H), 1.27 (d, J = 6.3 Hz, 3H), 0.98 (s, 3H), 0.19 (s, 6H). STEP 4 4-(4-hydroxyphenyl)butan-2-yl (2,5-dimethylphenyl)carbamate (50) - In a round bottom flask under argon was added 4-(4-((tert-butyldimethylsilyl)oxy)phenyl)butan-2-yl (2,5- dimethylphenyl)carbamate AK3 (0.09 mmol, 1.0 equiv., 42 mg) in THF (0.34 mL) followed by dropwise addition of TBAF (1M solution in THF, 0.14 mmol, 1.5 equiv., 43 µL). The reaction mixture was stirred at room temperature overnight. The solution was diluted with H2O (0.5 mL) and extracted with EtOAc (3 × 1 mL). The combined organic layers were dried over MgSO4, filtered and concentrated under reduced pressure. Purification was performed by preparative TLC (Cyclohexane/EtOAc – 100:0 to 70:30, Rf : 0.35) to give the pure 4-(4-hydroxyphenyl)butan-2-yl (2,5-dimethylphenyl)carbamate 50, 10 mg, yield: 32%, as a beige solid. Mp: 115°C.
Figure imgf000230_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.10 (s, 1H, OH 18), 7.70 (s, 1H, NH 9), 7.47 (d, J = 1.8 Hz, 1H, CH 8), 7.10 – 7.05 (m, 2+1H, CH 16+4 ), 6.89 – 6.80 (m, 1H, CH 3), 6.80 – 6.69 (m, 2H, CH 17), 4.84 (dqd, J = 7.7, 6.2, 5.1 Hz, 1H, CH 11), 2.72 – 2.49 (m, 2H, CH214), 2.27 (s, 3H, CH31), 2.24 (s, 3H, CH36), 1.99 – 1.69 (m, 2H, CH213), 1.27 (d, J = 6.2 Hz, 3H, CH312). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 157.5 (COH 18), 155.1 (CO 10), 138.4 (C 5), 136.7 (C 2), 133.2 (C 15), 131.2 (CH 16), 130.3 (CH 4), 126.0 (CH 3), 124.7 (CH 8), 116.3 (CH 7), 116.2 (CH 17) , 71.5 (CH 11), 38.7 (CH213), 32.3 (CH214), 21.4 (CH312), 20.8 (CH31), 17.8 (CH36). HRMS (ESI+): m/z calcd for C19H23N3O2Na+ (M+Na)+ 336.15701; found 336.15700. HPLC purity @ λ=254 nm: 95% (General conditions, Rt = 22.06 min). MS (ESI+): m/z (%) 314.2 (100) [M+H+]. EXAMPLE 51: 1-(2,4-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea STEP 1 1-(2,4-dimethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea (AL1) - According to Typical procedure 5 using 4-(4-methoxyphenyl)butan-2-amine W1 (0.276 mmol, 1.0 equiv., 41 mg) and isocyanato-2,4-dimethylbenzene (0.279 mmol, 1.0 equiv., 50 mg). The crude product was purified by SCC (PE/Acetone – 100:0 to 80:20), afforded 1-(2,4- dimethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea AL1, 103 mg, yield: 74%, as a white solid. Mp: 158°C. Rf (DCM/Acetone – 95:5): 0.35
Figure imgf000231_0001
1H NMR (300MHz, DMSO-d6, 20°C), δ (ppm): 7.67 (d, J = 8.1 Hz, 1H), 7.43 (s, 1H), 7.16 – 7.07 (m, 2H), 6.95 – 6.85 (m, 2H), 6.86 – 6.80 (m, 2H), 6.36 (d, J = 8.0 Hz, 1H), 3.71 (s, 3H), 3.68 – 3.59 (m, 1H), 2.60 – 2.51 (m, 2H), 2.19 (s, 3H), 2.14 (s, 3H), 1.74 – 1.55 (m, 1H), 1.10 (d, J = 6.5 Hz, 3H). 13C NMR (75MHz, DMSO-d6, 20°C), δ (ppm): 157.4, 155.0, 135.7, 133.8, 130.56, 130.5, 129.1, 126.7, 126.4, 120.6, 113.7, 54.9, 44.4, 38.8, 30.9, 21.3, 20.3, 17.8. STEP 2 1-(2,4-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea (51) - According to Typical procedure 6 using 1-(2,4-dimethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea AL1 (0.153 mmol, 1.0 equiv., 50 mg) as methoxy-protected urea in DCM (7 mL) with BBr3 (1M in DCM, 0.613 mmol, 4.0 equiv., 0.613 mL). The mixture was quenched by adding ice (#2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a purification by SCC (DCM/MeOH – 100:0 to 98:2), afforded the pure 1-(2,4-dimethylphenyl)-3-(4-(4- hydroxyphenyl)butan-2-yl)urea 51, 24 mg, yield: 51%, as a white solid.
Figure imgf000232_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.09 (s, 1H, OH 20), 7.76 (d, J = 8.9 Hz, 1H, CH 6), 7.05 (s, 1H, NH 9), 7.08 – 6.98 (m, 2H, CH 18), 6.92 (s, 1H, CH 3), 6.91 (d, J = 8.9 Hz, 1H, CH 7), 6.78 – 6.68 (m, 2H, CH 17), 5.81 (d, J = 8.2 Hz, 1H, NH 11), 3.85 (dtq, J = 8.2, 7.3, 6.5 Hz, 1H, CH 12), 2.70 – 2.48 (m, 2H, CH215), 2.22 (s, 3H, CH35), 2.17 (s, 3H, CH31), 1.79 – 1.60 (m, 2H, CH214), 1.15 (d, J = 6.6 Hz, 3H, CH313). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 156.3 (COH 19), 156.0 (CO 10), 136.8 (C 8), 133.8 (C 16), 132.3 (C 4), 132.0 (C 2), 131.5 (CH 3), 130.1 (CH 18), 127.5 (CH 7), 122.5 (CH 6), 115.9 (CH 17), 46.0 (CH 12), 40.4 (CH214), 32.3 (CH215), 21.9 (CH313), 20.7 (CH35), 18.1 (CH31). HRMS (ESI+): m/z calcd for C19H24N2O2Na+ (M+Na)+ 335.1730; found 335.1732. HPLC purity @ λ=254 nm: 96% (General conditions, Rt = 18.69 min). MS (ESI+): m/z (%) 313.2 (100) [M+H+]. EXAMPLE 52: 1-(3-cyclopropylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea STEP 1 1-(3-cyclopropylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea (AM1) - According to Typical procedure 5 Bis part 2 using 1-(3-isocyanatobutyl)-4-methoxybenzene (0.558 mmol, 1.0 equiv., 114 mg), 3-cyclopropylaniline hydrochloride (0.558 mmol, 1.0 equiv., 95 mg) and triethylamine (1.116 mmol, 2.0 equiv., 152 μl). The crude product was purified by SCC (PE/Acetone – 100:0 to 80:20), afforded 11-(3-cyclopropylphenyl)-3-(4-(4- methoxyphenyl)butan-2-yl)urea AM1, 93 mg, yield: 49%, as a colorless mixture.
Figure imgf000233_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 7.86 (s, 1H), 7.28 – 7.18 (m, 2H), 7.09 (m, 2H), 6.84 – 6.76 (m, 3H), 6.67 – 6.60 (m, 1H), 5.76 (d, J = 8.2 Hz, 1H), 3.97 – 3.78 (m, 1H), 3.73 (s, 3H), 2.73 – 2.50 (m, 2H), 1.90 – 1.77 (m, 1H), 1.76 – 1.64 (m, 2H), 1.15 (d, J = 6.6 Hz, 3H), 0.94 – 0.80 (m, 2H), 0.67 – 0.56 (m, 3H). STEP 2 1-(3-cyclopropylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea (52) - According to Typical procedure 6 using 1-(3-cyclopropylphenyl)-3-(4-(4-methoxyphenyl)butan-2- yl)urea AM1 (0.266 mmol, 1 eq., 90 mg) as methoxy-protected urea in DCM (4 mL) with BBr3 (1M in DCM, 1.064 mmol, 4.0 equiv., 1.064 mL). The mixture was quenched by adding ice (#2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 52, as a white solid, for characterization and biological assays.
Figure imgf000233_0002
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.10 (s, 1H, OH 20), 7.71 (s, 1H, NH 9), 7.25 – 7.22 (m, 2H, CH 6-7), 7.10 – 7.07 (m, 1H, CH 4), 7.06 – 7.00 (m, 2H, CH 18), 6.77 – 6.70 (m, 2H, CH 17), 6.65 – 6.62 (m, 1H, CH 5), 5.63 (d, J = 8.2 Hz, 1H, NH 11), 3.92 – 3.79 (m, 1H, CH 12), 2.67 – 2.50 (m, 2H, CH215), 1.89 – 1.80 (m, 1H, CH 2), 1.75 – 1.67 (m, 2H, CH214), 1.15 (d, J = 6.5 Hz, 3H, CH313), 0.93 – 0.87 (m, 2H, CH26), 0.65 – 0.60 (m, 2H, CH21). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 156.3 (COH 19), 155.7 (CO 10), 145.3 (C 3), 141.8 (C 5), 133.7 (CH 17), 130.1 (CH 18), 129.3 (C 7), 119.4 (C 8), 116.0 (C 6), 116.0 (C 4), 115.9 (C 16), 45.9 (CH 12), 40.3 (CH214), 32.3 (CH215), 21.8 (CH313), 16.0 (CH 2), 9.4 (CH21). HRMS (ESI+): m/z calcd for C20H24N2O2Na+ (M+Na)+ 347.1730; found 347.1733. HPLC purity @ λ=254 nm: 99% (General conditions, Rt = 20.16 min). MS (ESI+): m/z (%) 325.3 (100) [M+H+]. EXAMPLE 53: 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(5-isopropyl-2- methylphenyl)urea STEP 1 1-(5-isopropyl-2-methylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea (AN1) - According to Typical procedure 5 Bis part 2 using 1-(3-isocyanatobutyl)-4-methoxybenzene (0.244 mmol, 1.0 equiv., 50 mg) and 5-isopropyl-2-methylaniline (0.244 mmol, 1.0 equiv., 29 mg). The crude product was purified by SCC (PE/Acetone – 100:0 to 80:20), afforded 1-(5- isopropyl-2-methylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea AN1, 56 mg, yield: 66%, as a white solid. Rf (DCM/MeOH – 98:2): 0.65.
Figure imgf000234_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 7.89 (d, J = 1.9 Hz, 1H), 7.17 – 7.10 (m, 2H), 7.08 (s, 1H), 7.01 (d, J = 7.7 Hz, 1H), 6.86 – 6.80 (m, 2H), 6.78 (dd, J = 7.7, 1.9 Hz, 1H), 5.89 (d, J = 8.2 Hz, 1H), 3.93 – 3.82 (m, 1H), 3.75 (s, 3H), 2.83 – 2.79 (m, 1H), 2.72 – 2.55 (m, 2H), 2.16 (s, 3H), 1.82 – 1.63 (m, 2H), 1.21 (d, J = 6.9 Hz, 6H), 1.16 (d, J = 6.6 Hz, 3H). STEP 2 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(5-isopropyl-2-methylphenyl)urea (53) - According to Typical procedure 6 using 1-(5-isopropyl-2-methylphenyl)-3-(4-(4-methoxyphenyl)butan- 2-yl)urea AN1 (0.158 mmol, 1.0 equiv., 56 mg) as methoxy-protected urea in DCM (3 mL) with BBr3 (1M in DCM, 0.632 mmol, 4.0 equiv., 0.632 mL). The mixture was quenched by adding ice (#2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 53, as a white solid, for characterization and biological assays.
Figure imgf000235_0001
1H NMR (500MHz, Acetone-d6, 20°C), δ (ppm): 8.04 (s, 1H, OH 21), 7.88 (d, J = 1.9 Hz, 1H, CH 9), 7.15 (s, 1H, NH 10), 7.09 – 6.99 (m, 2H, CH 18), 7.01 (d, J = 7.7 Hz, 1H, CH 5), 6.77 (dd, J = 7.7, 1.9 Hz, 1H, CH 4), 6.79 – 6.69 (m, 2H, CH 19), 5.98 (d, J = 8.1 Hz, 1H, NH 12), 3.88 – 3.84 (m, 1H, CH 13), 2.83 (sept, J = 6.9 Hz, 1H, CH 2), 2.69 – 2.54 (m, 2H, CH216), 2.18 (s, 3H, CH37), 1.80 – 1.62 (m, 2H, CH215), 1.21 (d, J = 6.9 Hz, 6H, CH3 1), 1.16 (d, J = 6.6 Hz, 3H, CH314). 13C NMR (125MHz, Acetone-d6, 20°C), δ (ppm): 156.5 (COH 20), 156.1 (CO 11), 147.8 (C 3), 139.5 (C 8), 134.0 (C 17), 130.9 (CH 5), 130.2 (CH 18), 125.5 (C 6), 121.0 (CH 4), 120.3 (CH 9), 116.1 (CH 19), 46.2 (CH 13), 40.5 (CH215), 34.8 (CH 2), 32.5 (CH216), 24.6 (CH3 1), 22.0 (CH314), 17.9 (CH37). HRMS (ESI+): m/z calcd for C21H28N2O2Na+ (M+Na)+ 363.2048; found 363.2051. HPLC purity @ λ=254 nm: 93% (General conditions, Rt = 21.60 min). MS (ESI+): m/z (%) 341.3 (100) [M+H+]. EXAMPLE 54: 1-(2-ethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea STEP 1 1-(3-cyclopropylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea (AO1) - According to Typical procedure 5 Bis part 2 using 1-(3-isocyanatobutyl)-4-methoxybenzene (0.487 mmol, 1.0 equiv., 100 mg) and 2-ethylaniline (0.487 mmol, 1.0 equiv., 59 mg). The reaction flask was sealed and heat at 35°C overnight. The solution was evaporated, triturated with DiPE and filtered to afford 1-(2-ethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea AO1, 121 mg, yield: 76%, as a whitish solid. Mp: 115°C.
Figure imgf000236_0001
1H NMR (300MHz, Chloroform-d1,, 20°C), δ (ppm): 7.33 – 7.27 (m, 2H), 7.24 – 7.19 (m, 2H), 7.11 – 7.02 (m, 2H), 6.84 – 6.77 (m, 2H), 5.89 (s, 1H), 4.31 (s, 1H), 3.98 – 3.88 (m, 1H), 3.77 (s, 3H), 2.70 – 2.53 (m, 4H), 1.73 – 1.60 (m, 2H), 1.21 (t, J = 7.6 Hz, 3H), 1.14 (d, J = 6.5 Hz, 3H). 13C NMR (75MHz, Chloroform-d1, 20°C), δ (ppm): 158.0, 156.3, 139.8, 135.4, 134.0, 129.7, 129.3, 127.3, 127.0, 126.9, 114.0, 55.4, 46.2, 39.5, 31.7, 24.6, 21.7, 14.7. STEP 2 1-(2-ethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea (54) - According to Typical procedure 6 using 1-(2-ethylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea AO1 (0.077 mmol, 1.0 equiv., 25 mg) as methoxy-protected urea in DCM (2 mL) with BBr3 (1M in DCM, 0.308 mmol, 4.0 equiv., 0.308 mL). The mixture was quenched by adding ice (#2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 54, as a white solid, for characterization and biological assays.
Figure imgf000237_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.14 (s, 1H, OH 20), 7.92 (dd, J = 8.1, 1.3 Hz, 1H, CH 7), 7.18 (s, 1H, NH 9), 7.13 (dd, J = 8.1, 1.3 Hz, 1H, CH 4), 7.10 (td, J = 7.5, 1.4 Hz, 1H, CH 6), 7.06 – 6.98 (m, 2H, CH 18), 6.94 (td, J = 7.5, 1.4 Hz, 1H, CH 5), 6.78 – 6.69 (m, 2H, CH 17), 5.95 (d, J = 8.2 Hz, 1H, NH 11), 3.87 (dtq, J = 8.2, 7.3, 6.6 Hz, 1H, CH 12), 2.60 (q, J = 7.6 Hz, 2H, CH22), 2.64 – 2.52 (m, 2H, CH215), 1.80 – 1.61 (m, 2H, CH214), 1.17 (t, J = 7.6 Hz, 3H, CH31), 1.15 (d, J = 6.6 Hz, 3H, CH313). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 156.3 (COH 19), 156.1 (CO 10), 138.6 (C 8), 134.2 (C 3), 133.7 (C 16), 130.1 (CH 18), 129.1 (CH 4), 126.9 (CH 6), 123.5 (CH 5), 122.9 (CH 7), 115.9 (CH 17), 46.0 (CH 12), 40.4 (CH214), 32.3 (CH215), 24.9 (CH22), 21.9 (CH313), 14.5 (CH31). HRMS (ESI+): m/z calcd for C19H24N2O2Na+ (M+Na)+ 335.1730; found 335.1731. HPLC purity @ λ=254 nm: 98% (General conditions, Rt = 19.36 min). MS (ESI+): m/z (%) 313.3 (100) [M+H+]. EXAMPLE 55: 1-(3-fluoro-5-methylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea STEP 1 1-fluoro-3-isocyanato-5-methylbenzene (AP1) - According to Typical procedure 5 Bis part 1 using 3-fluoro-5-methylaniline (0.799 mmol, 1.0 equiv., 100 mg) in a solution of DCM (10 mL) under argon atmosphere with triethylamine (1.6 mmol, 2.0 equiv., 216 µL). The mixture was cooled to 0-5 °C then triphosgene (0.4 mmol, 0.5 equiv., 120 mg) previously dissolved in DCM (2 mL) was slowly added to the reaction and the mixture was allowed to return at room temperature. The reaction was monitored by TLC and stopped upon complete consumption of the amine derivative (4h). The reaction mixture was concentrated in vacuo, then filtered and washed with Et2O (3 × 10 mL). The organic layer was concentrated in vacuo to afford crude 1-fluoro-3-isocyanato-5-methylbenzene AP1 which was directly reengaged to the next step.
Figure imgf000238_0001
STEP 2 1-(3-fluoro-5-methylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea (AP2) - According to Typical procedure 5 Bis part 2 using a solution 4-(4-methoxyphenyl)butan-2-amine W1 (0.799 mmol, 1.0 equiv., 143 mg) in DCM (4 mL) and the freshly prepared 1-fluoro-3- isocyanato-5-methylbenzene AP1 in DCM (2 mL) which was added to the solution. The reaction was put under argon atmosphere and stirred overnight. Solvent was evaporated and the crude compound was purified with SCC (PE/Et2O – 100:0 to 50:50) to afford 1-(3-fluoro- 5-methylphenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea AP2, 194 mg, yield: 74%, as a white solid.
Figure imgf000238_0002
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.09 (s, 1H), 7.35 (dt, J = 11.7, 2.3 Hz, 1H), 7.14 – 7.06 (m, 2H), 6.92 – 6.85 (m, 1H), 6.83 – 6.76 (m, 2H), 6.49 (dddd, J = 9.5, 2.4, 1.5, 0.7 Hz, 1H), 5.90 (d, J = 8.2 Hz, 1H), 3.97 – 3.81 (m, 1H), 2.68 – 2.52 (m, 2H), 1.80 – 1.64 (m, 2H), 1.16 (d, J = 6.6 Hz, 3H). STEP 3 1-(3-fluoro-5-methylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea (55) - According to Typical procedure 6 using 1-(3-fluoro-5-methylphenyl)-3-(4-(4-methoxyphenyl)butan-2- yl)urea AP2 (0.303 mmol, 1.0 equiv., 100 mg) as methoxy-protected urea in DCM (6 mL) with BBr3 (1M in DCM, 1.212 mmol, 4.0 equiv., 1.212 mL). The mixture was quenched by adding ice (#2 g / 0.1 mmol) and the resulting precipitate was filtrated and dried to afford 1- (3-fluoro-5-methylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea 55, 62 mg, yield: 65%. A fraction (10 - 20 mg) of the crude mixture was purified by preparative TLC (PE/EtOAc – 70:30, Rf: 0.25) to afford sufficient quantity of pure compound, as a white solid, for characterization and biological assays.
Figure imgf000239_0001
1H NMR (300MHz, DMSO-d6, 20°C), δ (ppm): 8.44 (s, 1H, NH 8), 7.23 – 7.19 (m, 1H, CH 6), 6.99 – 6.96 (m, 2H, CH 17), 6.84 – 6.83 (m, 1H, CH 1), 6.67 – 6.64 (m, 2H, CH 16), 6.52 (dd, J = 9.9, 1.9 Hz, 1H, CH 4), 6.11 (d, J = 8.1 Hz, 1H, NH 10), 3.66 – 3.60 (m, 1H, CH 11), 2.56 – 2.45 (m, 2H, CH214), 2.23 (s, 3H, CH33), 1.67 – 1.58 (m, 2H, CH213), 1.08 (d, J = 6.6 Hz, 3H, CH312). 13C NMR (75MHz, DMSO-d6, 20°C), δ (ppm): 162.4 (d, J = 239.2 Hz, C 5), 155.2 (COH 18 ), 154.5 (CO 9), 142.1 (C 7), 140.0 (C 2), 131.9 (C 15), 129.0 (CH 17), 115.0 (CH 16), 113.5 (CH 1), 107.7 (d, J = 5.8 Hz, CH 4), 101.2 (d, J = 5.8 Hz, CH 6), 44.5 (CH 11), 38.5 (CH213), 30.9 (CH214), 21.2 (CH312), 21.0 (CH33). HRMS (ESI+): m/z calcd for C18H21FN2O2Na+ (M+Na)+ 339.14793; found 339.14770. HPLC purity @ λ=254 nm: 99% (General conditions, Rt = 17.78 min). MS (ESI+): m/z (%) 317.1 (100) [M+H+]. EXAMPLE 56: 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(1H-indol-6-yl)urea STEP 1 tert-butyl 6-isocyanato-1H-indole-1-carboxylate (AQ1) - According to Typical procedure 5 Bis part 1 using tert-butyl 6-amino-1H-indole-1-carboxylate (0.86 mmol, 1.0 equiv., 200 mg) in a solution of DCM (8 mL) under argon atmosphere with triethylamine (5.166 mmol, 6.0 equiv., 0.73 mL). The mixture was cooled to 0-5 °C then triphosgene (0.43 mmol, 0.5 equiv., 127 mg) was added, and the mixture was allowed to return at room temperature. The reaction was monitored by TLC and stopped upon complete consumption of the amine derivative (6h). The reaction mixture was concentrated in vacuo, then filtered and washed with Et2O (3 × 20 mL). The organic layer was concentrated in vacuo to afford tert-butyl 6- isocyanato-1H-indole-1-carboxylate AQ1, 237 mg, yield: quantitative, with sufficient purity to be used directly in the next step without further purification.
Figure imgf000240_0001
STEP 1 Bis 4-(4-acetoxyphenyl)butan-2-aminium chloride AQ2 - To a solution of DCM (8 mL) and TFA (8 mL) was added acetic anhydride (7.93 mmol, 4.0 equiv., 810 mg) and 4-(4- hydroxyphenyl)butan-2-aminium chloride (obtained from reductive amination of frambinone without basification, 1.98 mmol, 1.0 equiv., 400 mg) at room temperature. The resulting mixture was stirred (Monitored by TLC, around 4h), and stopped upon complete consumption. The solution was concentrated and was purified using SCC (DCM/MeOH – 100:0 to 95:5) to obtain 4-(4-acetoxyphenyl)butan-2-aminium chloride AQ2, 388 mg, yield: 80%. Rf (DCM/MeOH – 95:5): 0.3.
Figure imgf000241_0001
1H NMR (300MHz, Chloroform-d1, 20°C), δ (ppm): 7.85 – 7.66 (br s, 3H), 7.18 – 7.09 (m, 2H), 6.99 – 6.92 (m, 2H), 3.25 – 3.11 (m, 1H), 2.73 – 2.52 (m, 2H), 2.28 (s, 3H), 1.98 (m, 1H), 1.78 (m, 1H), 1.27 (d, J = 6.3 Hz, 3H). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 172.8, 169.8, 150.3, 139.4, 130.1, 122.6, 55.5, 48.3, 37.1, 32.0, 20.9, 20.6. STEP 2 tert-butyl 6-(3-(4-(4-acetoxyphenyl)butan-2-yl)ureido)-1H-indole-1-carboxylate (AQ3) - According to Typical procedure 5 Bis part 2 using 4-(4-acetoxyphenyl)butan-2-aminium chloride AQ2 (0.86 mmol, 1.0 equiv., 210 mg) and triethylamine (1.72 mmol, 2.0 equiv., 0.24 mL) dissolved in DCM (9 mL). tert-butyl 6-isocyanato-1H-indole-1-carboxylate AQ1 (0.86 mmol, 1.0 equiv., 222 mg) in THF (4 mL) was added and the reaction mixture was stirred at room temperature overnight. Solvent were evaporated and the crude was purified by SCC (Cyclohexane/EtOAc – 100:0 to 70:30) to obtain tert-butyl 6-(3-(4-(4- acetoxyphenyl)butan-2-yl)ureido)-1H-indole-1-carboxylate AQ3, 178 mg, yield: 45%, as a beige solid. Rf (Cyclohexane/EtOAc – 70:30): 0.25.
Figure imgf000241_0002
1H NMR (300MHz, DMSO-d6, 20°C), δ (ppm): 8.13 (m, 1H), 7.55 (d, J = 3.7 Hz, 1H), 7.48 (d, J = 8.3 Hz, 1H), 7.21 – 7.13 (m, 2H), 7.14 (d, J = 2.0 Hz, 1H), 7.00 – 6.92 (m, 2H), 6.52 (dd, J = 3.7, 0.8 Hz, 1H), 6.41 (s, 1H), 4.60 (d, J = 8.5 Hz, 1H), 4.00 – 3.96 (m, 1H), 2.70 – 2.54 (m, 2H), 2.28 (s, 3H), 1.81 – 1.67 (m, 2H), 1.64 (s, 3H), 1.17 (d, J = 6.6 Hz, 3H). 13C NMR (75MHz, DMSO-d6, 20°C), δ (ppm): 165.0, 144.0, 134.8, 130.0, 124.5, 122.9, 121.4, 116.8, 116.6, 113.8, 105.2, 102.2, 79.2, 41.4, 34.3, 27.2, 23.4, 16.8, 16.4. STEP 3 tert-butyl 6-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-1H-indole-1-carboxylate AQ4 - According to Typical procedure 15 using tert-butyl 6-(3-(4-(4-acetoxyphenyl)butan-2- yl)ureido)-1H-indole-1-carboxylate AQ3 (0.191 mmol, 1.0 equiv., 89 mg) in THF (6 mL) at room temperature with LiOH (0.76 mmol, 4.0 equiv., 19 mg) in H2O (2 mL). The reaction was stirred for 4h. Obtained crude of tert-butyl 6-(3-(4-(4-hydroxyphenyl)butan-2- yl)ureido)-1H-indole-1-carboxylate AQ4, 80 mg, yield: 95%, as a whitist solid. Rf (Cyclohexane/EtOAc – 70:30): 0.2.
Figure imgf000242_0001
1H NMR (300MHz, DMSO-d6, 20°C), δ (ppm): 9.11 (s, 1H), 8.43 – 8.36 (m, 2H), 7.53 – 7.37 (m, 2H), 7.12 (dd, J = 8.4, 2.0 Hz, 1H), 7.04 – 6.94 (m, 2H), 6.71 – 6.61 (m, 2H), 6.58 (dd, J = 3.7, 0.8 Hz, 1H), 5.99 (d, J = 8.1 Hz, 1H), 5.75 (s, 1H), 3.68 (hept, J = 6.8 Hz, 1H), 1.63 (s, 9H), 1.11 (d, J = 6.5 Hz, 3H). 13C NMR (75MHz, DMSO-d6, 20°C), δ (ppm): 155.3, 154.8, 149.2, 137.7, 135.2, 131.9, 129.0, 124.6, 124.2, 120.8, 115.0, 114.1, 107.3, 104.1, 83.4, 54.9, 44.4, 30.9, 30.7, 27.7, 21.3. STEP 3 Bis 4-(3-(3-(1H-indol-6-yl)ureido)butyl)phenyl acetate hydrochloride (AQ5) - According to Typical procedure 10 using tert-butyl 6-(3-(4-(4-acetoxyphenyl)butan-2-yl)ureido)-1H- indole-1-carboxylate AQ4 (0.189 mmol, 1.0 equiv., 88 mg). The mixture was stirred at room temperature for 24h. The resulting solution was concentrated to give the product AQ5 which was engaged directly in the next step.
Figure imgf000243_0001
STEP 4 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(1H-indol-6-yl)urea 57 - According to Typical procedure 15 using (3-(3-(1H-indol-6-yl)ureido)butyl)phenyl acetate hydrochloride AQ5 (0.189 mmol, 1.0 equiv., 75 mg) in THF (6 mL) at room temperature with LiOH (0.76 mmol, 4.0 equiv., 18 mg) in H2O (2 mL). The reaction was stirred for 2h (Monitored by TLC). The solution was diluted with H2O (4 mL) and extracted by EtOAc (3×3 mL). A fraction (10 - 20 mg) of the crude was purified by preparative TLC (Cyclohexane/EtOAc – 60:40, Rf : 0.3) to afford sufficient quantity of pure compound 56, as a white solid, for characterization and biological assays.
Figure imgf000243_0002
1H NMR (300 MHz, Acetone-d6, 20°C), δ (ppm): 10.03 (s, 1H, NH 1), 8.06 (s, 1H, OH 21), 7.93 (dt, J = 1.7, 0.8 Hz, 1H, CH 9), 7.68 (s, 1H, NH 10), 7.38 (dt, J = 8.4, 0.7 Hz, 1H, CH 6), 7.17 (dd, J = 3.2, 2.4 Hz, 1H, CH 2), 7.09 – 6.99 (m, 2H, CH 18), 6.84 (dd, J = 8.5, 1.9 Hz, 1H, CH 7), 6.77 – 6.70 (m, 2H, CH 19), 6.35 (ddd, J = 3.1, 2.0, 1.0 Hz, 1H, CH 3), 5.57 (d, J = 8.2 Hz, 1H, NH 12), 3.89 (hept, J = 6.6 Hz, 1H, CH 13), 2.71 – 2.49 (m, 2H, CH2 16), 1.82 – 1.60 (m, 2H, CH215), 1.16 (d, J = 6.5 Hz, 3H, CH314). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 156.3 (COH 21), 156.2 (CO 11), 137.8 (C 4), 136.1 (C 8), 133.9 (C 17), 130.1 (CH 18), 124.6 (CH 2), 120.8 (CH 6), 116.0 (CH 19), 113.1 (CH 7), 102.2 (CH 9), 102.1 (CH 3), 45.9 (CH 13), 40.4 (CH215), 32.3 (CH216), 21.9 (CH314). HRMS (ESI+): m/z calcd for C19H21N3O2Na+ (M+Na)+ 346.1526; found 346.1528. HPLC purity @ λ=254 nm: 97% (General conditions, Rt = 17.59 min). MS (ESI+): m/z (%) 324.3 (100) [M+H+] EXAMPLE 57: 1-(2,5-dimethylphenyl)-3-(6-(3-hydroxyphenyl)pyridin-2-yl)urea STEP 1 1-(6-(3-((tert-butyldimethylsilyl)oxy)phenyl)pyridin-2-yl)-3-(2,5-dimethylphenyl) urea (AR1) - In a mixture of dioxane:H2O (5:1, 4.5 mL : 0.9 mL) were added 1-(6-bromopyridin- 2-yl)-3-(2,5-dimethylphenyl)urea (0.78 mmol, 1.0 equiv., 250 mg), (3-((tert- butyldimethylsilyl)oxy)phenyl)boronic acid (0.78 mmol, 1.0 equiv., 197 mg) and Na2CO3 (3.12 mmol, 4.0 equiv., 331 mg). The mixture was degassed with argon and Pd(PPh3)4 (5 mol%, 45 mg) was added, and the mixture was heated at 70°C for 18 hours. The cooled reaction solution was portioned between EtOAc (12 mL) and H2O (12 mL). The organic layer was separated and washed with brine, dried over MgSO4 and concentrated in vacuo. Crude was purified by SCC (DCM/MeOH – 100 to 99.5:0.5), to afford 1-(6-(3-((tert- butyldimethylsilyl)oxy)phenyl)pyridin-2-yl)-3-(2,5-dimethylphenyl)urea AR1, 190 mg, yield: 54%, as a yellow solid. Mp: 145°C. Rf (DCM): 0.15.
Figure imgf000244_0001
1H NMR (300MHz, DMSO-d6, 20°C), δ (ppm): 9.82 (s, 1H), 9.74 (s, 1H), 7.82 (t, J = 8.3 Hz, 1H), 7.62 (d, J = 2.2 Hz, 1H), 7.55 (dt, J = 8.0, 1.1 Hz, 1H), 7.48 (dt, J = 7.7, 1.8 Hz, 2H), 7.36 (t, J = 8.0 Hz, 1H), 7.38 (t, J = 2.0 Hz, 1H), 7.05 (d, J = 7.7 Hz, 1H), 6.92 (ddd, J = 8.0, 2.5, 1.1 Hz, 1H), 6.81 (dd, J = 7.7, 2.2 Hz, 1H), 2.26 (s, 3H), 2.11 (s, 3H), 0.92 (s, 9H), 0.16 (s, 6H). 13C NMR (75MHz, DMSO-d6, 20°C), δ (ppm): 155.5, 153.9, 153.0, 152.5, 140.1, 139.5, 136.5, 135.1, 130.0, 130.0, 125.5, 124.2, 123.0, 120.4, 119.9, 118.0, 114.4, 110.9, 66.4, 25.5, 20.9, 17.9, 17.5, -4.6. STEP 2 1-(2,5-dimethylphenyl)-3-(6-(3-hydroxyphenyl)pyridin-2-yl)urea (57) - Under argon, in dry THF (1 mL) was added 1-(6-(3-((tert-butyldimethylsilyl)oxy)phenyl)pyridin-2-yl)-3-(2,5- dimethylphenyl)urea AR1 (0.056 mmol, 1.0 equiv., 25 mg) followed by a dropwise addition of tetrabutylammonium fluoride (TBAF) (1M in THF, 0.061 mmol, 1.1 equiv., 61 µL) at room temperature (Monitored by TLC). After completion, the mixture was quenched with H2O (2 mL), extracted with EtOAc (2 mL) and the organic layer was washed with brine, dried over MgSO4 and concentrated in vacuo to afford 1-(2,5-dimethylphenyl)-3-(6-(3- hydroxyphenyl)pyridin-2-yl)urea 57, 16 mg. The crude mixture was purified by semi- preparative HPLC to afford sufficient quantity of pure compound, as a white solid, for characterization and biological assays.
Figure imgf000245_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 10.65 (s, 1H, OH 23), 8.91 (s, 1H, NH 9/11), 8.59 (s, 1H, NH 9/11), 7.83 (t, J = 7.6 Hz, 1H, CH 14), 7.79 (d, J = 2.0 Hz, 1H, CH 8), 7.42 (t, J = 1.2 Hz, CH 22), 7.40 (d, J = 7.6 Hz, 1H, CH 13), 7.38 (ddd, J = 7.9, 2.5, 1.2 Hz, 1H, CH 20), 7.31 (d, J = 7.6 Hz, 1H, CH 15), 7.30 (t, J = 7.6 Hz, 1H, CH 19), 7.02 (d, J = 7.7 Hz, 1H, CH 4), 6.92 (ddd, J = 7.9, 2.5, 1.2 Hz, 1H, CH 18), 6.81 (dd, J = 7.7, 2.0 Hz, 1H, CH 3), 2.29 (s, 3H, CH31), 2.11 (s, 3H, CH36). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 158.7 (CO 10), 156.2 (C 21), 154.5 (C 16), 153.3 (C 12), 141.5 (C 17), 140.2(CH 14), 137.9 (C 7), 136.3 (C 2), 130.8 (CH 4), 130.7 (CH 19), 126.4 (C 5), 125.1 (CH 3), 124.2 (CH 8), 119.2 (CH 20), 116.9 (CH 18), 115.3 (CH 22), 114.9 (CH 13), 111.5 (CH 15), 21.3 (CH31), 18.0 (CH36). HRMS (ESI+): m/z calcd for C20H19N3O2Na+ (M+Na)+ 356.13695; found 356.13690. HPLC purity @ λ=254 nm: 98% (General conditions, Rt = 19.46 min). MS (ESI+): m/z (%) 333.2 (100) [M+H+]. EXAMPLE 58: 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)-1H-pyrrol-3-yl)urea
Figure imgf000246_0001
STEP 1 Methyl 1-(4-methoxyphenyl)-1H-pyrrole-3-carboxylate (AS1) - In an oven dry RB flask, under argon, were added methyl 1H-pyrrole-3-carboxylate (8 mmol, 1.0 equiv., 1 g), 4- iodoanisole (10.4 mmol, 1.3 equiv., 2.43 g), CuI (10 mol%, 150 mg) K3PO4 (16 mmol, 2.0 equiv., 3.39 g), trans-1,2-diaminocyclohexane (10 mol%, 96µL) in 1,4-dioxane (20 mL). The mixture was stirred at reflux overnight. After completion of the reaction (Monitored by TLC), the mixture was allowed to return at room temperature. The mixture was quenched with H2O (20 mL) and extracted with EtOAc (2 × 20 mL). The organic layer was filtered through Celite and concentrated under pressure. The obtained residue was triturated with DiPE and filtered to afford methyl 1-(4-methoxyphenyl)-1H-pyrrole-3-carboxylate AS1, 1.0 g, yield: 54%, as a dark green solid. Mp: 58°C. 1H NMR (300MHz, Chloroform-d1, 20°C), δ (ppm): 7.60 (dd, J = 2.3, 1.6 Hz, 1H), 7.35 – 7.26 (m, 2H), 7.02 – 6.92 (m, 2H), 6.93 (dd, J = 3.0, 2.3 Hz, 1H), 6.72 (dd, J = 3.0, 1.6 Hz, 1H), 3.83 (s, 3H), 3.84 (s, 3H). 13C NMR (75MHz, Chloroform-d1, 20°C), δ (ppm): 165.3, 161.9, 158.6, 133.6, 124.9, 122.7, 121.1, 117.5, 114.9, 111.3, 55.7, 51.3. STEP 2 1-(4-methoxyphenyl)-1H-pyrrole-3-carboxylic acid (AS2) - To a solution of methyl 1-(4- methoxyphenyl)-1H-pyrrole-3-carboxylate AS1 (2.16 mmol, 1.0 equiv., 500 mg) in THF (6 mL) and methanol (3 mL) at room temperature, was added 1N aqueous NaOH solution (5.40 mmol, 2.5 equiv., 5.4 mL). The resulting solution was stirred to 50°C overnight. After completion of the reaction (Monitored by TLC), the mixture was allowed to return at room temperature and was acidified using 1N aqueous HCl. The aqueous layer was extracted with EtOAc (3 × 15 mL), and the combined organic layer were dried over MgSO4, filtered and solvent was evaporated to afford 1-(4-methoxyphenyl)-1H-pyrrole-3-carboxylic acid AS2, 0.43 g, yield: 91%, as a dark brown solid. Mp: 144°C. Rf (Cyclohexane/EtOAc – 70:30): 0.2.
Figure imgf000247_0001
1H NMR (300MHz, Chloroform-d1, 20°C), δ (ppm): 7.72 – 7.64 (m, 1H), 7.39 – 7.28 (m, 2H), 7.03 – 6.92 (m, 3H), 6.78 (dd, J = 3.0, 1.6 Hz, 1H), 3.85 (s, 3H). 13C NMR (75MHz, Chloroform-d1, 20°C), δ (ppm): 158.6, 133.3, 125.9, 122.7, 121.4, 114.8, 111.7, 55.6. STEP 3 1-(4-methoxyphenyl)-1H-pyrrole-3-carbonyl azide (AS3) - In an oven dry flask under argon, was added 1-(4-methoxyphenyl)-1H-pyrrole-3-carboxylic acid AS2 (1.97 mmol, 1.0 equiv., 0.42 g) in dry toluene (11 mL). Triethylamine (2.06 mmol, 1.05 equiv., 0.29 mL) was added and the solution was stirred 15 minutes and DPPA (2.06 mmol, 1.05 equiv., 0.45 mL) was added too. After 5h of stirring at room temperature (Monitored by TLC), the solvent was removed and the mixture was purified using SCC to afford 1-(4-methoxyphenyl)-1H- pyrrole-3-carbonyl azide AS3, 0.42 g, yield: 89%, as a greenish solid. Mp: 85°C. Rf (Cyclohexane/EtOAc – 70:30): 0.5.
Figure imgf000248_0001
1H NMR (300MHz, DMSO-d6, 20°C), δ (ppm): 8.01 (dd, J = 2.3, 1.7 Hz, 1H), 7.65 – 7.54 (m, 2H), 7.41 (dd, J = 3.1, 2.3 Hz, 1H), 7.15 – 6.93 (m, 2H), 6.67 (dd, J = 3.1, 1.7 Hz, 1H), 3.80 (s, 3H). 13C NMR (75MHz, DMSO-d6, 20°C), δ (ppm): 168.4, 158.6, 132.6, 126.0, 122.9, 122.4, 118.4, 115.3, 111.2, 55.9. STEP 4 1-(2,5-dimethylphenyl)-3-(1-(4-methoxyphenyl)-1H-pyrrol-3-yl)urea (AS4) - In an oven dry flask under argon, was added the isolated 1-(4-methoxyphenyl)-1H-pyrrole-3-carbonyl azide AS3 (0.83 mmol, 1.0 equiv., 200 mg) in dry toluene (4 mL) and heated at reflux overnight to form the isocyanate compound by Curtius rearrangement (not isolated). The mixture was allowed to return at room temperature and 2,5-dimethylaniline (0.83 mmol, 1.0 equiv., 110 µL) The mixture was filtered and the precipitate was washed with cold toluene to afford 1-(2,5-dimethylphenyl)-3-(1-(4-methoxyphenyl)-1H-pyrrol-3-yl)urea AS4, 211 mg, yield: 74%, as a yellowish solid. Mp: 195°C. 1H NMR (300MHz, DMSO-d6, 20°C), δ (ppm): 8.71 (s, 1H), 7.78 – 7.69 (m, 2H), 7.48 – 7.37 (m, 2H), 7.33 (dd, J = 2.5, 1.7 Hz, 1H), 7.26 – 7.08 (m, 1H), 7.06 – 6.94 (m, 3H), 6.76 – 6.67 (m, 1H), 6.14 (dd, J = 3.0, 1.7 Hz, 1H), 3.77 (s, 3H), 2.24 (s, 3H), 2.17 (s, 3H). 13C NMR (75MHz, DMSO-d6, 20°C), δ (ppm): 157.1, 152.9, 138.1, 135.5, 130.4, 126.2, 124.0, 120.7, 117.5, 115.2, 103.2, 55.8, 21.4, 17.9. STEP 5 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)-1H-pyrrol-3-yl)urea (58) - According to Typical procedure 6 using 1-(2,5-dimethylphenyl)-3-(1-(4-methoxyphenyl)-1H-pyrrol-3- yl)urea AS4 (75 µmol, 1.0 equiv., 25 mg) as methoxy-protected urea in DCM (2 mL) with BBr3 (1M in DCM, 0.3 mmol, 4.0 equiv., 0.3 mL). The mixture was quenched by adding ice (#2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a trituration followed by a filtration with DiPE then ACN gave 1-(2,5-dimethylphenyl)-3-(1-(4- hydroxyphenyl)-1H-pyrrol-3-yl)urea 58 with sufficient purity for characterization and biological assays.
Figure imgf000249_0001
1H NMR (300MHz, DMSO-d6, 20°C), δ (ppm): 9.45 (s, 1H, OH 20), 8.69 (s, 1H, NH 11), 7.75 (d, J = 1.8 Hz, 1H, CH 8), 7.70 (s, 1H, NH 9), 7.32 – 7.24 (m, 2H, CH 18), 7.26 (s, 1H, CH 13), 7.04 (t, J = 2.9 Hz, CH 14), 7.01 (d, J = 7.6 Hz, 1H, CH 4), 6.86 – 6.77 (m, 2H, CH 17), 6.71 (dd, J = 7.6, 1.8 Hz, 1H, CH 3), 6.11 (dd, J = 2.9, 1.7 Hz, 1H, CH 15), 2.24 (s, 3H, CH31), 2.17 (s, 3H, CH36). 13C NMR (75MHz, DMSO-d6, 20°C), δ (ppm): 158.8 (CO 10), 154.8 (COH 19), 137.7 (C 7), 135.0 (C 2), 132.5 (C 16), 129.9 (CH 4), 125.5 (CH 13), 123.5 (C 5), 122.6 (CH 3), 120.7 (CH 8), 120.5 (CH 18), 116.9 (C 12), 115.9 (CH 17), 107.1 (CH 14/15), 102.4 (CH 14/15), 21.0 (CH31), 17.5 (CH36). HRMS (ESI+): m/z calcd for C19H19N3O2Na+ (M+Na)+ 344.13695; found 344.13740. HPLC purity @ λ=254 nm: 97% (General conditions, Rt = 19.36 min). MS (ESI+): m/z (%) 322.3 (100) [M+H+]. EXAMPLE 59: 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)-2-(prop-1-en-2-yl)- 1H-indol-3-yl)urea Compound 59 is a secondary product isolated during the synthesis of Compound 9.
Figure imgf000250_0001
1H NMR (500MHz, Acetone-d6, 20°C), δ (ppm): 8.74 (s, 1H, OH 24), 7.75 (d, J = 2.0 Hz, 1H, CH 8), 7.66 – 7.60 (m, 1H, CH 18), 7.38 (s, 1H, NH 11), 7.33 – 7.23 (m, 2H, CH 22), 7.27 (s, 1H, NH 9), 7.20 – 7.10 (m, 3H, CH 15/16/17), 7.08 – 6.99 (m, 2H, CH 21), 6.99 (d, J = 7.9 Hz, 1H, CH 4), 6.76 (dd, J = 7.9, 2.0 Hz, 1H, CH 3), 5.33 (dq, J = 37.5, 1.1 Hz, 1H, CH227), 2.26 (s, 3H, CH31), 2.07 (s, 3H, CH36), 1.81 (s, 3H, CH326). 13C NMR (126MHz, Acetone-d6, 20°C), δ (ppm): 156.6 (C 23), 137.8 (C 25), 136.7 (C 5), 136.4 (C 14), 134.9 (C 13), 130.0 (C 20), 129.8 (CH 4), 128.7 (CH 22), 124.5 (C 2), 123.5 (CH 3), 122.5 (CH 17), 122.3 (CH 8), 120.1 (CH 15/16), 119.8 (CH227), 118.7 (CH 18), 115.9 (CH 21), 110.2 (CH 15/16), 21.9 (CH326), 20.3 (CH31), 16.5 (CH36). HRMS (ESI+): m/z calcd for C26H25N3O2Na+ (M+Na)+ 434.1839; found 434.1841. HPLC purity @ λ=254 nm: 98% (General conditions, Rt = 22.28 min). MS (ESI+): m/z (%) 411.8 (100) [M+H+]. EXAMPLE 60: 1-(3,5-difluorophenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea STEP 1 1,3-difluoro-5-isocyanatobenzene (AT1) - According to Typical procedure 5 Bis part 1 using 3,5-difluoroaniline (0.775 mmol, 1.0 equiv., 100 mg) in a solution of DCM (10 mL) under argon atmosphere with triethylamine (1.55 mmol, 2.0 equiv., 0.211 ml). The mixture was cooled to 0-5 °C then triphosgene (0.388 mmol, 0.5 equiv., 115 mg) previously dissolved in DCM (2 mL) was slowly added to the reaction and the mixture was allowed to return at room temperature. The reaction was monitored by TLC and stopped upon complete consumption of the amine derivative (2h). The reaction mixture was concentrated in vacuo, then filtered and washed with Et2O (3 × 10 mL). The organic layer was concentrated in vacuo to afford crude 1,3-difluoro-5-isocyanatobenzene AT1 which was directly reengaged to the next step.
Figure imgf000251_0001
STEP 2 1-(3,5-difluorophenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea (AT2) - According to Typical procedure 5 Bis part 2 using a solution of 4-(4-methoxyphenyl)butan-2-amine W1 (0.775 mmol, 1.0 equiv., 139 mg) in DCM (4 mL) and the freshly prepared 1,3-difluoro-5- isocyanatobenzene AT1 in DCM (2 mL) which was added to the solution. The reaction was putted under argon atmosphere and stirred overnight. Solvent was evaporated and the crude compound was purified with SCC (PE/Et2O – 100:0 to 50:50) to afford 1-(3,5- difluorophenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea AT2, 168 mg, yield: 70%, as a white solid. 1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.28 (s, 1H), 7.25 – 7.13 (m, 2H), 7.13 – 7.05 (m, 2H), 6.84 – 6.77 (m, 2H), 6.50 (tt, J = 9.2, 2.4 Hz, 1H), 5.91 (d, J = 8.3 Hz, 1H), 3.98 – 3.78 (m, 1H), 2.72 – 2.50 (m, 2H), 1.83 – 1.63 (m, 2H), 1.17 (d, J = 6.6 Hz, 3H). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 164.1 (dd, J = 242.5, 15.5 Hz), 158.8, 155.3, 144.3 (t, J = 14.0 Hz), 134.7, 130.0, 114.5, 101.4 (m), 96.7 (t, J = 26.3 Hz), 55.3, 46.1, 39.9, 32.2, 21.6. STEP 3 1-(3,5-difluorophenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea (60) - According to Typical procedure 6 using 1-(3,5-difluorophenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea AT2 (0.239 mmol, 1.0 equiv., 80 mg) as methoxy-protected urea in DCM (4 mL) with BBr3 (1M in DCM, 0.270 mmol, 4.0 equiv., 0.270 mL). The mixture was quenched by adding ice (#2 g / 0.1 mmol), extraction with DCM. Solvents were removed and a purification of the crude residue by SCC (PE/EtOAc – 100:0 to 70:30) to afford 1-(3,5-difluorophenyl)-3-(4-(4- hydroxyphenyl)butan-2-yl)urea 60, 70 mg, yield: 91. A fraction (10 - 20 mg) of the crude mixture was purified by preparative TLC (PE/EtOAc – 70:30, Rf: 0.25) to afford sufficient quantity of pure compound, as a beige solid, for characterization and biological assays.
Figure imgf000252_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.17 (s, 1H, OH 16), 8.03 (s, 1H, NH 5), 7.22 – 7.12 (m, 2H, CH 3), 7.06 – 6.99 (m, 2H, CH 13/14), 6.77 – 6.69 (m, 2H, CH 13/14), 6.49 (tt, J = 9.3, 2.4 Hz, 1H, CH 1), 5.81 (d, J = 8.3 Hz, 1H, NH 7), 3.95 – 3.78 (m, 1H, CH 8), 2.68 – 2.48 (m, 2H, CH211), 1.82 – 1.63 (m, 2H, CH210), 1.17 (d, J = 6.6 Hz, 3H, CH3 9). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 164.1 (dd, J = 242.3, 15.3 Hz, C 2), 156.3 (CO 6), 155.2 (COH 17), 144.5 (t, J = 14.0 Hz, C 4), 133.6 (C 12), 130.0 (CH 14), 116.0 (CH 13), 101.4 (m, CH 3), 96.6 (t, J = 26.3Hz, CH 1), 46.2 (CH 8), 40.0 (CH210), 32.3 (CH2 11), 21.6 (CH39). HRMS (ESI+): m/z calcd for C17H18F2N2O2Na+ (M+Na)+ 343.12285; found 343.12280. HPLC purity @ λ=254 nm: 95% (General conditions, Rt = 19.89 min). MS (ESI+): m/z (%) 321.2 (100) [M+H+]. EXAMPLE 61: 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(3-methyl-5- (trifluoromethyl)phenyl)urea STEP 1 1-isocyanato-3-methyl-5-(trifluoromethyl)benzene AU1 - According to Typical procedure 5 Bis part 1 using 3-methyl-5-(trifluoromethyl)aniline (0.285 mmol, 1.0 eq., 50 mg) in a solution of DCM (10 mL) under argon atmosphere with triethylamine (0.571 mmol, 2.0 eq., 0.077 ml). The mixture was cooled to 0-5 °C then triphosgene (0.143 mmol, 0.5 eq., 43 mg) previously dissolved in DCM (2 mL) was slowly added to the reaction and the mixture was allowed to return at room temperature. The reaction was monitored by TLC and stopped upon complete consumption of the amine derivative (1h). The reaction mixture was concentrated in vacuo, then filtered and washed with Et2O (3×5 mL). The organic layer was concentrated in vacuo to afford 1-isocyanato-3-methyl-5-(trifluoromethyl)benzene AU1 which was directly reengaged to the next step. STEP 2 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(3-methyl-5-(trifluoromethyl)phenyl)urea AU2 - According to Typical procedure 5 Bis part 2 using a solution of 4-(4- methoxyphenyl)butan-2-amine W1 (0.285 mmol, 1.0 equiv., 51 mg) in DCM (4 mL) and the freshly prepared 1-isocyanato-3-methyl-5-(trifluoromethyl)benzene AU1 in DCM (2 mL) which was added to the solution. The reaction was putted under argon atmosphere and stirred overnight. Solvent was evaporated and the crude compound was purified with SCC (PE/Et2O – 100:0 to 60:40) to afford 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(3-methyl-5- (trifluoromethyl)phenyl)urea AU2, 48 mg, yield: 47%, as a yellowish oil. However, purification using SCC did not afford desired product with sufficient purity to obtain a clear 13C NMR spectra. 1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.09 (s, 1H), 7.84 – 7.82 (m, 1H), 7.51 – 7.43 (m, 1H), 7.15 – 7.10 (m, 2H), 7.06 – 7.02 (m, 1H), 6.83 – 6.80 (m, 2H), 5.82 (d, J = 8.3 Hz, 1H), 3.95 – 3.83 (m, 1H), 3.74 (s, 3H), 2.65 – 2.59 (m, 2H), 2.33 (s, 2H), 1.19 – 1.16 (d, J = 6.6 Hz, 3H). STEP 3 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(3-methyl-5-(trifluoromethyl)phenyl)urea (61) - According to Typical procedure 6 using 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(3-methyl- 5-(trifluoromethyl)phenyl)urea AU2 (0.213 mmol, 1.0 equiv., 48 mg) as methoxy-protected urea in DCM (4 mL) with BBr3 (1M in DCM, 0.505 mmol, 4.0 equiv., 0.505 mL) The mixture was quenched by adding ice (#2 g / 0.1 mmol), extraction with DCM. Solvents were removed and a purification of the crude residue by SCC (PE/EtOAc – 100:0 to 70:30), afforded 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(3-methyl-5-(trifluoromethyl)phenyl)urea 61, 37 mg, yield: 79%. A fraction (10 - 20 mg) of the crude mixture was purified by preparative TLC (PE/EtOAc – 70:30, Rf: 0.25) to afford sufficient quantity of pure compound, as a white solid, for characterization and biological assays. 1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.05 (s, 1H, OH 20), 8.02 (s, 1H, NH 9), 7.83 (t, J = 1.9 Hz, 1H, CH 7), 7.44 – 7.42 (m, 1H, CH 4), 7.09 – 6.98 (m, 3H, CH 1 & CH 18), 6.77 – 6.70 (m, 2H, CH 17), 5.75 (d, J = 8.2 Hz, 1H, NH 11), 3.97 – 3.77 (m, 1H, CH 12), 2.70 – 2.48 (m, 2H, CH215), 2.35 (d, J = 0.8 Hz, 3H, CH33), 1.83 – 1.63 (m, 2H, CH2 14), 1.17 (d, J = 6.6 Hz, 3H, CH313). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 156.3 (COH 19), 155.4 (CO 10), 142.4 (C 8), 133.7 (C 16), 140.4 (C 2), 130.1 (CH 18), 125.5 (C 6), 123.7 (C 5), 122.5 (CH 4), 118.9 (m, CH 1), 116.0 (CH 17), 112.4 (CH 7), 46.1 (CH 12), 40.2 (CH214), 32.3 (CH215), 21.7 (CH313), 21.4 (CH33). HRMS (ESI+): m/z calcd for C19H21F3N2O2Na+ (M+Na)+ 389.14473; found 389.14480. HPLC purity @ λ=254 nm: 100% (General conditions, Rt = 18.63 min). MS (ESI+): m/z (%) 367.3 (100) [M+H+]. EXAMPLE 62: 1-(3,5-bis(trifluoromethyl)phenyl)-3-(4-(4-hydroxyphenyl)butan-2- yl)urea STEP 1 1-isocyanato-3,5-bis(trifluoromethyl)benzene (AV1) - According to Typical procedure 5 Bis part 1 using 3,5-bis(trifluoromethyl)aniline (0.436 mmol, 1.0 equiv., 100 mg) in a solution of DCM (10 mL) under argon atmosphere with triethylamine (0.872 mmol, 2.0 equiv., 118 µl). The mixture was cooled to 0-5°C then triphosgene (0.218 mmol, 0.5 equiv., 65 mg) previously dissolved in DCM (2 mL) was slowly added to the reaction and the mixture was allowed to return at room temperature. The reaction was monitored by TLC and stopped upon complete consumption of the amine derivative (1h). The reaction mixture was concentrated in vacuo, then filtered and washed with Et2O (3 × 10 mL). The organic layer was concentrated in vacuo to afford 1-isocyanato-3,5-bis(trifluoromethyl)benzene AV1 which was directly reengaged to the next step.
Figure imgf000255_0001
STEP 2 1-(3,5-bis(trifluoromethyl)phenyl)-3-(4-(4-methoxyphenyl)butan-2-yl)urea (AV2) - According to Typical procedure 5 Bis part 2 using a solution of 4-(4- methoxyphenyl)butan-2-amine W1 (0.436 mmol, 1.0 equiv., 78 mg) in DCM (4 mL) and the freshly prepared 1-isocyanato-3,5-bis(trifluoromethyl)benzene isocyanatobenzene AV1 in DCM (2 mL) which was added to the solution. The reaction was putted under argon atmosphere and stirred overnight. Solvent was evaporated and the crude compound was purified with SCC (PE/Et2O – 100:0 to 60:40) to afford 1-(3,5-bis(trifluoromethyl)phenyl)- 3-(4-(4-methoxyphenyl)butan-2-yl)urea AV2, 94 mg, yield: 53%, as a colorless oil.
Figure imgf000256_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.48 (s, 1H), 8.19 – 8.12 (m, 2H), 7.51 (tq, J = 1.7, 0.8 Hz, 1H), 7.15 – 7.08 (m, 2H), 6.85 – 6.77 (m, 2H), 5.99 (d, J = 8.3 Hz, 1H), 3.96 – 3.86 (m, 1H), 3.74 (s, 3H), 2.74 – 2.52 (m, 2H), 1.87 – 1.67 (m, 2H), 1.20 (d, J = 6.6 Hz, 3H). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 158.9, 155.2, 143.7, 134.8, 132.4 (q, J = 32.9 Hz), 130.0, 126.3, 122.7, 118.4 (d, J = 4.5 Hz), 114.6, 55.4, 46.3, 39.9, 32.2, 21.6. STEP 3 1-(3,5-bis(trifluoromethyl)phenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea (62) - According to Typical procedure 6 using 1-(3,5-bis(trifluoromethyl)phenyl)-3-(4-(4- methoxyphenyl)butan-2-yl)urea AV2 (0.216 mmol, 1.0 equiv., 94 mg) as methoxy-protected urea in DCM (4 mL) with BBr3 (1M in DCM, 0.868 mmol, 4.0 equiv., 0.868 mL) The mixture was quenched by adding ice (#2 g / 0.1 mmol), extraction with DCM. Solvents were removed and a purification of the crude residue (121 mg) by SCC (PE/EtOAc – 100:0 to 70:30) to afford 1-(3,5-bis(trifluoromethyl)phenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea 62, 69 mg, yield: 76%. A fraction (10 - 20 mg) of the crude mixture was purified by preparative TLC (PE/EtOAc – 70:30, Rf: 0.25) to afford sufficient quantity of pure compound, as a colorless oil, for characterization and biological assays. 1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.47 (s, 1H, OH 17), 8.19 – 8.12 (m, 2H, CH 3), 8.07 (s, 1H, NH 6), 7.51 (tt, J = 1.7, 0.8 Hz, 1H, CH 1), 7.08 – 7.00 (m, 2H, CH 15), 6.77 – 6.70 (m, 2H, CH 14), 5.98 (d, J = 8.2 Hz, 1H, NH 8), 3.90 – 3.86 (m, 1H, CH 9), 2.71 – 2.49 (m, 2H, CH212), 1.84 – 1.65 (m, 2H, CH211), 1.19 (d, J = 6.5 Hz, 3H, CH310). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 156.4 (COH 16), 155.1 (CO 7), 143.8 (C 4), 133.6 (C 13), 132.3 (q, J = 32.9 Hz, C 2), 130.0 (CH 15), 124.5 (d, J = 271.9 Hz, C 5), 118.4 (CH 3), 116.0 (CH 14), 114.6(sept, J = 16.1 Hz, CH 1), 46.3 (CH 9), 40.0 (CH211), 32.3 (CH212), 21.6 (CH310). HRMS (ESI+): m/z calcd for C19H18F6N2O2Na+ (M+Na)+ 443.11647; found 443.11650. HPLC purity @ λ=254 nm: 98% (General conditions, Rt = 21.86 min). MS (ESI+): m/z (%) 421.3 (100) [M+H+]. EXAMPLE 63: 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-phenylurea STEP 1 1-(4-(4-methoxyphenyl)butan-2-yl)-3-phenylurea (AW1) - According to Typical procedure 5 Bis part 2 using 1-(3-isocyanatobutyl)-4-methoxybenzene (0.487 mmol, 1.0 equiv., 100 mg) and aniline (0.487 mmol, 1.0 equiv., 45 mg, 44 µL). The crude product was purified by SCC (DCM/MeOH – 100 to 99:1), afforded 1-(4-(4-methoxyphenyl)butan-2-yl)- 3-phenylurea AW1, 96 mg, yield: 66%, as a beige solid. Mp: 132°C.
Figure imgf000257_0001
1H NMR (300MHz, Chloroform-d1, 20°C), δ (ppm): 7.36 – 7.21 (m, 5H), 7.12 – 7.03 (m, 3H), 6.84 – 6.77 (m, 2H), 6.40 (s, 1H), 4.64 (d, J = 8.4 Hz, 1H), 3.92 (dq, J = 8.4, 6.5 Hz, 1H), 3.77 (s, 3H), 2.66 – 2.55 (m, 2H), 1.79 – 1.66 (m, 2H), 1.17 (d, J = 6.5 Hz, 3H). 13C NMR (75MHz, Chloroform-d1, 20°C), δ (ppm): 158.0, 155.4, 138.8, 134.0, 129.5, 129.3, 123.9, 121.2, 114.0, 55.4, 46.2, 39.4, 31.7, 21.7. STEP 2 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-phenylurea (63) - According to Typical procedure 6 using 1-(4-(4-methoxyphenyl)butan-2-yl)-3-phenylurea AW1 (0.084 mmol, 1.0 equiv., 25 mg) as methoxy-protected urea in DCM (2 mL) with BBr3 (1M in DCM, 0.336 mmol, 4.0 equiv., 0.336 mL). The mixture was quenched by adding ice (#2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 63 for characterization and biological assays.
Figure imgf000258_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.03 (s, 1H, OH 16), 7.76 (s, 1H, NH 5), 7.53 – 7.46 (m, 2H, CH 3), 7.26 – 7.18 (m, 2H, CH 2), 7.09 – 6.99 (m, 2H, CH 13), 6.91 (tt, J = 7.3, 1.2 Hz, 1H, CH 1), 6.78 – 6.71 (m, 2H, CH 14), 5.63 (d, J = 8.3 Hz, 1H, NH 7), 3.94 – 3.82 (m, 1H, CH 8), 2.69 – 2.54 (m, 2H, CH211), 1.82 – 1.66 (m, 2H, CH210), 1.18 (d, J = 6.6 Hz, 3H, CH39). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 155.5 (COH 15), 153.4 (CO 6), 140.9 (C 4), 132.8 (C 12), 129.1 (CH 13), 128.5 (CH 2), 121.1 (CH 1), 117.9 (CH 3), 115.1 (CH 14), 45.0 (CH 8), 39.4 (CH210), 31.4 (CH211), 20.9 (CH39). HRMS (ESI+): m/z calcd for C17H20N2O2Na+ (M+Na)+ 307.1417; found 307.1417. HPLC purity @ λ=254 nm: 92% (General conditions, Rt = 17.74 min). MS (ESI+): m/z (%) 285.1 (100) [M+H+]. EXAMPLE 64: 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(m-tolyl)urea STEP 1 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(m-tolyl)urea (AX1) - According to Typical procedure 5 Bis part 2 using 1-(3-isocyanatobutyl)-4-methoxybenzene (0.244 mmol, 1.0 equiv., 50 mg) and m-toluidine (0.244 mmol, 1.0 equiv., 26 mg). The crude product was purified by SCC (DCM/MeOH – 100:0 to 98:2), yielded 1-(4-(4-methoxyphenyl)butan-2- yl)-3-(m-tolyl)urea AX1, 80 mg, yield: 66%. Rf (DCM/MeOH – 98:2): 0.7
Figure imgf000259_0001
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 7.91 (s, 1H), 7.30 – 7.26 (m, 2H), 7.11 – 7.08 (m, 3H), 6.82 – 6.79 (m, 2H), 6.72 – 6.72 (m, 1H), 5.81 (d, J = 8.3 Hz, 1H), 3.93 – 3.84 (m, 1H), 3.73 (s, 3H), 2.64 – 2.57 (m, 2H), 2.23 (s, 3H), 1.75 – 1.66 (m, 2H), 1.16 – 1.14 (d, J = 6.6 Hz, 3H). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 158.8, 156.1, 141.5, 138.9, 134.9, 130.0, 129.3, 123.0, 119.7, 116.2, 114.5, 55.4, 45.9, 40.1, 32.2, 21.8, 21.6. STEP 2 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(m-tolyl)urea (64) - According to Typical procedure 6 using 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(m-tolyl)urea AX1 (0.128 mmol, 1.0 equiv., 40 mg) as methoxy-protected urea in DCM (3 mL) with BBr3 (1M in DCM, 0.512 mmol, 4.0 equiv., 0.512 mL). The mixture was quenched by adding ice (#2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a purification by SCC (PE/acetone – 100:0 to 80:20), afforded the pure 1-(4-(4-methoxyphenyl)butan-2-yl)-3-(m-tolyl)urea 64, 36 mg, yield: 37%. 1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.09 (s, 1H, OH 19), 7.78 (s, 1H, NH 8), 7.31 (m, 1H, CH 6), 7.27 – 7.24 (m, 1H, CH 1), 7.08 (t, J = 7.8 Hz, 1H, CH 2), 7.05 – 6.99 (m, 2H, CH 16), 6.77 – 6.70 (m, 3H, CH 17 & 3), 5.70 (d, J = 8.2 Hz, 1H, NH 10), 3.92 – 3.83 (m, 1H, CH 11), 2.62 – 2.55 (m, 2H, CH214), 2.24 (s, 3H, CH35), 1.96 – 1.66 (m, 2H, CH213), 1.16 (d, J = 6.5 Hz, 3H, CH312). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 156.3 (COH 18), 155.9 (CO 9), 141.5 (CH 1), 138.9 (C 4), 133.7 (CH 2), 130.0 (CH 16), 129.3 (C 15), 123.0 (CH 3), 119.7 (CH 6), 116.2 (C 7), 116.0 (CH 17), 46.0 (CH 11), 40.3 (CH213), 32.3 (CH214), 21.8 (CH312), 21.6 (CH35). HRMS (ESI+): m/z calcd for C18H22N2O2Na+ (M+Na)+ 321.15735; found 321.15740. HPLC purity @ λ=254 nm: 98% (General conditions, Rt = 18.95 min). MS (ESI+): m/z (%) 322.1 (100) [M+H+]. EXAMPLE 65: 1-cyclohexyl-3-(4-(4-hydroxyphenyl)butan-2-yl)urea STEP 1 1-cyclohexyl-3-(4-(4-methoxyphenyl)butan-2-yl)urea (AY1) - According to Typical procedure 5 using 4-(4-methoxyphenyl)butan-2-amine 1 (0.558 mmol, 1.0 equiv., 100 mg) and isocyanatocyclohexane (0.558 mmol, 1.0 equiv., 70 mg). The reaction flask was sealed and heat at 35°C overnight. The solution was evaporated, triturated with DiPE and filtered to afford 1-cyclohexyl-3-(4-(4-methoxyphenyl)butan-2-yl)urea AY1, 130 mg, yield: 77%, as a colorless oil. 1H NMR (300MHz, Chloroform-d1, 20°C), δ (ppm): 7.15 – 7.04 (m, 2H), 6.86 – 6.79 (m, 2H), 3.98 (dd, J = 11.8, 8.2 Hz, 2H), 3.77 (s, 3H), 3.74 – 3.70 (m, 1H), 3.57 – 3.37 (m, 1H), 2.67 – 2.55 (m, 2H), 2.00 – 1.86 (m, 2H), 1.78 – 1.62 (m, 4H), 1.45 – 1.22 (m, 2H), 1.15 (d, J = 6.5 Hz, 3H), 1.15 – 1.01 (m, 2H). 13C NMR (75MHz, Chloroform-d1, 20°C), δ (ppm): 158.1, 157.9, 157.0, 134.1, 129.4, 114.0, 55.4, 49.3, 46.1, 39.6, 34.1, 34.1, 31.7, 25.7, 25.1, 21.8. STEP 2 1-cyclohexyl-3-(4-(4-hydroxyphenyl)butan-2-yl)urea (65) - According to Typical procedure 6 using 1-cyclohexyl-3-(4-(4-methoxyphenyl)butan-2-yl)urea AY1 (0.082 mmol, 1.0 equiv., 25 mg) as methoxy-protected urea in DCM (2 mL) with BBr3 (1M in DCM, 0.328 mmol, 4.0 equiv., 0.328 mL). The mixture was quenched by adding ice (#2 g / 0.1 mmol), extraction with DCM and EtOAc. Solvents were removed and a fraction (10 - 20 mg) of the crude mixture was purified by semi-preparative HPLC to afford sufficient quantity of pure compound 65, as a white solid, for characterization and biological assays.
Figure imgf000261_0001
1H NMR (300MHz, Acetone-d6, 20°C): δ (ppm): 8.14 (s, 1H, OH 16), 7.03 – 7.00 (m, 2H, CH 14), 6.74 – 6.71 (m, 2H, CH 13), 5.19 – 5.16 (m, 2H, NH 5-7), 3.83 – 3.69 (m, 1H, CH 8) 3.56 – 3.44 (m, 1H, CH 4), 2.57 – 2.50 (m, 2H, CH211), 1.88 – 1.83 (m, 2H, CH23), 1.70 – 1.61 (m, 2H, CH210), 1.61 – 1.54 (m, 3H, CH21 & 2), 1.39 – 1.27 (m, 2H, CH22), 1.21 – 1.13 (m, 2H, CH23), 1.10 (m, 1H, CH21), 1.08 (d, J = 6.5 Hz, 3H, CH39). 13C NMR (75MHz, Acetone-d6, 20°C): δ (ppm): 157.9 (CO 6), 156.3 (COH 15), 133.9 (C 12), 130.0 (CH 13), 115.9 (CH 14), 49.2 (CH 4), 45.8 (CH 8), 40.6 (CH210), 34.6 (CH23), 32.3 (CH211), 26.5 (CH21), 25.7 (CH22), 22.0 (CH39). HRMS (ESI+): m/z calcd for C21H28N2O2Na+ (M+Na)+ 313.18865; found 313.18860. HPLC purity @ λ=254 nm: 65%. MS (ESI+): m/z (%) 291.1 (100) [M+H+]. EXAMPLE 66: 1-(benzo[d][1,3]dioxol-5-yl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea STEP 1 5-isocyanatobenzo[d][1,3]dioxole (AZ1) - According to Typical procedure 5 Bis part 1 using benzo[d][1,3]dioxol-5-amine (1.24 mmol, 1.0 equiv., 170 mg) in a solution of DCM (6 mL) under argon atmosphere with triethylamine (4.96 mmol, 4.0 equiv., 0.69 mL). The mixture was cooled to 0-5 °C then triphosgene (1.24 mmol, 1.0 equiv., 368 mg) was added, and the mixture was allowed to return at room temperature. The reaction was monitored by TLC and stopped upon complete consumption of the amine derivative (overnight). The reaction mixture was concentrated in vacuo, then filtered and washed with Et2O (3 × 30 mL). The organic layer was concentrated in vacuo to afford 5-isocyanatobenzo[d][1,3]dioxole AZ1, 160 mg, yield: 79%, with sufficient purity to be used directly in the next step without further purification.
Figure imgf000262_0001
STEP 2 4-(3-(3-(benzo[d][1,3]dioxol-5-yl)ureido)butyl)phenyl acetate (AZ2) - According to Typical procedure 5 Bis part 2 using 5-isocyantobenzo[d][1,3]dioxole AZ1 (0.981 mmol, 1.0 equiv., 160 mg), triethylamine (2.944 mmol, 3.0 equiv., 0.38 mL) and 4-(4- acetoxyphenyl)butan-2-aminium chloride AR2 (0.981 mmol, 1.0 equiv., 203 mg) in DCM (8 mL). The reaction mixture was stirred at room temperature, overnight, monitored by TLC and stopped upon complete consumption of the amine derivative. The resulting mixture was evaporated and the crude product was purified by SCC (PE/Acetone – 100:0 to 70:30), to obtain 4-(3-(3-(benzo[d][1,3]dioxol-5-yl)ureido)butyl)phenyl acetate AZ2, 140 mg, 38%, as a beige solid.
Figure imgf000263_0001
1H NMR (300MHz, Acetone-d6, 20 °C), δ (ppm): 7.71 (s, 1H), 7.29 – 7.23 (m, 3H), 7.03 – 6.97 (m, 2H), 6.74 – 6.68 (m, 2H), 5.90 (s, 2H), 5.61 (d, J = 6 Hz, 1H), 3.90 – 3.86 (m, 1H), 2.76 – 2.60 (m, 2H), 2.23 (s, 3H), 1.81 – 1.69 (m, 2H), 1.16 (d, J = 6.6 Hz, 3H). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 169.7, 155.9, 150.0, 148.5, 143.0, 140.6, 136.2, 130.0, 122.4, 111.5, 108.6, 101.9, 101.7, 46.0, 40.0, 32.5, 21.8, 21.0. STEP 3 1-(benzo[d][1,3]dioxol-5-yl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea (66) - According to Typical procedure 15 using 4-(3-(3-(benzo[d][1,3]dioxol-5-yl)ureido)butyl)phenyl acetate AZ2 (0.379 mmol, 1.0 equiv., 140 mg) in THF (6 mL) at room temperature with LiOH (1.512 mmol, 4.0 equiv., 36 mg) in H2O (2 mL). The reaction was stirred for 24 h. Crude was purified by SCC (PE/Acetone – 100:0 to 70:30), to obtain 1-(benzo[d][1,3]dioxol-5-yl)- 3-(4-(4-hydroxyphenyl)butan-2-yl)urea 66, 120 mg, yield: 96%. A fraction (10 - 20 mg) was purified by semi-preparative HPLC to afford sufficient quantity of pure compound, as a white solid, for characterization and biological assays.
Figure imgf000263_0002
1H NMR (300MHz, Acetone-d6, 20°C), δ (ppm): 8.07 (s, 1H, OH 19), 7.70 (s, 1H, NH 8), 7.30 (dd, J = 1.9, 0.6 Hz, 1H, CH 6), 7.04 – 7.02 (m, 2H, CH 17), 6.76 – 6.70 (m, 2H, CH 16), 6.70 (dd, J = 6.6, 0.6 Hz, 1H, CH 2), 6.69 (dd, J = 6.6, 1.9 Hz, 1H, CH 1), 5.90 (s, 2H, CH24), 5.57 (d, J = 8.2 Hz, 1H, NH 10), 3.91 – 3.77 (m, 1H, CH 11), 2.64 – 2.50 (m, 2H, CH214), 1.75 – 1.65 (m, 2H, CH213), 1.15 (d, J = 6.6 Hz, 3H, CH312). 13C NMR (75MHz, Acetone-d6, 20°C), δ (ppm): 156.3 (CO 9), 155.8 (COH 18), 148.5 (C 5), 142.9 (C 3), 136.3 (C 7), 133.8 (C 15), 130.0 (CH 17), 115.9 (CH 16), 111.4 (CH 1), 108.6 (CH 2), 101.8 (CH24), 101.7 (CH 6), 45.9 (CH 11), 40.3 (CH213), 32.3 (CH214), 21.8 (CH312). HRMS (ESI+): m/z calcd for C18H20N2O4Na+ (M+Na)+ 351.13153; found 351.13190. HPLC purity @ λ=254 nm: 98% (General conditions, Rt = 17.36 min). MS (ESI+): m/z (%) 329.3 (100) [M+H+]. BIOLOGICAL EVALUATION Materials and methods Materials – IRE1 wild-type recombinant protein encoding the cytoplasmic domain (amino acids 465–977) with N-terminal polyhistidine-tag and GST tag was from Sinobiological (Sino Biological Europe GmbH, Eschborn, Germany, #11905-H20B). The fluorescent probe used for the in vitro IRE1 RNase assay was from Eurogentec. Tunicamycin was purchased from Calbiochem (Merck KGaA, Darmstadt, Germany). All the inhibitors were synthesized by and purchased from Enamine (Riga, Latvia). MicroScale Thermophoresis - The direct binding of compound 33 (Z4) and compound 28 (Z4P) to IRE1 protein was measured using MicroScale Thermophoresis (MST). IRE recombinant protein was labelled using RED-Tris-NTA fluorescent dye (RED-Tris-NTA 2nd Generation, NanoTemper, Munich, Germany; # MO-L018). For the labelling step, 100 µL of 20 nM protein solution is mixed with 100 µL of 10 nM RED-Tris-NTA dye in PBST buffer (PBS with 0.05% Tween-20) and incubated for 30 min at RT. The protein–dye mixture was centrifuged for 10 min at 4°C and 15000xg. For measurement of direct binding, the compounds were analyzed in a 16-point dilution series mixed in a 1:1 ratio with the labelled protein in PBST buffer. The assay was performed in standard Monolith NT.115 Capillaries (NanoTemper; #MO-K022), and all measurements were performed at 60% MST power and 60% excitation power using the Monolith NT.115 Pico machine (NanoTemper). The dissociation constant (Kd) was calculated by taking the average of triplicate normalized fluorescence data using NANOTEMPER analysis software (MO.Affinity Analysis v2.3). The normalized values were converted to fraction-bound data, and the resulting binding curves are plotted using GRAPHPAD PRISM software (GraphPad Software). IRE1-mediated in vitro RNase assay – Organic molecules (Compounds 28 (Z4P), and 33 (Z4), 34 (Z4A), 35 (Z4B), 36 (Z4C), 37 (Z4D) and 38 (Z4E)) were diluted in minimal volume of DMSO and subsequently re-diluted in reaction buffer (20 mM HEPES pH 7.5; 1 mM MgOAc; 50 mM KOAc). Maximum volume of DMSO per reaction never exceeded 1%. Reaction volume was 25 μL. Recombinant IRE1 (0.6 μg/ reaction) was incubated at room temperature for 10 minutes with varying concentrations (0-100 μM) of inhibitor and reaction buffer. The assay relied on the use of fluorescence resonance energy transfer (FRET)— quenched mini Xbp1 RNA substrate probe, which when cleaved by IRE1 emits fluorescence at 590 nm (cy3) wavelength (F. Prischi et al., Nature Communications, 2014, 5, 3554). Subsequently equal volume of mixture of reaction buffer, 20 mM ATP, 2 mM DTT and 1 μg of fluorescent probe were added to each sample and fluorescence was read in 96 well plates flat bottom, black polystyrene, matrix active group High Bind (Corning®) every minute for 25 minutes, at 37°C, using a Tecan 200 plate reader. Cell culture and treatments – U87MG (U87; ATCC) cells were authenticated as recommended by AACR (http://aacrjournals.org/content/cell-line-authentication- information) and regularly tested for the absence of mycoplasma using MycoAlert® (Lonza, Basel, Switzerland) or MycoFluor (Invitrogen, Carlsbad, CA, USA). U87 were grown in DMEM Glutamax (Invitrogen, Carlsbad, CA, USA) supplemented with 10% FBS. GB immortalized U251 and primary RADH87 (T. Avril et al., Brain Pathol., 2012, 22, 159–174) cells were grown in DMEM supplemented with 10% FBS in a 5% CO2 humidified atmosphere at 37°C. GB cell lines were modified for IRE1 activity by overexpressing dominant negative (DN) forms of IRE1 that lack the RNase domain (IRE1.NCK or IRE1 Q780stop) as previously described (S. Lhomond et al., EMBO Molecular Medicine, 2018, 10, e7929; J. Obacz et al., bioRxiv, 2020, 533018). For XBP1s induction experiments, tunicamycin was used at 1 μg/mL for the indicated periods of time. For inhibitor cell toxicity assays cells were plated in 96 well plates at 5000 cells per well and treated with 0, 5, 10, 25, 50, 100, 250, 500, 1000 and 2500 μM concentrations of each inhibitor. After 6 days of incubation, WST1 reagent (Roche) was added to each well and post 2-hour incubation the plate was read using a Tecan 200 colorimeter. For TMZ sensitivity assays cells were plated in a 96 well plate at 5000 cells per well and co-treated with 0, 5, 10, 25, 50, 100, 250, 500, 1000 and 2500 μM of TMZ plus a non-toxic dose of inhibitor. After 6 days of incubation, WST1 reagent (Roche) was added to each well and post 2-hour incubation the plate was read using a Tecan 200 colorimeter. Western blotting - All IRE1 signaling analyses were carried out as described previously (S. Lhomond et al., Methods Mol. Biol., 2015, 1292, 177-194). Cells grown on 6 well plates were washed with PBS and lysed with RIPA lysis buffer at 4°C, over 25 minutes to extract protein. IRE1 and phosphorylated IRE1 were stained using anti‐IRE1 antibody (Anti- human; rabbit polyclonal; SantaCruz Biotechnologies, H-190) and pS724-IRE1 antibody (Anti-human; rabbit polyclonal; Abcam, ab48187), respectively. The phosphorylated form of eIF2α was stained with anti-phospho-eIF2α (Ser51) Antibody #9721 (CellSignalling®). Actin was used as a loading control (β-Actin (C4): sc-47778; SantaCruz Biotechnologies). Cell extracts were resolved by SDS-PAGE and transferred to nitrocellulose membranes for 30 minutes using a Trans-Blot® Turbo™ (BioRad® Transfer System #1704150). The resulting membranes were incubated with primary antibodies for 16 hours at 4°C, washed with PBST, and incubated for 1 hour with goat anti‐rabbit or goat anti‐mouse secondary antibodies at room temperature (Invitrogen, Carlsbad, CA, USA) prior revelation using chemiluminescence (ECL RevelBlOt® Intense, Ozyme). Proteomic analyses - Untreated and MKC8866 and compound 33 (Z4)-treated (during 48 hours) parental U251 and RADH87 GB cells as well as DN IRE1 U251 and RADH87 cells were lysed with lysis buffer composed of 20 mM Tris pH 8, 1.5 mM EDTA, 150 mM NaCl, 1% Triton X-100, 0.1% SDS, supplemented with proteases and phosphatases inhibitor cocktails (Roche). Total proteins were precipitated with 80% ice-cold acetone. Washed pellet were then denatured with 8 M urea in Tris-HCl 0.1 mM, reduced with 5 mM TCEP for 30 minutes, and then alkylated with 10 mM iodoacetamide for 30 minutes in the dark. Double digestion was performed with endoproteinase Lys-C (Ref 125-05061, Wako) at a ratio 1/100 (enzyme/proteins) in 8 M urea for 4h, followed by an overnight modified trypsin digestion (Ref V511A, Promega) at a ratio 1/100 (enzyme/proteins) in 2 M urea. Both Lys- C and Trypsin digestions were performed at 37°C. Peptide mixtures were then desalted on C18 spin-column and dried on Speed-Vacuum before LC-MS/MS analysis. Samples were analyzed using an Ultimate 3000 nano-RSLC (Thermo Scientific, San Jose California) coupled in line with a LTQ-Orbitrap ELITE mass spectrometer via a nano-electrospray ionization source (Thermo Scientific, San Jose California). Proteins were identified and quantified by database searching using SequestHT (Thermo Fisher Scientific) with Proteome Discoverer 2.4 software (PD2.4, Thermo Fisher Scientific) against Homo sapiens reviewed SwissProt database. Peptides and proteins were filtered with a false discovery rate (FDR) at 1% and their quantification was based on XIC (Extracted Ion Chromatogram). Proteomic Data analyses and representation - Differential expression analysis was executed to identify the impact of compound 33 (Z4) treatment and IRE1 knock on the proteome, comparing the parental strains of U251 and RADH7 cell lines with the compound 33 (Z4)-treated, dimethyl sulfoxide (DMSO)-treated and DN forms of them. The statistical analysis was based on the non-parametric method of rank products, using the RankProd (R/Bioconductor) package (F. Hong, et al., Bioinformatics, 2006, 22, 2825–2827). Its RP and topGene functions were utilized to identify the most significantly over and under- expressed proteins, setting the p-value threshold at 0.05. Differentially expressed proteins in the wild type – compound 33 (Z4)-treatment comparison with absolute log fold change value lower than that of wild type - DMSO comparison, were filtered out, as their regulation cannot be exclusively attributed to the effect of compound 33 (Z4) administration. BioInfoMiner (T. Koutsandreas, et al., Int. J. Monit. Surveill. Technol. Res., 2016, 4, 30–49) was used for the functional interpretation of the sets of significantly perturbed proteins and the comparative analysis of the derived semantic networks. Pathway analysis was performed, exploiting the annotation of Gene Ontology (Biological Process domain – GO-BP) (Nucleic Acids Research, 2021, 49, D325–D334), with hyper-geometric and adjusted p-value thresholds at 0.05. A network of statistically important GO-BP terms was identified for each list of proteins. Then, all significantly over-represented terms were clustered, using the Resnik semantic similarity measure (P. Resnik, Journal of Artificial Intelligence Research, 1999, 11, 95–130) In each iteration of the agglomerative clustering process, the most similar pair of terms was substituted with its most informative common ancestor (mica). Semantic similarity threshold was set to 0.25, as lower values might produce semantic clusters, with overly generic semantic description. Examining the membership of each GO-BP cluster and the initial lists of over-represented terms, a binary association matrix was constructed, to reveal and compare the membership profiles of the regulated sets of proteins in cellular functionality. Tumor cell orthotopic implantation - Tumor cells (U87-Luc (A. Jabouille, et al., Oncotarget, 2015, 6, 24922–24934)) were implanted into the brain of immunodeficient NMRI-Foxn1nu/Foxn1nu, 8 weeks old male mice (Janvier Laboratories, Laval, France). All animal procedures met the European Community Directive guidelines (Agreement B35-238- 40 Biosit Rennes, France/ No DIR 13480) and were approved by the local (University of Rennes) ethics committee and ensuring the breeding and the daily monitoring of the animals in the best conditions of wellbeing according to the law and the rule of 3R (Reduce-Refine- Replace). U87-Luc cells were implanted in the mouse brain by intracerebral injection followed by tumor growth monitoring using bioluminescence (PhotonIMAGER™ systems, BIOSPACE LAB). The mice were anesthetized intraperitoneally (i.p. mix Xylazine 10mg/kg and Ketamin 60 mg/kg) and then fixed on a stereotactic frame. This framework makes it possible to manipulate the brains of living animals, and to reach isolated areas of the brain precisely relative to markings visible to the naked eye through the use of three- dimensional coordinates. After incising the scalp, the stereotaxic coordinates were calculated for injection of tumor cells into a specific point of the brain, and reproducible for all the mice used. In the study, the tumor cells (5 × 104 cells per mice in 1 μL) were injected at Bregma 0, 2.2 mm to the left of the bregma and 3.2 mm deep to perform the implantation at the level of the striatum. Mouse treatments – Four days after tumor cells implantation, compound 28 (Z4P) treatments were started consisting in treatments of 300 µg/kg/day intraperitonially. For the RNA quantification experiment, performed on tumors generated in mice, three mice per group were treated according to the methods above. Mice were sacrificed at day 34 and the brains were removed and extract the tumor. The samples were used to extract RNA for the analysis, as described below. For the survival experiment, performed on tumors generated in mice, co-treatment with TMZ was performed one week after the beginning of compound 33 (Z4) treatment (n=7 per group for the TMZ and COMBO, n=8 per group for the control and compound 28 (Z4P)). The animals were injected with 10mg/kg/day of TMZ delivered intraperitoneally for ten days. All the treatments were performed with 2 days of rest each week. Survival was measured as the time between implantation and sacrifice, which was performed by cervical dislocation in case of critical clinical signs or loss of weight >10%. Bioluminescence monitoring - Mice were evaluated with in vivo bioluminescence imaging every 3 days starting from the day 4, for the first week, and weekly onward for tumor progression and followed for signs of neurologic deterioration daily. Mice were injected i.p. with 100 μL of luciferin (Promega, Charbonnières-les-Bains, France). The luciferin was allowed to circulate for 5 min before the mice were anesthetized with a mix of O2 and isoflurane (2.5%). Quantitative real‐time PCR - Total RNA was prepared using TRIzol reagent (Invitrogen, Carlsbad, CA, USA). All RNAs were reverse‐transcribed with Maxima Reverse Transcriptase (Thermo Scientific, Waltham, MA, USA), according to manufacturer protocol. qPCR was performed via a StepOnePlus™ Real‐Time PCR Systems from Applied Biosystems and the SYBR Green PCR Core reagents kit (Takara). Analysis was carried out using QuantStudioTM Design and Analysis software version 1.3.1. Three technical repeats were performed per experiment and at least three biological repeats were performed per point per experiment. Each sample was extract individually for RNA and performed the quantification of mRNA levels by qPCR for several targets. These data were then plotted using Morpheus’ tool from the Broad institute (https://software.broadinstitute.org/morpheus). Statistical analyses - Data are presented as mean ± SEM or ± SD as indicated in each figure. Statistical significance (P < 0.05 or less) was determined using unpaired t‐tests or ANOVA as appropriate and performed using GraphPad Prism software (GraphPad Software, San Diego, CA, USA). Curve’s extrapolations were performed using curve fit hypotheses by GraphPad Prism software (GraphPad Software, San Diego, CA, USA). Luciferase assays - Cells were seeded in 96-well plate. Cells were treated with increasing concentration of Z4 and analogues. Medium was discarded and the plate was tapped to remove residual medium. 100 µL of lysis buffer (25 mM Tricine pH 7.8; 15 mM Potassium Phosphate pH 7.8; 15 mM MgSO4; 4 mM EGTA; 1% Triton X-100; 1 mM DTT) were added per well and incubated for 20 minutes at RT.50 µL were transferred to a white 96-well plate and 50 µL of substrate buffer (25 mM Tricine pH 7.8; 15 mM Potassium Phosphate pH 7.8; 15 mM MgSO4; 4 mM EGTA; 1% Triton X-100; 1 mM DTT; 1 mM ATP; 0.2 mM luciferin) were added in each well and the luminescence was read. Results IRE1-fragment-derived organic molecules dock onto the ATP binding pocket of IRE1 in vitro To monitor the interaction between IRE1 and the compounds, micro-scale thermophoresis (MST) was used with recombinant IRE1 and increasing concentrations of compound 33 (Z4) and compound 28 (Z4P) (racemic mixtures) and the pure compound 28 (Z4P) enantiomers. It is shown that IRE1 interacts with both compound 33 (Z4) and compound 28 (Z4P) following a saturation one-site binding curve exhibiting respective KD of ~20 and of ~120 µM, respectively (data not shown). Since compound 28 (Z4P) possesses a stereocenter, we next tested if both enantiomers would bind in the ATP-binding pocket of IRE1 and if they both do, which of them have a better affinity for the target. To this end, the racemic compound was synthesized and the enantiomers separated by chiral chromatography. Both enantiomers of compound 28, named S-28 (Z4P (S)) and R-28 (Z4P (R)), were subjected to the same MST assay and were found to efficiently associate with IRE1 following a one-site saturation binding curves and with a marginal 2-fold difference in KD, with ~60 and ~29 µM, respectively (data not shown). Additionally, compounds 28 (Z4P), 33 (Z4), 34 (Z4A), 35 (Z4B), 36 (Z4C), 37 (Z4D) and 38 (Z4E) were tested for inhibitory activity using a fluorescence resonance energy transfer (FRET)—quenched IRE1 RNA substrate probe and yielded IC50s in the low micromolar range (Figure 1). IC50s of compound S-28 and R-28 (Z4P (S) and Z4P (R)) were found to differ only by a 3-fold factor, highlighting again the rather unimportant nature of the stereocenter (data not shown). Compounds of the invention modulate IRE1 activity in vitro and in cellular models of GB It was investigated whether compounds 28 (Z4P) and 33 (Z4) could affect IRE1 activity in GB cells. Their effect on IRE1 phosphorylation was first documented. Indeed, upon treatment with these compounds IRE1 phosphorylation was reduced by about 25% (Figure 2A). Since kinase interference may mediate RNase effects (W. Tirasophon et al., Genes Dev., 1998, 12, 1812-1824) and since IRE1 has two distinct functional outputs through its RNase domain (XBP1 splicing and RIDD), the effect of compounds 28 (Z4P) and 33 (Z4) on Xbp1 mRNA splicing and RIDD mediated RNA decay was investigated. Compounds 28 (Z4P), and 33 (Z4) inhibited Xbp1 mRNA splicing in U87 cells in the presence of the ER stressor tunicamycin (Figure 2B). The effect of compounds 28 (Z4P) and 33 (Z4) on RIDD was then tested by interrogating the levels of the known RIDD target Sparc mRNA (46) in U87 cells upon treatment with the transcriptional blocker actinomycin D and the ER stressor tunicamycin. Both compound 33 (Z4) and 28 (Z4P) blocked tunicamycin-induced degradation of Sparc mRNA compared to control treatment (Figure 2C) whilst had no effect on Sparc mRNA in cells overexpressing a dominant negative (DN) form of IRE1 (data not shown). The specificity of these compounds in the context of the other arms of the UPR, PERK in particular, was then investigated. This was done to ensure that the stress responses related to compounds 28 (Z4P) and 33 (Z4) were wholly due to their action on IRE1 rather than other UPR transducers. The expression of downstream effectors of PERK and ATF6 was evaluated. Neither Chop nor Herpud1 mRNA were significantly affected by compound 33 (Z4) treatment (data not shown). This was also confirmed at the level of eIF2α phosphorylation and ATF4 protein expression which were not altered upon exposure to compound 33 (Z4) (data not shown). These experiments gave confidence that compounds 28 (Z4P), 33 (Z4), 34 (Z4A), 35 (Z4B), 36 (Z4C), 37 (Z4D) and 38 (Z4E) did not significantly impact the other UPR branches. Consequently, to further document the characteristics of compound 33 (Z4) at the protein level, U251 (commonly available GB cell line) and RADH87 (G. Auf et al., Proc Natl Acad Sci U S A, 2010, 107, 15553–15558) (human tumor-derived primary GB cell line) cells were treated and their proteome were evaluated. Treatment of U251 and RADH87 cells with compound 33 (Z4) impacted on particular pathways such as cell adhesion, circulatory system development, cell locomotion and cell migration; all properties involved in tumor establishment (e.g. angiogenesis) and tumor invasion (e.g. metastasis) (data not shown). The proteomes of these GB lines exposed to compound 33 (Z4) was then functionally compared to those of the same cell lines in which IRE1 activity was invalidated genetically either using a dominant negative construct reported previously (B. Drogat, et al., Cancer Res., 2007, 67, 6700–6707) or a truncated mutant variant of IRE1 lacking an RNase domain (Q780stop) (S. Lhomond, et al., EMBO Molecular Medicine, 2018, 10, e7929) (data not shown). Genes that were down- or up-regulated were compared according to the cell lines and conditions. Proteome alterations upon compound 33 (Z4) exposure phenocopied those observed in cells in which IRE1 activity was genetically blunted, thus reinforcing the selectivity of compound 33 (Z4) towards IRE1 (data not shown). Having extensively documented the effect and specificity of Z4-compounds on IRE1, it was then tested whether compounds 28 (Z4P), 33 (Z4), 34 (Z4A), 35 (Z4B), 36 (Z4C), 37 (Z4D) and 38 (Z4E) could sensitize GB cells to the standard of care chemotherapy, TMZ. Cell viability assays were carried out on U87 cells treated with sub-toxic doses of compound 33 (Z4) and compound 28 (Z4P) (data not shown) and escalating doses of TMZ. Both compound 33 (Z4) and compound 28 (Z4P) produced similar effects, namely to significantly sensitize the cells to TMZ by about 1.5-fold (Figures 2D and 3). The results show that compound 28 (Z4P) (i) binds IRE1 in vitro; (ii) inhibits IRE1 activity in vitro and in cellular models; (iii) sensitizes GB cells to TMZ and; (iv) is predicted to cross the BBB. Combination therapy of compound 28 (Z4P) and TMZ against in vivo models of GB arrests tumor growth and prolongs relapse-free survival To characterize compound 28 (Z4P) in vivo, murine models growing orthotopic tumors generated by injection of U87-luc cells were utilized (25). First, intraperitoneal (IP) escalating doses of compound 28 (Z4P) (50 µg/kg to 300 µg/kg) were administered to determine compound 28 (Z4P) toxicity in vivo. This was done in WT mice and no toxicity was detected in any of the concentrations tested (Figure 4A). Since compound 28 (Z4P) displayed zero toxicity in WT mice, it was then sought to (i) characterize the effect of compound 28 (Z4P) on IRE1 biology in an in vivo tumor; (ii) test whether compound 28 (Z4P) was able to cross the BBB and reach tumors cells; (iii) determine if co-treatment of compound 28 (Z4P) alongside SOC chemotherapy TMZ conferred any anti-tumorigenic, anti-relapse or pro-survival advantages. To do so, U87-luc cells were orthotopically implanted in the mouse brain and the tumor was allowed to grow. At day 4 post implantation, a small tumor formation was detected using bioluminescence (data not shown). Thereafter, mice were randomized in different groups and compound 28 (Z4P) daily treatments (i.p.300 µg/kg) were administered for 34 consecutive days (Figure 5A). Upon completion of this time period, mouse brains were entirely excised and dissected. The corresponding tumors were resected, dissociated and RNA was extracted from them. The splicing of Xbp1 mRNA was evaluated in control tumors and in animals treated with compound 28 (Z4P). Treatment with compound 28 (Z4P) decreased the splicing of Xbp1 mRNA thus demonstrating that compound 28 (Z4P) mediates IRE1 inhibition in vivo when administered IP, also implying that it crossed the BBB (Figure 5B). Using the same mRNA samples, the expression levels of other GB invasion, inflammation and UPR markers using RT-qPCR were also evaluated (data not shown). A remarkable decrease of mRNA levels of genes related to inflammation (Il-6 and Il-8) whilst no expression difference could be seen in stemness genes (Cd133, Olig2, Sox2, Cd44 and Sall2) was observed. Vegfa mRNA, one of the genes related to vascular formation, had its levels decreased under compound 28 (Z4P) treatment while Vimentin has a clear upregulation on Z4P treated group, results that were consistent with what was observed in U87 cells in which IRE1 activity was genetically invalidated (B. Drogat, et al., Cancer Res., 2007, 67, 6700–6707; G. Auf et al., Proc Natl Acad Sci U S A, 2010, 107, 15553–15558). The expression levels of E-cadherin (Cdh1) mRNA remained unchanged upon compound 28 (Z4P) treatment. Lastly, apart from Bip, mRNA expression of the other ER stress markers tested (Chop, Herpud1 and Perk) decreased upon compound 28 (Z4P) treatment. The latest results, although not identical to those obtained in cellular models, might reflect a more complex mode of action of compound 28 (Z4P) than expected. Finally, the potential of compound 28 (Z4P) as monotherapy or neo-adjuvant therapy alongside TMZ in a clinically relevant, surgical model of GB was investigated. Utilizing our compound 28 (Z4P)-treated, orthotopic mouse model (i.p. 300 μg/kg daily, starting on D4), we administered 10 daily treatments of TMZ (10 mg/kg; Figure 5C) after day 11-post tumor cell implantation. Whilst compound 28 (Z4P) treatment alone had no effect on tumor growth or mouse survival (Figure 4B and 4C), co-treatment with TMZ significantly decreased tumor growth. This effect was observed as early as during the first week of treatment (Figure 5D). One of the aggressive hallmarks of clinical GB is the very high probability of tumor recurrence despite treatment with TMZ. To this end, it was investigated whether co- treatment of compound 28 (Z4P) and TMZ would have an effect on tumor recurrence. Over 200 days, expectedly, mice treated with TMZ alone relapsed (Figure 5D, black curve) forming new tumors; a result consistent with what is observed in clinic. In contrast, no tumor relapse was observed in the group of animals treated with both TMZ and compound 28 (Z4P) (Figure 5D, grey curve). In conclusion, compound 28 (Z4P), administered intraperitoneally, sensitizes orthotopically implanted GB tumors to TMZ in mice. It prevents tumor relapse when used as an adjuvant therapy alongside TMZ. This has major translational clinical implications as GB tumor relapse is the major disease evolution characteristic causing death in human patients. IRE1-mediated in vitro RNase assay and Luciferase assay Results of the IRE1-mediated in vitro RNase assay and Luciferase assay for the compounds of the invention are listed in the Table 5 below: Table 5
Figure imgf000274_0001
Figure imgf000275_0001
Figure imgf000276_0001
Figure imgf000277_0001
Figure imgf000278_0001

Claims

CLAIMS 1. A compound of Formula I:
Figure imgf000279_0001
a pharmaceutically acceptable salt or a solvate thereof, wherein A is selected from –NH–, –N(Me)– and –O–;
Figure imgf000279_0002
, Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O–, –CH2– and –C(O)NH– or is a single bond; Z is O or S; R1 and R2 are independently selected from H and OH, with the proviso that at least one of R1 and R2 is H and that R1 and R2 are not both H; Cy is selected from: -
Figure imgf000280_0001
R3 is selected from H, C1-C4-alkyl and halogen; R4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; R5 is H or C1-C4-alkyl; R6 is selected from H, C1-C4-alkyl, C3-C4-cycloalkyl, halogen, C(O)NR8R9, NHC(O)R10 and C(O)OR11 wherein R8 are R9 are independently selected from hydrogen, C1-C4-alkyl and C3- C4-cycloalkyl; R9 is C1-C4-alkyl; and R10 is selected from H and C1-C4-alkyl; and R7 is H or C1-C4-alkyl; with the proviso that R3, R4, R5, R6 and R7, are not all H; -
Figure imgf000281_0001
wherein R11 and R12 are independently selected from hydrogen and C1-C4-alkyl; - - - -
Figure imgf000281_0002
- with the proviso that, when
Figure imgf000282_0001
are not all H, and the compound of Formula I is none of the following: (R)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; (S)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N,4-dimethylbenzamide; (R)-4-fluoro-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)benzamide; (S)-N-ethyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; (S)-N-cyclopropyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; (S)-N1-(5-carbamoyl-2-fluorophenyl)-N2-(4-(4-hydroxyphenyl)butan-2-yl)oxalamide; (R)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N,4-dimethylbenzamide; (S)-N-(3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylphenyl)acetamide; (S)-1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(5-iodo-2-methylphenyl)urea; (S)-N-(3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylphenyl)propionamide; (S)-3-acetamido-N-(4-(4-hydroxyphenyl)butan-2-yl)-4-methylbenzamide; (S)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)benzamide; (R)-N-ethyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; Methyl (S)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzoate; (R)-N-(3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylphenyl)isobutyramide; (S)-1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; (S)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N,N,4-trimethylbenzamide; (S)-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; (R)-N-(3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylphenyl)acetamide; (R)-N1-(5-carbamoyl-2-methylphenyl)-N2-(4-(4-hydroxyphenyl)butan-2- yl)oxalamide; N-ethyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; 3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N,4-dimethylbenzamide; 3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; 3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N,2-dimethylbenzamide; 4-fluoro-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)benzamide; 4-fluoro-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N-methylbenzamide; N-cyclopropyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; N-(3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)phenyl)isobutyramide; N-(5-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-2-methylphenyl)acetamide; (S)-1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(m-tolyl)urea; 1-(2,5-dimethylphenyl)-3-(4-phenylbutan-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-hydroxyphenethyl)urea; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(2-fluorophenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(2-ethyl-6-methylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; N-(2,5-dimethylphenyl)-4-(3-hydroxyphenyl)piperazine-1-carboxamide; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-phenylurea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(m-tolyl)urea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(p-tolyl)urea; 1-cyclohexyl-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; and 1-(benzo[d][1,3]dioxol-5-yl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea. 2. The compound according to claim 1, wherein Z is O. 3. The compound according to claim 1 or 2, wherein R1 is OH and R2 is H. 4. The compound according to claim 1, having the formula VII: VII, or a pharmaceutically acceptable salt or solvate thereof, wherein R1, R2, L, A, Z, Y, R3, R4, R5, R6 and R7 are as defined in claim 1. 5. The compound according to claim 1, having the formula IX:
Figure imgf000284_0001
IX, or a pharmaceutically acceptable salt or solvate thereof, wherein L, A, Y, R3, R4, R5, R6 and R7 are as defined in claim 1. 6. The compound according to claim 1, selected from the group consisting of: 1-(2,5-dimethylphenyl)-3-(4-hydroxybenzyl)urea; 1-(2,5-dimethylbenzyl)-3-(4-hydroxybenzyl)urea; 1-(2,5-dimethylbenzyl)-3-(4-hydroxyphenethyl)urea; 1-(2,5-dimethylphenyl)-3-(3-(4-hydroxyphenyl)propyl)urea; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)-2-methylbutan-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)but-3-yn-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(3-(4-hydroxyphenyl)prop-2-yn-1-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)-2-methylbut-3-yn-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)-1H-indol-3-yl)urea; N-(2,5-dimethylphenyl)-4-(4-hydroxyphenyl)piperazine-1-carboxamide; 1-(2,5-dimethylphenyl)-3-(3'-hydroxy-[1,1'-biphenyl]-4-yl)urea; 1-(2,5-dimethylphenyl)-3-(4'-hydroxy-[1,1'-biphenyl]-4-yl)urea; 1-(2,5-dimethylphenyl)-3-(4'-hydroxy-[1,1'-biphenyl]-3-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)cyclohexyl)urea; 1-(2,5-dimethylphenyl)-3-(1-(3-hydroxyphenyl)piperidin-3-yl)urea; 3-(2,5-dimethylphenyl)-1-(4-(4-hydroxyphenyl)butan-2-yl)-1-methylurea; 2-(2,5-dimethylphenyl)-N-(4-(4-hydroxyphenyl)butan-2-yl)acetamide; 2,5-dimethylphenyl (4-(4-hydroxyphenyl)butan-2-yl)carbamate; 4-(4-hydroxyphenyl)butan-2-yl 2-(2,5-dimethylphenyl)acetate; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)thiourea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(o-tolyl)urea; 1-(3,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(3-isopropylphenyl)urea; 1-(benzo[d]thiazol-6-yl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(quinolin-7-yl)urea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(2-methylnaphthalen-1-yl)urea; 1-(2-(tert-butyl)phenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(2,5-dimethylbenzyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(3-(3-hydroxyphenyl)prop-2-yn-1-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-(3-hydroxyphenyl)but-3-yn-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-(3-hydroxyphenyl)-2-methylbut-3-yn-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(6-(4-hydroxyphenyl)pyridin-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(3-(4-hydroxyphenyl)cyclohexyl)urea; 1-(2,5-dimethylphenyl)-3-(1-(3-hydroxyphenyl)piperidin-4-yl)urea; 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)piperidin-4-yl)urea; 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)piperidin-3-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)-1-methylurea; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)-1,3-dimethylurea; 4-(4-hydroxyphenyl)butan-2-yl (2,5-dimethylphenyl)carbamate; 1-(2,4-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(3-cyclopropylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(5-isopropyl-2-methylphenyl)urea; 1-(2-ethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(3-fluoro-5-methylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; and 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(1H-indol-6-yl)urea. 7. A pharmaceutical composition comprising a compound Formula I:
Figure imgf000286_0001
I, or a pharmaceutically acceptable salt or solvate thereof and at least one pharmaceutically acceptable carrier, diluent, excipient and/or adjuvant, wherein A is selected from –NH–, –N(Me)– and –O–; Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O–, –CH2– and –C(O)NH– or is a single bond; Z is O or S; R1 and R2 are independently selected from H, OH and OMe, with the proviso that at least one of R1 and R2 is H; Cy is selected from: - R3 is selected from H, C1-C4-alkyl, and halogen; R4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; R5 is H or C1-C4-alkyl; R6 is selected from H, C1-C4-alkyl, C3-C4-cycloalkyl, halogen, C(O)NR8R9, NHC(O)R10 and C(O)OR11 wherein R8 are R9 are independently selected from hydrogen, C1-C4-alkyl and C3- C4-cycloalkyl; R9 is C1-C4-alkyl; and R10 is selected from H and C1-C4-alkyl; and R7 is H or C1-C4-alkyl; -
Figure imgf000288_0001
wherein R11 and R12 are independently selected from hydrogen and C1-C4-alkyl; - -
Figure imgf000288_0002
- - - 8. A compound of Formula I:
Figure imgf000289_0001
a pharmaceutically acceptable salt or a solvate thereof, wherein A is selected from –NH–, –N(Me)– and –O–; Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O–, –CH2– and –C(O)NH– or is a single bond; Z is O or S; R1 and R2 are independently selected from H, OH and OMe, with the proviso that at least one of R1 and R2 is H; Cy is selected from: - R3 is selected from H, C1-C4-alkyl, and halogen; R4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; R5 is H or C1-C4-alkyl; R6 is selected from H, C1-C4-alkyl, C3-C4-cycloalkyl, halogen, C(O)NR8R9, NHC(O)R10 and C(O)OR11 wherein R8 are R9 are independently selected from hydrogen, C1-C4-alkyl and C3- C4-cycloalkyl; R9 is C1-C4-alkyl; and R10 is selected from H and C1-C4-alkyl; and R7 is H or C1-C4-alkyl; -
Figure imgf000291_0001
wherein R11 and R12 are independently selected from hydrogen and C1-C4-alkyl; - -
Figure imgf000291_0002
- - - in combination with an anticancer agent for use in treating cancer. 9. A compound of Formula I:
Figure imgf000292_0001
I, a pharmaceutically acceptable salt or a solvate thereof, wherein A is selected from –NH–, –N(Me)– and –O–; Y is selected from –NH–, –N(Me)–, –NH–CH2–, –O–, –CH2– and –C(O)NH– or is a single bond; Z is O or S; R1 and R2 are independently selected from H, OH and OMe, with the proviso that at least one of R1 and R2 is H; Cy is selected from: - R3 is selected from H, C1-C4-alkyl, and halogen; R4 is selected from H, C1-C4-alkyl and C3-C4-cycloalkyl; R5 is H or C1-C4-alkyl; R6 is selected from H, C1-C4-alkyl, C3-C4-cycloalkyl, halogen, C(O)NR8R9, NHC(O)R10 and C(O)OR11 wherein R8 are R9 are independently selected from hydrogen, C1-C4-alkyl and C3- C4-cycloalkyl; R9 is C1-C4-alkyl; and R10 is selected from H and C1-C4-alkyl; and R7 is H or C1-C4-alkyl; -
Figure imgf000294_0001
wherein R11 and R12 are independently selected from hydrogen and C1-C4-alkyl; - -
Figure imgf000294_0002
- - - for use in increasing the sensitivity of cancer cells to an anticancer agent in a treatment of cancer. 10. The compound for use according to claim 8 or 9, wherein Z is O. 11. The compound for use according to claim 8 or 9, wherein R1 is OH and R2 is H. 12. The compound for use according to claim 8 or 9, wherein the compound is selected from the group consisting of: 1-(2,5-dimethylphenyl)-3-(4-hydroxybenzyl)urea; 1-(2,5-dimethylbenzyl)-3-(4-hydroxybenzyl)urea; 1-(2,5-dimethylbenzyl)-3-(4-hydroxyphenethyl)urea; 1-(2,5-dimethylphenyl)-3-(3-(4-hydroxyphenyl)propyl)urea; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)-2-methylbutan-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)but-3-yn-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(3-(4-hydroxyphenyl)prop-2-yn-1-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)-2-methylbut-3-yn-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)-1H-indol-3-yl)urea; N-(2,5-dimethylphenyl)-4-(4-hydroxyphenyl)piperazine-1-carboxamide; 1-(2,5-dimethylphenyl)-3-(3'-hydroxy-[1,1'-biphenyl]-4-yl)urea; 1-(2,5-dimethylphenyl)-3-(4'-hydroxy-[1,1'-biphenyl]-4-yl)urea; 1-(2,5-dimethylphenyl)-3-(4'-hydroxy-[1,1'-biphenyl]-3-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)cyclohexyl)urea; 1-(2,5-dimethylphenyl)-3-(1-(3-hydroxyphenyl)piperidin-3-yl)urea; 3-(2,5-dimethylphenyl)-1-(4-(4-hydroxyphenyl)butan-2-yl)-1-methylurea; 2-(2,5-dimethylphenyl)-N-(4-(4-hydroxyphenyl)butan-2-yl)acetamide; 2,5-dimethylphenyl (4-(4-hydroxyphenyl)butan-2-yl)carbamate; 4-(4-hydroxyphenyl)butan-2-yl 2-(2,5-dimethylphenyl)acetate; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)thiourea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(o-tolyl)urea; 1-(3,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(3-isopropylphenyl)urea; 1-(benzo[d]thiazol-6-yl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(quinolin-7-yl)urea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(2-methylnaphthalen-1-yl)urea; 1-(2-(tert-butyl)phenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; (S)-1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; (R)-1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-phenylbutan-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-hydroxyphenethyl)urea; N-(2,5-dimethylphenyl)-4-(3-hydroxyphenyl)piperazine-1-carboxamide; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(p-tolyl)urea; 3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; 3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-N,4-dimethylbenzamide; 4-fluoro-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)benzamide; N-ethyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; N-cyclopropyl-3-(3-(4-(4-hydroxyphenyl)butan-2-yl)ureido)-4-methylbenzamide; N1-(5-carbamoyl-2-fluorophenyl)-N2-(4-(4-hydroxyphenyl)butan-2-yl)oxalamide; 1-(2,5-dimethylbenzyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(3-(3-hydroxyphenyl)prop-2-yn-1-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-(3-hydroxyphenyl)but-3-yn-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-(3-hydroxyphenyl)-2-methylbut-3-yn-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(6-(4-hydroxyphenyl)pyridin-2-yl)urea; 1-(2,5-dimethylphenyl)-3-(3-(4-hydroxyphenyl)cyclohexyl)urea; 1-(2,5-dimethylphenyl)-3-(1-(3-hydroxyphenyl)piperidin-4-yl)urea; 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)piperidin-4-yl)urea; 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)piperidin-3-yl)urea; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)-1-methylurea; 1-(2,5-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)-1,3-dimethylurea; 4-(4-hydroxyphenyl)butan-2-yl (2,5-dimethylphenyl)carbamate; 1-(2,4-dimethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(3-cyclopropylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(5-isopropyl-2-methylphenyl)urea; 1-(2-ethylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(3-fluoro-5-methylphenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(1H-indol-6-yl)urea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-phenylurea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(m-tolyl)urea; 1-cyclohexyl-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; and 1-(benzo[d][1,3]dioxol-5-yl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea. 13. The compound for use according to any one of claims 8 to 12, wherein the anticancer agent is an alkylating agent. 14. The compound for use according to claim 13, wherein the alkylating agent is temozolomide. 15. The compound for use according to any one of claims 8 to 14, wherein the cancer is selected from glioblastoma triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer. 16. A hydroxyphenyl compound selected from the group consisting of: 1-(2,5-dimethylphenyl)-3-(6-(3-hydroxyphenyl)pyridin-3-yl)urea; 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)-1H-pyrrol-3-yl)urea; 1-(2,5-dimethylphenyl)-3-(1-(4-hydroxyphenyl)-2-(prop-1-en-2-yl)-1H-indol-3- yl)urea; 1-(3,5-difluorophenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; 1-(4-(4-hydroxyphenyl)butan-2-yl)-3-(3-methyl-5-(trifluoromethyl)phenyl)urea; and 1-(3,5-bis(trifluoromethyl)phenyl)-3-(4-(4-hydroxyphenyl)butan-2-yl)urea; or a pharmaceutically acceptable salt or solvate thereof. 17. A pharmaceutical composition comprising a hydroxyphenyl compound according to claim 16 or a pharmaceutically acceptable salt or solvate thereof and at least one pharmaceutically acceptable carrier, diluent, excipient and/or adjuvant. 18. The hydroxyphenyl compound according to claim 16, in combination with an anticancer agent, for use in treating cancer. 19. The hydroxyphenyl compound according to claim 16, for use in increasing the sensitivity of cancer cells to an anticancer agent in a treatment of cancer. 20. The hydroxyphenyl compound for use according to claim 18 or 19, wherein the anticancer agent is an alkylating agent. 21. The hydroxyphenyl compound for use according to claim 20, wherein the alkylating agent is temozolomide. 22. The hydroxyphenyl compound for use according to any one of claims 18 to 21, wherein the cancer is selected from glioblastoma triple-negative breast cancer, lung cancer, osteosarcoma, prostate cancer, ovarian cancer and pancreatic cancer.
PCT/EP2023/050228 2022-01-07 2023-01-06 Compounds containing a hydroxyphenyl moiety and their use WO2023131677A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP22305014 2022-01-07
EP22305014.7 2022-01-07

Publications (1)

Publication Number Publication Date
WO2023131677A1 true WO2023131677A1 (en) 2023-07-13

Family

ID=80113278

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2023/050228 WO2023131677A1 (en) 2022-01-07 2023-01-06 Compounds containing a hydroxyphenyl moiety and their use

Country Status (1)

Country Link
WO (1) WO2023131677A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019046711A2 (en) * 2017-09-01 2019-03-07 Optikira Llc Compounds and compositions for ire1 inhibition

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019046711A2 (en) * 2017-09-01 2019-03-07 Optikira Llc Compounds and compositions for ire1 inhibition

Non-Patent Citations (33)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 2011
A. JABOUILLE ET AL., ONCOTARGET, vol. 6, 2015, pages 24922 - 24934
B. DROGAT ET AL., CANCER RES., vol. 67, 2007, pages 6700 - 6707
C. HETZ ET AL., NAT. REV. DRUG DISCOV., vol. 12, 2013, pages 703 - 719
CHEM. COMMUN., vol. 49, 2013, pages 4157
CHEM., vol. 53, 2010, pages 8679
D. DOULTSINOS ET AL., SLAS DISCOV. ADV. LIFE SCI. R&D, vol. 22, 2017, pages 787 - 800
D. PELIZZARI-RAYMUNDO ET AL., TRENDS IN CANCER, vol. 6, 2020, pages 1018 - 1030
F. HONG ET AL., BIOINFORMATICS, vol. 22, 2006, pages 2825 - 2827
F. PRISCHI ET AL., NATURE COMMUNICATIONS, vol. 5, 2014, pages 3554
G. AUF ET AL., PROC NATL ACAD SCI USA, vol. 107, 2010, pages 15553 - 15558
H.C. FELDMAN ET AL., CS CHEM. BIOL., vol. 11, 2016, pages 219 - 2205
J. MED. CHEM., vol. 50, 2007, pages 1076
J. OBACZ ET AL., BIORXIV, 2020, pages 533018
J. OBACZ ET AL., SCI. SIGNAL., vol. 10, 2017, pages eaal2323
L. WANG ET AL., CHEM. BIOL., vol. 8, 2012, pages 982 - 989
LETT, vol. 22, 2020, pages 194 - 1981
M. BOUCHECAREILH ET AL., FASEB J., vol. 25, 2011, pages 3115 - 3129
N. ZHAO ET AL., J. CLIN. INVEST., vol. 128, 2018, pages 1283 - 1299
NUCLEIC ACIDS RESEARCH, vol. 49, 2021, pages D325 - D334
ORG. LETT., vol. 6, 2004, pages 3111 - 3114
P. RESNIK, JOURNAL OF ARTIFICIAL INTELLIGENCE RESEARCH, vol. 11, 1999, pages 95 - 130
PELIZZARI-RAYMUNDO DIANA ET AL: "A novel blood brain barrier-permeable IRE1 kinase inhibitor sensitizes glioblastoma to chemotherapy in mice", CHEMRXIV, 11 January 2022 (2022-01-11), pages 1 - 30, XP055935455, Retrieved from the Internet <URL:-> [retrieved on 20220625] *
R. STUPP ET AL., N. ENGL. J. MED., vol. 352, 2005, pages 987 - 996
S. LHOMOND ET AL., EMBO MOL. MED, vol. 10, 2018, pages 139 - 308
S. LHOMOND ET AL., METHODS MOL. BIOL., vol. 1292, 2015, pages 177 - 194
S. LHOMOND ET AL.: "EMBO Molecular Medicine", vol. 10, 2018, pages: e7929
S.E. LOGUE ET AL., NAT. COMMUN., vol. 9, 2018, pages 3267
T. AVRIL ET AL., BRAIN PATHOL., vol. 22, 2012, pages 159 - 174
T. KOUTSANDREAS ET AL., INT. J. MONIT. SURVEILL. TECHNOL. RES., vol. 4, 2016, pages 30 - 49
T. LANGLAIS, BIOCHEM. J., vol. 478, 2021, pages 2953 - 2975
W. TIRASOPHON ET AL., GENES DEV., vol. 12, 1998, pages 1812 - 1824
WANG LIKUN ET AL: "Divergent allosteric control of the IRE1? endoribonuclease using kinase inhibitors", vol. 8, no. 12, 21 October 2012 (2012-10-21), New York, pages 982 - 989, XP055797928, ISSN: 1552-4450, Retrieved from the Internet <URL:http://www.nature.com/articles/nchembio.1094> DOI: 10.1038/nchembio.1094 *

Similar Documents

Publication Publication Date Title
JP6970802B2 (en) Methods of Inducing Targeted Proteolysis by Bifunctional Molecules
JP7033764B2 (en) Substituted heterocyclyl derivative as a CDK inhibitor
JP7042812B2 (en) Crystal form of triazolopyrimidine compound
CA3041563C (en) Pyrimidin-2-amine derivatives and pharmaceutical compositions thereof useful as inhibitors of cyclin-dependent kinase 12 (cdk12)
KR102021642B1 (en) Methods of treating cancer
EP3572400B1 (en) Ezh2 inhibitor and use thereof
JP5997143B2 (en) Isoxazolo-quinazolines as regulators of protein kinase activity
JP2020502065A (en) 8,9-dihydroimidazole [1,2-a] pyrimido [5,4-e] pyrimidine-5 (6H) -ketone compound
WO2014134774A1 (en) Compounds inhibiting leucine-rich repeat kinase enzyme activity
IL229387A (en) Spiro-oxindole mdm2 antagonists
KR20170095964A (en) Piperidine derivatives as hdac1/2 inhibitors
JP2020512315A (en) Heteroaryl compounds useful as MK2 inhibitors
WO2021222138A1 (en) Compounds for mutant ras protein degradation
JP2020511468A (en) Deuterated analogs of MK2 inhibitors and uses thereof
JP2022033753A (en) Inhibitors of tryptophan dioxygenases (ido1 and tdo) and their use in therapy
US10501466B2 (en) WDR5 inhibitors and modulators
CA3174507A1 (en) Imidazotriazines acting on cancer via inhibition of cdk12
KR102288246B1 (en) Mechanical targets of rapamycin signaling pathway inhibitors and their therapeutic applications
EP3108883A1 (en) Therapeutic uses of non-peptide inhibitors of the calcineurin - nfat signalling pathway
WO2023131677A1 (en) Compounds containing a hydroxyphenyl moiety and their use
JP6257835B2 (en) Novel triazolopyrimidinone or triazolopyridinone derivatives and their uses
EP3114114B1 (en) Tricyclic prodrugs
WO2022006412A2 (en) Conjugates of ampk inhibitors and protac degraders and related uses
Yeh et al. Discovery and development of a novel N-(3-bromophenyl)-{[(phenylcarbamoyl) amino] methyl}-N-hydroxythiophene-2-carboximidamide indoleamine 2, 3-dioxygenase inhibitor using knowledge-based drug design
WO2021003339A1 (en) Amp-activated protein kinase inhibitors and methods of making and using the same

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: 23702391

Country of ref document: EP

Kind code of ref document: A1