US20220135571A1 - Therapeutic egfr inhibitors - Google Patents

Therapeutic egfr inhibitors Download PDF

Info

Publication number
US20220135571A1
US20220135571A1 US17/558,053 US202117558053A US2022135571A1 US 20220135571 A1 US20220135571 A1 US 20220135571A1 US 202117558053 A US202117558053 A US 202117558053A US 2022135571 A1 US2022135571 A1 US 2022135571A1
Authority
US
United States
Prior art keywords
pyrrolo
imidazol
dihydro
indazol
chloro
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
US17/558,053
Inventor
Cosimo Dolente
Annick Goergler
Georg Jaeschke
Bernd Kuhn
Christa Ulrike Obst-Sander
Antonio Ricci
Daniel Rueher
Sandra Steiner
David Hewings
Yvonne Alice Nagel
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Hoffmann La Roche Inc
Original Assignee
Hoffmann La Roche Inc
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 Hoffmann La Roche Inc filed Critical Hoffmann La Roche Inc
Publication of US20220135571A1 publication Critical patent/US20220135571A1/en
Assigned to F. HOFFMANN-LA ROCHE AG reassignment F. HOFFMANN-LA ROCHE AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NAGEL, YVONNE ALICE, DOLENTE, COSIMO, GOERGLER, ANNICK, HEWINGS, DAVID, KUHN, BERND, OBST-SANDER, Christa Ulrike, RICCI, ANTONIO, RUEHER, DANIEL, JAESCHKE, GEORG, STEINER, SANDRA
Assigned to HOFFMANN-LA ROCHE INC. reassignment HOFFMANN-LA ROCHE INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: F. HOFFMANN-LA ROCHE AG
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/485Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4858Organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention provides compounds which are selective allosteric inhibitors of T790M/L858R, T790M/L858R/C797S, L858R, L858R/C797S containing EGFR mutants, their manufacture, pharmaceutical compositions containing them and their use as therapeutically active substances.
  • the present invention provides a novel compounds selected from
  • the HER family receptor tyrosine kinases are mediators of cell growth, differentiation and survival.
  • the receptor family includes four distinct members, i.e. epidermal growth factor receptor (EGFR, ErbB1, or HER1) HER2 (ErbB2), HER3 (ErbB3) and HER4 (ErbB4).
  • EGFR epidermal growth factor receptor
  • ErbB1 HER2
  • HER3 ErbB3
  • HER4 ErbB4
  • EGFR C797S mutation mediates resistance to third-generation inhibitors in T790M-positive non-small cell lung cancer, J Hematol Oncol. 2016; 9: 59). Additional mutations that cause resistance to Osimertinib are described by Yang, for example L718Q. (Yang et al, Investigating Novel Resistance Mechanisms to Third-Generation EGFR Tyrosine Kinase Inhibitor Osimertinib in Non-Small Cell Lung Cancer Patients, Clinical Cancer Research, DOI: 10.1158/1078-0432.CCR-17-2310) Lu et al.
  • WO2009158369 describes certain heterocyclic antibacterial agents.
  • WO2016183534 describes certain heterocyclic compounds suitable as EBNA1 inhibitors.
  • WO2011128279 describes certain heterocyclic compounds suitable as mGluR5 modulators.
  • salts refers to those salts which retain the biological effectiveness and properties of the free bases or free acids, which are not biologically or otherwise undesirable.
  • the salts are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, in particular hydrochloric acid, and organic acids such as acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, N-acetylcystein and the like.
  • salts may be prepared by addition of an inorganic base or an organic base to the free acid.
  • Salts derived from an inorganic base include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium salts and the like.
  • Salts derived from organic bases include, but are not limited to salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, lysine, arginine, N-ethylpiperidine, piperidine, polyimine resins and the like.
  • Particular pharmaceutically acceptable salts of compounds of the present invention are the hydrochloride salts, methanesulfonic acid salts and citric acid salts.
  • uM means microMolar and is equivalent to the symbol ⁇ M.
  • the abbreviation uL means microliter and is equivalent to the symbol ⁇ L.
  • the abbreviation ug means microgram and is equivalent to the symbol ⁇ g.
  • the compounds of the present invention can contain several asymmetric centers and can be present in the form of optically pure enantiomers, mixtures of enantiomers such as, for example, racemates, optically pure diastereoisomers, mixtures of diastereoisomers, diastereoisomeric racemates or mixtures of diastereoisomeric racemates.
  • the asymmetric carbon atom can be of the “R” or “S” configuration.
  • an embodiment of the present invention is a compound as described herein and pharmaceutically acceptable salts thereof, more particularly a compound as described herein.
  • the corresponding pharmaceutically acceptable salts with acids can be obtained by standard methods known to the person skilled in the art, e.g. by dissolving the compound of the present invention in a suitable solvent such as e.g. dioxane or tetrahydrofuran and adding an appropriate amount of the corresponding acid.
  • a suitable solvent such as e.g. dioxane or tetrahydrofuran
  • the products can usually be isolated by filtration or by chromatography.
  • the conversion of a compound of the present invention into a pharmaceutically acceptable salt with a base can be carried out by treatment of such a compound with such a base.
  • One possible method to form such a salt is e.g. by addition of 1/n equivalents of a basic salt such as e.g.
  • a suitable solvent e.g. ethanol, ethanol-water mixture, tetrahydrofuran-water mixture
  • Particular salts are hydrochloride, formate and trifluoroacetate.
  • the compounds of the present invention may be derivatised at functional groups to provide derivatives which are capable of conversion back to the parent compound in vivo.
  • a certain embodiment of the invention relates to the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, for use as therapeutically active substance.
  • a certain embodiment of the invention relates to the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, for the use in the therapeutic and/or prophylactic treatment of cancer, in particular non-small-cell lung cancer.
  • a certain embodiment of the invention relates to the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, for the use in the therapeutic and/or prophylactic treatment of non-small-cell lung cancer.
  • a certain embodiment of the invention relates to the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the therapeutic and/or prophylactic treatment of cancer, in particular non-small-cell lung cancer.
  • a certain embodiment of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable auxiliary substance.
  • a certain embodiment of the invention relates to a method for the therapeutic and/or prophylactic treatment of cancer, in particular non-small-cell lung cancer by administering the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, to a patient.
  • a certain embodiment of the invention relates to the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, for the use as a medicament in therapeutic and/or prophylactic treatment of a patient with EGFR activating mutations suffering from cancer, in particular non-small-cell lung cancer, comprising determining the EGFR activating mutations status in said patient and then administering the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, to said patient.
  • a certain embodiment of the invention relates to the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, for the use as a medicament in therapeutic and/or prophylactic treatment of a patient with EGFR mutations T790M/L858R, T790M/L858R/C797S, L858R and/or L858R/C797S suffering from cancer, in particular non-small-cell lung cancer, comprising determining the EGFR activating mutations status in said patient and then administering the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, to said patient.
  • a certain embodiment of the invention relates to the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, for the use as a medicament in therapeutic and/or prophylactic treatment of a patient with EGFR activating mutations as determined with a Cobas® EGFR Mutation Test v2 suffering from cancer, in particular non-small-cell lung cancer, comprising determining the EGFR activating mutations status in said patient and then administering the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, to said patient.
  • the invention includes all substituents in its corresponding deuterated form, wherever applicable, of the compounds of the present invention.
  • the invention includes all optical isomers, i.e. diastereoisomers, diastereomeric mixtures, racemic mixtures, all their corresponding enantiomers and/or tautomers as well as their solvates, wherever applicable, of the compounds of the present invention.
  • the compounds of the present invention may contain one or more asymmetric centers and can therefore occur as racemates, racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. Additional asymmetric centers may be present depending upon the nature of the various substituents on the molecule. Each such asymmetric center will independently produce two optical isomers and it is intended that all of the possible optical isomers and diastereomers in mixtures and as pure or partially purified compounds are included within this invention. The present invention is meant to encompass all such isomeric forms of these compounds. The independent syntheses of these diastereomers or their chromatographic separations may be achieved as known in the art by appropriate modification of the methodology disclosed herein.
  • Their absolute stereochemistry may be determined by the x-ray crystallography of crystalline products or crystalline intermediates which are derivatized, if necessary, with a reagent containing an asymmetric center of known absolute configuration.
  • racemic mixtures of the compounds may be separated so that the individual enantiomers are isolated. The separation can be carried out by methods well known in the art, such as the coupling of a racemic mixture of compounds to an enantiomerically pure compound to form a diastereomeric mixture, followed by separation of the individual diastereomers by standard methods, such as fractional crystallization or chromatography.
  • optically pure enantiomer means that the compound contains >90% of the desired isomer by weight, particularly >95% of the desired isomer by weight, or more particularly >99% of the desired isomer by weight, said weight percent based upon the total weight of the isomer(s) of the compound.
  • Chirally pure or chirally enriched compounds may be prepared by chirally selective synthesis or by separation of enantiomers. The separation of enantiomers may be carried out on the final product or alternatively on a suitable intermediate.
  • an embodiment of the present invention are compounds of the present invention as described herein, when manufactured according to any one of the described processes.
  • H1975 cell line (CRL-5908; Lot BA70803) was obtained from American Type Culture Collection (Manassas, Va., USA). Cells were maintained at 37° C., 5% CO 2 in complete Media RPMI 1640 without phenol red containing 0.3 mg/ml glutamine, 100 IU/ml penicillin, and 100 mg/ml streptomycin (Gibco, 15140-122) supplemented with 10% fetal bovine serum (FBS, Gibco, 10091-148).
  • FBS fetal bovine serum
  • Cells were cultured for 24 h in a 384-well plate (Greiner Bio-One, Nr. 784-080; 5000 cells/well) using 8 ⁇ l of complete medium/well. Then 4 l/well of the 3 ⁇ compound solution, containing a factor 3 dilution series of the compound or DMSO in starving medium, were added to the cells (final DMSO 0.33%). After 16 hours at 37° C., 5% CO2, 95% rel. humidity cells were lysed by adding to the compound mix 4 l/well of the supplemented lysis buffer (Cis-bio, Phospho-EGFR HTRF kit, 64EG1PEH), followed by incubation for 30 min at room temperature with shaking (400 rpm).
  • the supplemented lysis buffer Cis-bio, Phospho-EGFR HTRF kit, 64EG1PEH
  • BaF3-TMLRCS cell line were obtained from Crownbio (San Diego, Calif., USA). Cells were maintained at 37° C., 5% CO 2 in RPMI ATCC (Gibco 31870)+2 mM Glutamine+0.5 ⁇ g/ml Puromycin supplemented with 10% fetal bovine serum (FBS) (Gibco).
  • FBS fetal bovine serum
  • the compounds of the present invention and their pharmaceutically acceptable salts can be used as medicaments (e.g. in the form of pharmaceutical preparations).
  • the pharmaceutical preparations can be administered internally, such as orally (e.g. in the form of tablets, coated tablets, dragées, hard and soft gelatin capsules, solutions, emulsions or suspensions), nasally (e.g. in the form of nasal sprays), rectally (e.g. in the form of suppositories) or topical ocularly (e.g. in the form of solutions, ointments, gels or water soluble polymeric inserts).
  • the administration can also be effected parenterally, such as intramuscularly, intravenously, or intraocularly (e.g. in the form of sterile injection solutions).
  • the compounds of the present invention and their pharmaceutically acceptable salts can be processed with pharmaceutically inert, inorganic or organic adjuvants for the production of tablets, coated tablets, dragées, hard gelatin capsules, injection solutions or topical formulations Lactose, corn starch or derivatives thereof, talc, stearic acid or its salts etc. can be used, for example, as such adjuvants for tablets, dragées and hard gelatin capsules.
  • Suitable adjuvants for soft gelatin capsules are, for example, vegetable oils, waxes, fats, semi-solid substances and liquid polyols, etc.
  • Suitable adjuvants for the production of solutions and syrups are, for example, water, polyols, saccharose, invert sugar, glucose, etc.
  • Suitable adjuvants for injection solutions are, for example, water, alcohols, polyols, glycerol, vegetable oils, etc.
  • Suitable adjuvants for suppositories are, for example, natural or hardened oils, waxes, fats, semi-solid or liquid polyols, etc.
  • Suitable adjuvants for topical ocular formulations are, for example, cyclodextrins, mannitol or many other carriers and excipients known in the art.
  • the pharmaceutical preparations can contain preservatives, solubilizers, viscosity-increasing substances, stabilizers, wetting agents, emulsifiers, sweeteners, colorants, flavorants, salts for varying the osmotic pressure, buffers, masking agents or antioxidants. They can also contain still other therapeutically valuable substances.
  • the dosage can vary in wide limits and will, of course, be fitted to the individual requirements in each particular case.
  • the formulation can contain 0.001% to 15% by weight of medicament and the required dose, which can be between 0.1 and 25 mg in can be administered either by single dose per day or per week, or by multiple doses (2 to 4) per day, or by multiple doses per week It will, however, be clear that the upper or lower limit given herein can be exceeded when this is shown to be indicated.
  • a compound of the present invention, lactose and corn starch are firstly mixed in a mixer and then in a comminuting machine.
  • the mixture is returned to the mixer; the talc is added thereto and mixed thoapproximatively.
  • the mixture is filled by machine into suitable capsules, e.g. hard gelatin capsules.
  • the pure enantiomers can be obtained by methods described herein or by methods known to those skilled in the art, such as e.g. chiral chromatography or crystallization.
  • Substituted indazoles (II) are known or can be prepared in analogy to known methods or using the methods described below.
  • Step 1 tert-Butyl (2S)-2-(2-(6-bromo-4-chloro-2H-indazol-2-yl)-3-ethoxy-3-oxopropanoyl)pyrrolidine-1-carboxylate
  • Step 2 Ethyl 2-(6-bromo-4-chloro-2H-indazol-2-yl)-2-(3-thioxo-2,5,6,7-tetrahydro-3H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • Step 3 Ethyl 2-(6-bromo-4-chloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • Step 4 tert-Butyl 4-(4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-ethoxy-2-oxoethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • Step 5 tert-Butyl 4-(4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • Step 6 2-(4-Chloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride
  • Step 1 tert-Butyl 4-(2-chloro-4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-ethoxy-2-oxoethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • step 4 ethyl 2-(6-bromo-4-chloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate (Example 1, step 3) was treated with (4-(4-(tert-butoxycarbonyl)piperazin-1-yl)-3-chlorophenyl)boronic acid in toluene in the presence of cesium carbonate and [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) to give the title compound as off-white solid.
  • MS: m/e 639.5 ([M+H] + )
  • Step 2 tert-Butyl 4-(2-chloro-4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • Step 2 2-(4-Chloro-6-(3-chloro-4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride
  • step 6 a solution of tert-butyl 4-(2-chloro-4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate in dichloromethane and methanol was treated with HCl in dioxane to give the title compound as off-white solid.
  • MS: m/e 591.5 ([M ⁇ H] ⁇ )
  • Step 1 tert-Butyl (2S,4R)-2-(3-ethoxy-2-(4-fluoro-6-iodo-2H-indazol-2-yl)-3-oxopropanoyl)-4-fluoropyrrolidine-1-carboxylate
  • step 1 (2S,4R)-1-(tert-butoxycarbonyl)-4-fluoropyrrolidine-2-carboxylic acid was treated with carbonyldiimidazole to give solution A.
  • Ethyl 2-(4-fluoro-6-iodo-2H-indazol-2-yl)acetate was deprotonated with LDA and treated with solution A at ⁇ 78° C. After stirring at room temperature overnight and workup in analogy to Example 1, step 1, the crude title compound was obtained which was used for the next step without further purification.
  • MS: m/e 564.3 ([M+H] + )
  • Step 2 Ethyl 2-((R)-6-fluoro-3-thioxo-2,5,6,7-tetrahydro-3H-pyrrolo[1,2-c]imidazol-1-yl)-2-(4-fluoro-6-iodo-2H-indazol-2-yl)acetate
  • Step 3 Ethyl 2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-(4-fluoro-6-iodo-2H-indazol-2-yl)acetate
  • Step 4 tert-butyl 4-(2-chloro-4-(2-(2-ethoxy-1-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxoethyl)-4-fluoro-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • step 4 ethyl 2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-(4-fluoro-6-iodo-2H-indazol-2-yl)acetate was reacted with (4-(4-(tert-butoxycarbonyl)piperazin-1-yl)-3-chlorophenyl)boronic acid in toluene in the presence of [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) and cesium carbonate to give the title compound as light brown solid.
  • MS: m/e 641.5 ([M+H] + )
  • Step 5 tert-Butyl 4-(2-chloro-4-(4-fluoro-2-(1-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • Step 6 2-(6-(3-Chloro-4-(piperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride
  • Step 1 tert-Butyl (2S)-2-(2-(6-bromo-4,7-dichloro-2H-indazol-2-yl)-3-ethoxy-3-oxopropanoyl)pyrrolidine-1-carboxylate
  • step 1 (tert-butoxycarbonyl)-L-proline was treated with carbonyldiimidazole to give solution A.
  • Ethyl 2-(6-bromo-4,7-dichloro-2H-indazol-2-yl)acetate was deprotonated with LDA and treated with solution A at ⁇ 78° C. After stirring at room temperature overnight and workup in analogy to Example 1, step 1, the crude title compound was obtained which was used for the next step without further purification.
  • MS: m/e 550.2 ([M+H] + )
  • Step 2 Ethyl 2-(6-bromo-4,7-dichloro-2H-indazol-2-yl)-2-(3-thioxo-2,5,6,7-tetrahydro-3H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • step 2 tert-butyl (2S)-2-(2-(6-bromo-4,7-dichloro-2H-indazol-2-yl)-3-ethoxy-3-oxopropanoyl)pyrrolidine-1-carboxylate was deprotected using HCl in dioxane followed by reaction with potassium thiocyanate to give the crude title compound which was used for the next step without further purification.
  • MS: m/e 491.1 ([M+H] + , Br)
  • Step 3 Ethyl 2-(6-bromo-4,7-dichloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • Step 4 tert-Butyl 4-(4-(4,7-dichloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-ethoxy-2-oxoethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • step 4 ethyl 2-(6-bromo-4,7-dichloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate was treated with (4-(4-(tert-butoxycarbonyl)piperazin-1-yl)phenyl)boronic acid in toluene in the presence of cesium carbonate and [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) to give the title compound as light brown solid.
  • MS: m/e 639.5 ([M+H] + )
  • Step 5 tert-Butyl 4-(4-(4,7-dichloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • Step 6 2-(4,7-Dichloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride
  • step 6 a solution of tert-butyl 4-(4-(4,7-dichloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate in dichloromethane and methanol was treated with HCl in dioxane to give the title compound as light yellow solid.
  • MS: m/e 591.4 ([M ⁇ H] ⁇ )
  • Step 1 tert-Butyl (2S)-2-(2-(6-bromo-4-chloro-7-fluoro-2H-indazol-2-yl)-3-ethoxy-3-oxopropanoyl)pyrrolidine-1-carboxylate
  • step 1 (tert-butoxycarbonyl)-L-proline was treated with carbonyldiimidazole to give solution A.
  • Ethyl 2-(6-bromo-4-chloro-7-fluoro-2H-indazol-2-yl)acetate was deprotonated with LDA and treated with solution A at ⁇ 78° C. After stirring at room temperature overnight and workup in analogy to Example 1, step 1, the crude title compound was obtained which was used for the next step without further purification.
  • Step 2 Ethyl 2-(6-bromo-4-chloro-7-fluoro-2H-indazol-2-yl)-2-(3-thioxo-2,5,6,7-tetrahydro-3H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • Step 3 Ethyl 2-(6-bromo-4-chloro-7-fluoro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • Step 4 tert-Butyl 4-(4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-ethoxy-2-oxoethyl)-7-fluoro-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • step 4 ethyl 2-(6-bromo-4-chloro-7-fluoro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate was treated with (4-(4-(tert-butoxycarbonyl)piperazin-1-yl)phenyl)boronic acid in toluene in the presence of cesium carbonate and [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) to give the title compound as off-white solid.
  • MS: m/e 623.4 ([M+H] + )
  • Step 5 tert-Butyl 4-(4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-7-fluoro-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • Step 6 2-(4-Chloro-7-fluoro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride
  • Step 1 Ethyl 2-(4-chloro-7-fluoro-6-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • step 4 ethyl 2-(6-bromo-4-chloro-7-fluoro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate (Example 15, step 3) was reacted with 1-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,4-dihydroquinolin-2(1H)-one in toluene in the presence of [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II), cesium carbonate and 10 equivalents of water to give the title compound as off-white solid.
  • MS: m/e 522.3 ([M+H] + )
  • Step 2 2-(4-Chloro-7-fluoro-6-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide
  • Step 1 tert-Butyl (2S,4R)-2-(2-(6-bromo-4-chloro-7-methoxy-2H-indazol-2-yl)-3-ethoxy-3-oxopropanoyl)-4-fluoropyrrolidine-1-carboxylate
  • step 1 (2S,4R)-1-(tert-butoxycarbonyl)-4-fluoropyrrolidine-2-carboxylic acid was treated with carbonyldiimidazole to give solution A.
  • Ethyl 2-(6-bromo-4-chloro-7-methoxy-2H-indazol-2-yl)acetate was deprotonated with LDA and treated with solution A at ⁇ 78° C. After stirring at room temperature for 90 min and workup in analogy to Example 1, step 1, the crude title compound was obtained which was used for the next step without further purification.
  • MS: m/e 564.2 ([M+H] + , Br)
  • Step 2 Ethyl 2-(6-bromo-4-chloro-7-methoxy-2H-indazol-2-yl)-2-((R)-6-fluoro-3-thioxo-2,5,6,7-tetrahydro-3H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • Step 3 Ethyl 2-(6-bromo-4-chloro-7-methoxy-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • Step 4 Ethyl 2-(4-chloro-7-methoxy-6-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • step 4 ethyl 2-(6-bromo-4-chloro-7-methoxy-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate (Example 19, step 3) was reacted with 1-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,4-dihydroquinolin-2(1H)-one in toluene in the presence of [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II), cesium carbonate and 10 equivalents of water to give the title compound as off-white solid.
  • MS: m/e 552.3 ([M+H] + )
  • Step 5 2-[4-Chloro-7-methoxy-6-(1-methyl-2-oxo-3,4-dihydroquinolin-6-yl)indazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide
  • Step 1 tert-Butyl 4-(4-(4-chloro-2-(2-ethoxy-1-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxoethyl)-7-methoxy-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • step 4 ethyl 2-(6-bromo-4-chloro-7-methoxy-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate (Example 20, step 3) was reacted with (4-(4-(tert-butoxycarbonyl)piperazin-1-yl)phenyl)boronic acid in toluene in the presence of [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) and cesium carbonate to give the title compound as off-white solid.
  • MS: m/e 653.4 ([M+H] + )
  • Step 2 tert-Butyl 4-(4-(4-chloro-2-(1-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-7-methoxy-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • Step 3 2-(4-Chloro-7-methoxy-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride
  • Step 4 2-[4-Chloro-6-[4-(4-ethylpiperazin-1-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide
  • Step 1 Ethyl 2-(4-chloro-6-(4-(1-cyclopropylpiperidin-4-yl)phenyl)-7-methoxy-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • step 4 ethyl 2-(6-bromo-4-chloro-7-methoxy-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate (Example 20, step 3) was reacted with 1-cyclopropyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine (CAS #2244702-81-4) in THF and water in the presence of [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) and cesium carbonate to give the title compound as light brown solid.
  • MS: m/e 592.4 ([M+H] + )
  • Step 2 2-[4-Chloro-6-[4-(1-cyclopropylpiperidin-4-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide
  • Step 1 Ethyl 2-(4-chloro-7-fluoro-6-(1-isopropyl-2-oxo-1,2-dihydropyridin-4-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • step 4 ethyl 2-(6-bromo-4-chloro-7-fluoro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate (Example 15, step 3) was reacted with 1-isopropyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2(1H)-one in toluene in the presence of [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II), cesium carbonate and equivalents of water to give the title compound as off-white solid.
  • MS: m/e 498.4 ([M+H] + )
  • Step 2 2-(4-Chloro-7-fluoro-6-(1-isopropyl-2-oxo-1,2-dihydropyridin-4-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide

Abstract

The invention provides novel compounds as described herein, compositions including the compounds and methods of using the compounds.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of International Application No. PCT/EP2020/067082, filed in the European Receiving Office on Jun. 19, 2020, which claims the benefit of European Patent Application 19181754.3, filed Jun. 21, 2019. The entirety of these applications are hereby incorporated by reference herein for all purposes.
  • SUMMARY OF THE INVENTION
  • The present invention provides compounds which are selective allosteric inhibitors of T790M/L858R, T790M/L858R/C797S, L858R, L858R/C797S containing EGFR mutants, their manufacture, pharmaceutical compositions containing them and their use as therapeutically active substances.
  • The present invention provides a novel compounds selected from
    • 2-(4-chloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
    • 2-(6-(4-(4-acetylpiperazin-1-yl)phenyl)-4-chloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
    • 2-(4-chloro-6-(3-chloro-4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
    • 2-(4-chloro-6-(3-chloro-4-(4-ethylpiperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
    • 2-(6-(3-chloro-4-(piperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
    • 2-(6-(4-(4-acetylpiperazin-1-yl)-3-chlorophenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
    • 2-(6-(3-chloro-4-(4-ethylpiperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
    • 2-(4,7-dichloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
    • 2-(6-(4-(4-acetylpiperazin-1-yl)phenyl)-4,7-dichloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
    • 2-(4,7-dichloro-6-(4-(4-ethylpiperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
    • 2-(4-chloro-7-fluoro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
    • 2-(4-chloro-6-(4-(4-ethylpiperazin-1-yl)phenyl)-7-fluoro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
    • 2-(4-chloro-7-fluoro-6-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
    • 2-[4-chloro-7-methoxy-6-(1-methyl-2-oxo-3,4-dihydroquinolin-6-yl)indazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide;
    • 2-[4-chloro-6-[4-(4-ethylpiperazin-1-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide;
    • 2-[4-chloro-6-[4-(1-cyclopropylpiperidin-4-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide;
    • 2-(4-chloro-7-fluoro-6-(1-isopropyl-2-oxo-1,2-dihydropyridin-4-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide or pharmaceutically acceptable salts.
  • The HER family receptor tyrosine kinases are mediators of cell growth, differentiation and survival. The receptor family includes four distinct members, i.e. epidermal growth factor receptor (EGFR, ErbB1, or HER1) HER2 (ErbB2), HER3 (ErbB3) and HER4 (ErbB4). Upon ligand binding the receptors form homo and heterodimers and subsequent activation of the intrinsic tyrosine kinase activity leads to receptor auto-phosphorylation and the activation of downstream signaling molecules (Yarden, Y., Sliwkowski, MX. Untangling the ErbB signalling network. Nature Review Mol Cell Biol. 2001 February; 2(2): 127-37). De-regulation of EGFR by overexpression or mutation has been implicated in many types of human cancer including colorectal, pancreatic, gliomas, head and neck and lung cancer, in particular non-small cell lung cancer (NSCLC) and several EGFR targeting agents have been developed over the years (Ciardiello, F., and Tortora, G. (2008). EGFR antagonists in cancer treatment. The New England journal of medicine 358, 1160-1174). Erlotinib (Tarceva®), a reversible inhibitor of the EGFR tyrosine kinase was approved in numerous countries for the treatment of recurrent NSCLC.
  • An impressive single agent activity of EGFR tyrosine kinase inhibitors is observed in a subset of NSCLC patients whose tumors harbor somatic kinase domain mutations, whereas clinical benefit in wild-type EGFR patients is greatly diminished (Paez, J. et al. (2004). EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science (New York, N.Y. 304, 1497-1500). The most common somatic mutations of EGFR are exon 19 deletions with delta 746-750 the most prevalent mutation and the exon 21 amino acid substitutions with L858R the most frequent mutation (Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer. 2007 March; 7(3): 169-81).
  • Treatment resistance arises frequently, often due to the secondary T790M mutation within the ATP site of the receptor. Some developed mutant-selective irreversible inhibitors are highly active against the T790M mutant, but their efficacy can be compromised by acquired mutation of C797S, that is the cysteine residue with which they form a key covalent bond (Thress, K. S. et al. Acquired EGFR C797S mutation mediates resistance to AZD9291 in non-small cell lung cancer harboring EGFR T790M. Nat. Med. 21, 560-562 (2015)). C797S mutation was further reported by Wang to be a major mechanism for resistance to T790M-targeting EGFR inhibitors (Wang et al. EGFR C797S mutation mediates resistance to third-generation inhibitors in T790M-positive non-small cell lung cancer, J Hematol Oncol. 2016; 9: 59). Additional mutations that cause resistance to Osimertinib are described by Yang, for example L718Q. (Yang et al, Investigating Novel Resistance Mechanisms to Third-Generation EGFR Tyrosine Kinase Inhibitor Osimertinib in Non-Small Cell Lung Cancer Patients, Clinical Cancer Research, DOI: 10.1158/1078-0432.CCR-17-2310) Lu et al. (Targeting EGFRL858R/T790M and EGFRL858R/T790M/C797S resistance mutations in NSCLC: Current developments in medicinal chemistry, Med Res Rev 2018; 1-32) report in a review article on Targeting EGFRL858R/T790M and EGFRL858R/T790M/C797S resistance mutations in NSCLC treatment.
  • As most available EGFR tyrosine kinase inhibitors target the ATP-site of the kinase, there is a need for new therapeutic agents that work differently, for example through targeting drug-resistant EGFR mutants.
  • Recent studies suggest that purposefully targeting allosteric sites might lead to mutant-selective inhibitors (Jia et al. Overcoming EGFR(T790M) and EGFR(C797S) resistance with mutant-selective allosteric inhibitoRS, June 2016, Nature 534, 129-132)
  • There is just a need in the generation of selective molecules that specifically inhibit T790M/L858R, T790M/L858R/C797S, L858R, L858R/C797S containing EGFR mutants useful for the therapeutic and/or prophylactic treatment of cancer, in particular T790M and C797S containing EGFR mutants.
  • WO2009158369 describes certain heterocyclic antibacterial agents. WO2016183534 describes certain heterocyclic compounds suitable as EBNA1 inhibitors. WO2011128279 describes certain heterocyclic compounds suitable as mGluR5 modulators.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The term “pharmaceutically acceptable salts” refers to those salts which retain the biological effectiveness and properties of the free bases or free acids, which are not biologically or otherwise undesirable. The salts are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, in particular hydrochloric acid, and organic acids such as acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, N-acetylcystein and the like. In addition, these salts may be prepared by addition of an inorganic base or an organic base to the free acid. Salts derived from an inorganic base include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium salts and the like. Salts derived from organic bases include, but are not limited to salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, lysine, arginine, N-ethylpiperidine, piperidine, polyimine resins and the like. Particular pharmaceutically acceptable salts of compounds of the present invention are the hydrochloride salts, methanesulfonic acid salts and citric acid salts.
  • The abbreviation uM means microMolar and is equivalent to the symbol μM.
  • The abbreviation uL means microliter and is equivalent to the symbol μL.
  • The abbreviation ug means microgram and is equivalent to the symbol μg.
  • The compounds of the present invention can contain several asymmetric centers and can be present in the form of optically pure enantiomers, mixtures of enantiomers such as, for example, racemates, optically pure diastereoisomers, mixtures of diastereoisomers, diastereoisomeric racemates or mixtures of diastereoisomeric racemates.
  • According to the Cahn-Ingold-Prelog Convention the asymmetric carbon atom can be of the “R” or “S” configuration.
  • Also an embodiment of the present invention is a compound as described herein and pharmaceutically acceptable salts thereof, more particularly a compound as described herein.
  • In another embodiment a compound as described herein is selected from
    • 2-(4-chloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
    • 2-(6-(4-(4-acetylpiperazin-1-yl)phenyl)-4-chloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
    • 2-(4-chloro-6-(3-chloro-4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
    • 2-(4-chloro-6-(3-chloro-4-(4-ethylpiperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
    • 2-(6-(3-chloro-4-(piperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
    • 2-(6-(4-(4-acetylpiperazin-1-yl)-3-chlorophenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
    • 2-(6-(3-chloro-4-(4-ethylpiperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
    • 2-(4,7-dichloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
    • 2-(6-(4-(4-acetylpiperazin-1-yl)phenyl)-4,7-dichloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
    • 2-(4,7-dichloro-6-(4-(4-ethylpiperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
    • 2-(4-chloro-7-fluoro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
    • 2-(4-chloro-6-(4-(4-ethylpiperazin-1-yl)phenyl)-7-fluoro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
    • 2-(4-chloro-7-fluoro-6-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
    • 2-[4-chloro-7-methoxy-6-(1-methyl-2-oxo-3,4-dihydroquinolin-6-yl)indazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide;
    • 2-[4-chloro-6-[4-(4-ethylpiperazin-1-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide;
    • 2-[4-chloro-6-[4-(1-cyclopropylpiperidin-4-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide;
    • 2-(4-chloro-7-fluoro-6-(1-isopropyl-2-oxo-1,2-dihydropyridin-4-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide.
  • Processes for the manufacture of a compound of the present invention as described herein are also an object of the invention.
  • The corresponding pharmaceutically acceptable salts with acids can be obtained by standard methods known to the person skilled in the art, e.g. by dissolving the compound of the present invention in a suitable solvent such as e.g. dioxane or tetrahydrofuran and adding an appropriate amount of the corresponding acid. The products can usually be isolated by filtration or by chromatography. The conversion of a compound of the present invention into a pharmaceutically acceptable salt with a base can be carried out by treatment of such a compound with such a base. One possible method to form such a salt is e.g. by addition of 1/n equivalents of a basic salt such as e.g. M(OH)n, wherein M=metal or ammonium cation and n=number of hydroxide anions, to a solution of the compound in a suitable solvent (e.g. ethanol, ethanol-water mixture, tetrahydrofuran-water mixture) and to remove the solvent by evaporation or lyophilisation. Particular salts are hydrochloride, formate and trifluoroacetate.
  • Insofar as their preparation is not described in the examples, the compounds of the present invention as well as all intermediate products can be prepared according to analogous methods or according to the methods set forth herein. Starting materials are commercially available, known in the art or can be prepared by methods known in the art or in analogy thereto.
  • It will be appreciated that the compounds of the present invention may be derivatised at functional groups to provide derivatives which are capable of conversion back to the parent compound in vivo.
  • A certain embodiment of the invention relates to the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, for use as therapeutically active substance.
  • A certain embodiment of the invention relates to the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, for the use in the therapeutic and/or prophylactic treatment of cancer, in particular non-small-cell lung cancer.
  • A certain embodiment of the invention relates to the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, for the use in the therapeutic and/or prophylactic treatment of non-small-cell lung cancer.
  • A certain embodiment of the invention relates to the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the therapeutic and/or prophylactic treatment of cancer, in particular non-small-cell lung cancer.
  • A certain embodiment of the invention relates to a pharmaceutical composition comprising the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable auxiliary substance.
  • A certain embodiment of the invention relates to a method for the therapeutic and/or prophylactic treatment of cancer, in particular non-small-cell lung cancer by administering the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, to a patient.
  • A certain embodiment of the invention relates to the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, for the use as a medicament in therapeutic and/or prophylactic treatment of a patient with EGFR activating mutations suffering from cancer, in particular non-small-cell lung cancer, comprising determining the EGFR activating mutations status in said patient and then administering the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, to said patient.
  • A certain embodiment of the invention relates to the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, for the use as a medicament in therapeutic and/or prophylactic treatment of a patient with EGFR mutations T790M/L858R, T790M/L858R/C797S, L858R and/or L858R/C797S suffering from cancer, in particular non-small-cell lung cancer, comprising determining the EGFR activating mutations status in said patient and then administering the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, to said patient.
  • A certain embodiment of the invention relates to the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, for the use as a medicament in therapeutic and/or prophylactic treatment of a patient with EGFR activating mutations as determined with a Cobas® EGFR Mutation Test v2 suffering from cancer, in particular non-small-cell lung cancer, comprising determining the EGFR activating mutations status in said patient and then administering the compound of the present invention as described herein, or a pharmaceutically acceptable salt thereof, to said patient.
  • Furthermore, the invention includes all substituents in its corresponding deuterated form, wherever applicable, of the compounds of the present invention.
  • Furthermore, the invention includes all optical isomers, i.e. diastereoisomers, diastereomeric mixtures, racemic mixtures, all their corresponding enantiomers and/or tautomers as well as their solvates, wherever applicable, of the compounds of the present invention.
  • The compounds of the present invention may contain one or more asymmetric centers and can therefore occur as racemates, racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. Additional asymmetric centers may be present depending upon the nature of the various substituents on the molecule. Each such asymmetric center will independently produce two optical isomers and it is intended that all of the possible optical isomers and diastereomers in mixtures and as pure or partially purified compounds are included within this invention. The present invention is meant to encompass all such isomeric forms of these compounds. The independent syntheses of these diastereomers or their chromatographic separations may be achieved as known in the art by appropriate modification of the methodology disclosed herein. Their absolute stereochemistry may be determined by the x-ray crystallography of crystalline products or crystalline intermediates which are derivatized, if necessary, with a reagent containing an asymmetric center of known absolute configuration. If desired, racemic mixtures of the compounds may be separated so that the individual enantiomers are isolated. The separation can be carried out by methods well known in the art, such as the coupling of a racemic mixture of compounds to an enantiomerically pure compound to form a diastereomeric mixture, followed by separation of the individual diastereomers by standard methods, such as fractional crystallization or chromatography.
  • In the embodiments, where optically pure enantiomers are provided, optically pure enantiomer means that the compound contains >90% of the desired isomer by weight, particularly >95% of the desired isomer by weight, or more particularly >99% of the desired isomer by weight, said weight percent based upon the total weight of the isomer(s) of the compound. Chirally pure or chirally enriched compounds may be prepared by chirally selective synthesis or by separation of enantiomers. The separation of enantiomers may be carried out on the final product or alternatively on a suitable intermediate.
  • Also an embodiment of the present invention are compounds of the present invention as described herein, when manufactured according to any one of the described processes.
  • Assay Procedures
  • HTRF Phospho EGFR TMLR Assay (Cellular)
  • Cell Line and Media
  • H1975 cell line (CRL-5908; Lot BA70803) was obtained from American Type Culture Collection (Manassas, Va., USA). Cells were maintained at 37° C., 5% CO2 in complete Media RPMI 1640 without phenol red containing 0.3 mg/ml glutamine, 100 IU/ml penicillin, and 100 mg/ml streptomycin (Gibco, 15140-122) supplemented with 10% fetal bovine serum (FBS, Gibco, 10091-148). Compounds were diluted into starving medium RPMI 1640 Media (Gibco, 11835-030) without phenol red containing 0.3 mg/ml glutamine, 100 IU/ml penicillin, and 100 mg/ml streptomycin (Gibco).
  • Protocol
  • Cells were cultured for 24 h in a 384-well plate (Greiner Bio-One, Nr. 784-080; 5000 cells/well) using 8 μl of complete medium/well. Then 4 l/well of the 3× compound solution, containing a factor 3 dilution series of the compound or DMSO in starving medium, were added to the cells (final DMSO 0.33%). After 16 hours at 37° C., 5% CO2, 95% rel. humidity cells were lysed by adding to the compound mix 4 l/well of the supplemented lysis buffer (Cis-bio, Phospho-EGFR HTRF kit, 64EG1PEH), followed by incubation for 30 min at room temperature with shaking (400 rpm). Then 4 μl of a mixture of anti-Phospho-EGFR Cryptate and of anti-Phospho-EGFR-d2 antibody solutions prepared in the detection buffer was added. The plates were then incubated for 4 h at room temperature before reading the fluorescence emission at 620 and 665 nm using an Envision reader (Perkin Elmer).
  • HTRF Phospho EGFR TMLRCS Assay (Cellular) Cell Line and Media
  • BaF3-TMLRCS cell line were obtained from Crownbio (San Diego, Calif., USA). Cells were maintained at 37° C., 5% CO2 in RPMI ATCC (Gibco 31870)+2 mM Glutamine+0.5 μg/ml Puromycin supplemented with 10% fetal bovine serum (FBS) (Gibco).
  • Protocol
  • Cells are transferred as above to Greiner Bio-One, Nr. 784-08 micro-titerplate at 20000 cells/well in 12.5 μl of growth medium/well after the plates had been pre-filled with 12.5 nl of DMSO solutions of the to be tested compounds (in dose response) or DMSO only. After spinning the plates at 300×g for 30 seconds the cells were incubated for 4 hours at 37 C, 5% CO2, 95% humidity. The cells were lysed by adding to the compound mix 4 l/well of the supplemented lysis buffer (Cis-bio, Phospho-EGFR HTRF kit, 64EG1PEH), followed by incubation for 30 min at room temperature with shaking (400 rpm). The plates were then frozen and stored overnight at −80 C. On the next day and after thawing the plates, 4 μl of a mixture of anti-Phospho-EGFR Cryptate and of anti-Phospho-EGFR-d2 antibody solutions prepared in the supplied detection buffer was added to each well. The lidded plates were then incubated for 4 h at room temperature before reading the fluorescence emission at 616 and 665 nm using an Envision reader (Perkin Elmer). Data was analyzed in similar fashion as above using the normalized ratio of the 665 to 616 signals multiplied by 10000.
  • The results are shown in Table 1
  • IC50 (H1975) IC50 (BaF3)
    Example μM μM
    1 0.003 0.007
    2 0.0004 0.005
    3 0.009
    4 0.011 0.014
    6 0.003
    7 0.001
    8 0.005
    9 0.002
    12 0.001
    13 0.001 0.011
    15 0.002
    16 0.002
    18 0.001
    20 0.001
    21 0.003
    22 0.005
    23 0.001
  • The compounds of the present invention and their pharmaceutically acceptable salts can be used as medicaments (e.g. in the form of pharmaceutical preparations). The pharmaceutical preparations can be administered internally, such as orally (e.g. in the form of tablets, coated tablets, dragées, hard and soft gelatin capsules, solutions, emulsions or suspensions), nasally (e.g. in the form of nasal sprays), rectally (e.g. in the form of suppositories) or topical ocularly (e.g. in the form of solutions, ointments, gels or water soluble polymeric inserts). However, the administration can also be effected parenterally, such as intramuscularly, intravenously, or intraocularly (e.g. in the form of sterile injection solutions).
  • The compounds of the present invention and their pharmaceutically acceptable salts can be processed with pharmaceutically inert, inorganic or organic adjuvants for the production of tablets, coated tablets, dragées, hard gelatin capsules, injection solutions or topical formulations Lactose, corn starch or derivatives thereof, talc, stearic acid or its salts etc. can be used, for example, as such adjuvants for tablets, dragées and hard gelatin capsules.
  • Suitable adjuvants for soft gelatin capsules, are, for example, vegetable oils, waxes, fats, semi-solid substances and liquid polyols, etc.
  • Suitable adjuvants for the production of solutions and syrups are, for example, water, polyols, saccharose, invert sugar, glucose, etc.
  • Suitable adjuvants for injection solutions are, for example, water, alcohols, polyols, glycerol, vegetable oils, etc.
  • Suitable adjuvants for suppositories are, for example, natural or hardened oils, waxes, fats, semi-solid or liquid polyols, etc.
  • Suitable adjuvants for topical ocular formulations are, for example, cyclodextrins, mannitol or many other carriers and excipients known in the art.
  • Moreover, the pharmaceutical preparations can contain preservatives, solubilizers, viscosity-increasing substances, stabilizers, wetting agents, emulsifiers, sweeteners, colorants, flavorants, salts for varying the osmotic pressure, buffers, masking agents or antioxidants. They can also contain still other therapeutically valuable substances.
  • The dosage can vary in wide limits and will, of course, be fitted to the individual requirements in each particular case. In general, in the case of oral administration a daily dosage of about 0.1 mg to 20 mg per kg body weight, preferably about 0.5 mg to 4 mg per kg body weight (e.g. about 300 mg per person), divided into preferably 1-3 individual doses, which can consist, for example, of the same amounts, should it be appropriate. In the case of topical administration, the formulation can contain 0.001% to 15% by weight of medicament and the required dose, which can be between 0.1 and 25 mg in can be administered either by single dose per day or per week, or by multiple doses (2 to 4) per day, or by multiple doses per week It will, however, be clear that the upper or lower limit given herein can be exceeded when this is shown to be indicated.
  • Preparation of Pharmaceutical Compositions Comprising Compounds of the Invention
  • Tablets of the following composition are manufactured in the usual manner:
  • mg/tablet
    Ingredient 5 25 100 500
    Compound 5 25 100 500
    Lactose Anhydrous DTG 125 105 30 150
    Sta-Rx 1500 6 6 6 60
    Microcrystalline Cellulose 30 30 30 450
    Magnesium Stearate 1 1 1 1
    Total 167 167 167 831
  • Manufacturing Procedure
    • 1. Mix ingredients 1, 2, 3 and 4 and granulate with purified water.
    • 2. Dry the granules at 50° C.
    • 3. Pass the granules through suitable milling equipment.
    • 4. Add ingredient 5 and mix for three minutes; compress on a suitable press.
  • Capsules of the following composition are manufactured:
  • mg/capsule
    Ingredient 5 25 100 500
    Compound 5 25 100 500
    Hydrous Lactose 159 123 148
    Corn Starch 25 35 40 70
    Talk 10 15 10 25
    Magnesium Stearate 1 2 2 5
    Total 200 200 300 600
  • Manufacturing Procedure
    • 1. Mix ingredients 1, 2 and 3 in a suitable mixer for 30 minutes.
    • 2. Add ingredients 4 and 5 and mix for 3 minutes.
    • 3. Fill into a suitable capsule.
  • A compound of the present invention, lactose and corn starch are firstly mixed in a mixer and then in a comminuting machine. The mixture is returned to the mixer; the talc is added thereto and mixed thoapproximatively. The mixture is filled by machine into suitable capsules, e.g. hard gelatin capsules.
  • Injection solutions of the following composition are manufactured:
  • Ingredient mg/injection solution.
    Compound 3
    Polyethylene Glycol 400 150
    acetic acid q.s. ad pH 5.0
    water for injection solutions ad 1.0 ml
  • The invention is illustrated hereinafter by Examples, which have no limiting character.
  • In case the preparative examples are obtained as a mixture of enantiomers, the pure enantiomers can be obtained by methods described herein or by methods known to those skilled in the art, such as e.g. chiral chromatography or crystallization.
  • EXAMPLES
  • The following examples are provided for illustration of the invention. They should not be considered as limiting the scope of the invention, but merely as being representative thereof.
  • Synthesis of Intermediates
  • Substituted indazoles (II) are known or can be prepared in analogy to known methods or using the methods described below.
  • 6-Bromo-4,7-dichloro-1H-indazole Step 1: 4-Bromo-3,6-dichloro-2-fluorobenzaldehyde
  • A solution of 1-bromo-2,5-dichloro-3-fluorobenzene (9.414 g) in tetrahydrofuran (70 ml) was cooled in a dry ice/acetone bath. LDA, 2 mol/l in THE (21.2 ml) was added and the mixture was stirred at −75° C. for 20 min. N,N-dimethylformamide (2.82 g) was added dropwise and stirred for 1 h. A solution of AcOH in Et2O (1:1, 10 ml) was added. The mixture was allowed to warm to room temperature. Water was added and the mixture was extracted with EtOAc. The organic layers were washed with water, dried (MgSO4), filtered and concentrated in vacuo to give the crude title compound (11.323 g) as light yellow solid. The compound was used for the next step without further purification.
  • Step 2: 6-Bromo-4,7-dichloro-1H-indazole
  • To a solution of 4-bromo-3,6-dichloro-2-fluorobenzaldehyde (10.5 g) in Dioxane (50 ml) was added hydrazine hydrate (3.86 g). The mixture was stirred at room temperature for 3 days. Hydrazine hydrate (386 mg) was added and the mixture was warmed to 70° C. for 7 h. After cooling to room temperature water was added and the precipitated solid was collected by filtration. To the solid was added a small amount of acetonitrile and stirred for 2 h. The solid was collected by filtration, washed with a small amount of acetonitrile and dried to give the title compound (7.842 g) as off-white solid. MS: m/e=267.0 ([M+H]+, Br)
  • 6-Bromo-4-chloro-7-fluoro-2H-indazole Step 1: 4-Bromo-6-chloro-2,3-difluorobenzaldehyde
  • In analogy to the synthesis of 4-bromo-3,6-dichloro-2-fluorobenzaldehyde, 1-bromo-5-chloro-2,3-difluorobenzene was first treated with LDA in tetrahydrofuran at −75° C. followed by treatment with N,N-dimethylformamide. Workup in analogy to the synthesis of 4-bromo-3,6-dichloro-2-fluorobenzaldehyde gave the crude title compound as light brown solid.
  • Step 2: 6-Bromo-4-chloro-7-fluoro-2H-indazole
  • 4-Bromo-6-chloro-2,3-difluorobenzaldehyde (5 g) was combined under argon with 1,4-dioxane (19.6 ml) and hydrazine hydrate (1.18 g) and stirred overnight. Hunig's base (2.53 g) was added and stirred for 18 h at 60° C., for 3 h at 80° C. and overnight at 100° C. The mixture was cooled down to room temperature and concentrated. After addition of CH2Cl2 a solid precipitated. The solid was collected by filtration, washed with CH2Cl2 and dried to give the title compound (3.9 g) as off-white solid. MS: m/e=251.0 ([M+H]+, Br)
  • 6-Bromo-4-chloro-7-methoxy-2H-indazole Step 1: 4-Bromo-6-chloro-2-fluoro-3-methoxybenzaldehyde
  • In analogy to the synthesis of 4-bromo-3,6-dichloro-2-fluorobenzaldehyde, 1-bromo-5-chloro-3-fluoro-2-methoxybenzene was first treated with LDA in Tetrahydrofuran at −75° C. followed by treatment with N,N-dimethylformamide. Workup in analogy to the synthesis of 4-bromo-3,6-dichloro-2-fluorobenzaldehyde gave the crude title compound as light brown solid.
  • Step 2: 6-Bromo-4-chloro-7-methoxy-2H-indazole
  • Crude 4-bromo-6-chloro-2-fluoro-3-methoxybenzaldehyde (11.75 g) was dissolved in THE (50 ml). After addition of hydrazine hydrate (3.83 g) the mixture was stirred at room temperature for 42 h. The temperature was increased to 70° C. and the mixture was stirred for 36 h. The temperature was increased to 90° C. and the mixture was stirred for 7 days. After addition of hydrazine hydrate (15.3 g) the mixture was stirred for 18 h at 90° C. After cooling to room temperature, the mixture was diluted with water and stirred for 30 min. The sticky solid was collected by filtration, washed with water and dried to give the slightly impure title compound (6.88 g) as a light brown solid which was used for the next step without further purification. MS: m/e=261.0 ([M+H]+, Br)
  • General Method A: Alkylation of Indazoles
  • A mixture of indazole (II, 1 eq), ethyl 2-bromoacetate (2 eq) and N,N-dimethylacetamide (small amount to produce a solution) is heated to 100° C. until completion of the reaction (usually 5-48 h). After cooling to room temperature, ice is added and the precipitated solid is collected by filtration and washed with water. Purification of the desired regioisomer can be accomplished by chromatography or in certain cases by re-crystallization from solvents such as EtOH, acetonitrile or dichloromethane.
  • Using General Method A, the following intermediates (III) were prepared:
  • Alkylating MS
    Intermediate (III) Indazole (II) agent m/e
    ethyl 2-(6-bromo-4-chloro-2H- 6-bromo-4-chloro-1H- ethyl 2- 319.0
    indazol-2-yl)acetate indazole (CAS 885518- bromoacetate ([M + H]+,
    99-0) Br)
    ethyl 2-(4-fluoro-6-iodo-2H- 4-fluoro-6-iodo-1H- ethyl 2- 349.1
    indazol-2-yl)acetate indazole (CAS 887568- bromoacetate ([M + H]+)
    03-8)
    ethyl 2-(6-bromo-4,7-dichloro- 6-bromo-4,7-dichloro-1H- ethyl 2- 353.0
    2H-indazol-2-yl)acetate indazole bromoacetate ([M + H]+,
    Br)
    ethyl 2-(6-bromo-4-chloro-7- 6-bromo-4-chloro-7- ethyl 2- 336.96
    fluoro-2H-indazol-2-yl)acetate fluoro-2H-indazole bromoacetate ([M + H]+,
    Br)
    ethyl 2-(6-bromo-4-chloro-7- 6-bromo-4-chloro-7- ethyl 2- 348.98
    methoxy-2H-indazol-2- methoxy-2H-indazole bromoacetate ([M + H]+,
    yl)acetate Br)
  • Synthesis of Examples Example 1
  • Figure US20220135571A1-20220505-C00001
  • 2-(4-Chloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride Step 1: tert-Butyl (2S)-2-(2-(6-bromo-4-chloro-2H-indazol-2-yl)-3-ethoxy-3-oxopropanoyl)pyrrolidine-1-carboxylate
  • A solution of (tert-butoxycarbonyl)-L-proline (1.58 g) in Tetrahydrofuran (7.5 ml) was cooled in an ice bath. Carbonyldiimidazole (1.19 g) was added. The cooling bath was removed and the mixture was stirred for 3 h to give solution A. A solution of ethyl 2-(6-bromo-4-chloro-2H-indazol-2-yl)acetate (1.5 g) in tetrahydrofuran (7.5 ml) was cooled to −75° C. LDA, 2 mol/l in THE (3.66 ml) was added dropwise within 5 min. The mixture was stirred for 30 min at −75° C. Solution A was added dropwise within 5 min. The mixture was allowed to warm to room temperature in the cooling bath overnight. After addition of sat. aqueous NH4Cl-solution, the mixture was extracted twice with EtOAc. The organic layers were washed with water, combined, dried over sodium sulphate and concentrated to dryness to give the crude title compound (2.2 g) which was used for the next step without further purification. MS: m/e=516.2 ([M+H]+, Br)
  • Step 2: Ethyl 2-(6-bromo-4-chloro-2H-indazol-2-yl)-2-(3-thioxo-2,5,6,7-tetrahydro-3H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • A solution of tert-butyl (2S)-2-(2-(6-bromo-4-chloro-2H-indazol-2-yl)-3-ethoxy-3-oxopropanoyl)pyrrolidine-1-carboxylate (2.2 g) in HCl, 4M in dioxane (10.7 ml) was stirred for 1 h at room temperature. The mixture was concentrated to dryness. The residue was dissolved in Ethanol (25 ml), potassium thiocyanate (558 mg) and HCl, 1 M in EtOH (4.27 ml) were added and stirred over night. Water was added and the mixture was extracted with EtOAc. The organic layers were washed with water, dried over MgSO4, filtered, concentrated and dried to give the crude title compound (2.1 g) which was used for the next step without further purification. MS: m/e=457.1 ([M+H]+, Br)
  • Step 3: Ethyl 2-(6-bromo-4-chloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • A solution of ethyl 2-(6-bromo-4-chloro-2H-indazol-2-yl)-2-(3-thioxo-2,5,6,7-tetrahydro-3H-pyrrolo[1,2-c]imidazol-1-yl)acetate (2.13 g) in AcOH (10 ml) was cooled to 10° C. Hydrogen peroxide, 35% (2.78 g) was added dropwise. The reaction mixture was stirred for 45 min at room temperature. The excess of hydrogen peroxide was destroyed by addition of saturated sodium sulfit solution. After addition of some water (just enough to dissolve all salts) and EtOAc the mixture was brought to pH 9 by careful addition of solid sodium carbonate. The mixture was extracted with EtOAc. The organic layers were washed with water, dried over sodium sulphate and concentrated. The product was purified by chromatography (SiO2, 0-20% EtOH in EtOAc) to give the title compound (0.64 g) as light brown solid. MS: m/e=425.2 ([M+H]+, Br)
  • Step 4: tert-Butyl 4-(4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-ethoxy-2-oxoethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • Ethyl 2-(6-bromo-4-chloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate (100 mg), (4-(4-(tert-butoxycarbonyl)piperazin-1-yl)phenyl)boronic acid (217 mg) and cesium carbonate (231 mg) were mixed with toluene (3.08 ml), degassed by bubbling argon through the mixture under ultra sonic treatment. [1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (18 mg) was added and the mixture was stirred for 30 min at 115° C. in a sealed tube. The mixture was cooled to room temperature, diluted with EtOAc, washed with half concentrated sodium carbonate solution, dried over sodium sulphate and concentrated. The crude material was purified by flash chromatography (SiO2, 0% to 100% EtOAc in n-heptane) to give the title compound (83 mg) as light brown solid. MS: m/e=605.6 ([M+H]+)
  • Step 5: tert-Butyl 4-(4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • To a solution of tert-butyl 4-(4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-ethoxy-2-oxoethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate (80.9 mg) in THE (1.81 ml) were added LiOH 1M (201 μl) and Water (722 μl). The mixture was stirred for 30 min at room temperature. The mixture was concentrated and dried. The residue was dissolved in DMF (1.81 ml). After addition of thiazol-2-amine (16.1 mg), HATU (61 mg) and Hunig's base (51.8 mg) the mixture was stirred for 45 min at room temperature. Water was added and the mixture was extracted with EtOAc. The org layers were combined, dried, filtered and concentrated. The crude material was purified by flash chromatography (SiO2, 0% to 40% MeOH in AcOEt) followed by another chromatography (SiO2, 0% to 20% MeOH in dichloromethane) to give the title compound (49 mg) as off-white solid. MS: m/e=659.5 ([M+H]+)
  • Step 6: 2-(4-Chloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride
  • To a solution of tert-butyl 4-(4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate (47.4 mg) in dichloromethane (1.69 ml) and methanol (967 μl) was added HCl, 4M in dioxan (899 μl). The mixture was stirred for 45 min at room temperature. The mixture was concentrated, once suspended in dichloromethane and concentrated and dried to give the title compound (47 mg) as light yellow solid. MS: m/e=559.4 ([M+H]+)
  • Example 2
  • Figure US20220135571A1-20220505-C00002
  • 2-(6-(4-(4-Acetylpiperazin-1-yl)phenyl)-4-chloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide
  • To a suspension of 2-(4-chloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride (Example 1, 40 mg) in dichloromethane (5 ml) were added triethylamine (20.4 mg) and acetyl chloride (5.8 mg) and the mixture was stirred over night. Acetyl chloride (5.8 mg) was added and the mixture was stirred for 1 h. The mixture was diluted with dichloromethane, washed with sat. sodium carbonate solution and with water. The organic layers were dried over sodium sulphate, filtered, concentrated, and dried under high vacuum at 60° C. to give the title compound (37 mg) as a grey solid. MS: m/e=599.5 ([M−H])
  • Example 3
  • Figure US20220135571A1-20220505-C00003
  • 2-(4-Chloro-6-(3-chloro-4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride Step 1: tert-Butyl 4-(2-chloro-4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-ethoxy-2-oxoethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • In analogy to Example 1, step 4, ethyl 2-(6-bromo-4-chloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate (Example 1, step 3) was treated with (4-(4-(tert-butoxycarbonyl)piperazin-1-yl)-3-chlorophenyl)boronic acid in toluene in the presence of cesium carbonate and [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) to give the title compound as off-white solid. MS: m/e=639.5 ([M+H]+)
  • Step 2: tert-Butyl 4-(2-chloro-4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • In analogy to Example 1, step 5, tert-butyl 4-(2-chloro-4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-ethoxy-2-oxoethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate was first treated with LiOH and the resulting acid salt was reacted with thiazol-2-amine in the presence of HATU and Hunig's base to give the title compound as off-white solid. MS: m/e=693.4 ([M+H]+)
  • Step 2: 2-(4-Chloro-6-(3-chloro-4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride
  • In analogy to Example 1, step 6, a solution of tert-butyl 4-(2-chloro-4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate in dichloromethane and methanol was treated with HCl in dioxane to give the title compound as off-white solid. MS: m/e=591.5 ([M−H])
  • Example 4
  • Figure US20220135571A1-20220505-C00004
  • 2-(4-Chloro-6-(3-chloro-4-(4-ethylpiperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide
  • To a solution of 2-(4-chloro-6-(3-chloro-4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride (58.0 mg) in N,N-dimethylformamide (2.14 ml) were added iodoethane (15.8 mg) and Hunig's base (35.7 mg) and the mixture was stirred overnight. The reaction mixture was poured into H2O (25 ml) and extracted with EtOAc, dried, filtered and concentrated. The crude material was purified by preparative TLC (SiO2, 2.0 mm, 9:1 dichloromethane/MeOH) to give the title compound (17 mg) as off-white solid. MS: m/e=619.6 ([M−H])
  • Example 6
  • Figure US20220135571A1-20220505-C00005
  • 2-(6-(3-Chloro-4-(piperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride Step 1: tert-Butyl (2S,4R)-2-(3-ethoxy-2-(4-fluoro-6-iodo-2H-indazol-2-yl)-3-oxopropanoyl)-4-fluoropyrrolidine-1-carboxylate
  • In analogy to Example 1, step 1, (2S,4R)-1-(tert-butoxycarbonyl)-4-fluoropyrrolidine-2-carboxylic acid was treated with carbonyldiimidazole to give solution A. Ethyl 2-(4-fluoro-6-iodo-2H-indazol-2-yl)acetate was deprotonated with LDA and treated with solution A at −78° C. After stirring at room temperature overnight and workup in analogy to Example 1, step 1, the crude title compound was obtained which was used for the next step without further purification. MS: m/e=564.3 ([M+H]+)
  • Step 2: Ethyl 2-((R)-6-fluoro-3-thioxo-2,5,6,7-tetrahydro-3H-pyrrolo[1,2-c]imidazol-1-yl)-2-(4-fluoro-6-iodo-2H-indazol-2-yl)acetate
  • In analogy to Example 1, step 2, tert-butyl (2S,4R)-2-(3-ethoxy-2-(4-fluoro-6-iodo-2H-indazol-2-yl)-3-oxopropanoyl)-4-fluoropyrrolidine-1-carboxylate was deprotected using HCl in dioxane followed by reaction with potassium thiocyanate to give the crude title compound which was used for the next step without further purification. MS: m/e=505.2 ([M+H]+)
  • Step 3: Ethyl 2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-(4-fluoro-6-iodo-2H-indazol-2-yl)acetate
  • In analogy to Example 1, step 3, ethyl 2-((R)-6-fluoro-3-thioxo-2,5,6,7-tetrahydro-3H-pyrrolo[1,2-c]imidazol-1-yl)-2-(4-fluoro-6-iodo-2H-indazol-2-yl)acetate was treated with hydrogen peroxide in AcOH to give the title compound as light brown solid. MS: m/e=473.3 ([M+H]+)
  • Step 4: tert-butyl 4-(2-chloro-4-(2-(2-ethoxy-1-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxoethyl)-4-fluoro-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • In analogy to Example 1, step 4, ethyl 2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-(4-fluoro-6-iodo-2H-indazol-2-yl)acetate was reacted with (4-(4-(tert-butoxycarbonyl)piperazin-1-yl)-3-chlorophenyl)boronic acid in toluene in the presence of [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) and cesium carbonate to give the title compound as light brown solid. MS: m/e=641.5 ([M+H]+)
  • Step 5: tert-Butyl 4-(2-chloro-4-(4-fluoro-2-(1-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • In analogy to Example 1, step 5, tert-butyl 4-(2-chloro-4-(2-(2-ethoxy-1-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxoethyl)-4-fluoro-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate was first treated with LiOH and the resulting acid salt was reacted with thiazol-2-amine in the presence of HATU and Hunig's base to give the title compound as off-white solid. MS: m/e=695.5 ([M+H]+)
  • Step 6: 2-(6-(3-Chloro-4-(piperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride
  • In analogy to Example 1, step 6, a solution of tert-butyl 4-(2-chloro-4-(4-fluoro-2-(1-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate in dichloromethane and methanol was treated with HCl in dioxane to give the title compound as off-white solid. MS: m/e=595.4 ([M+H]+)
  • Example 7
  • Figure US20220135571A1-20220505-C00006
  • 2-(6-(4-(4-Acetylpiperazin-1-yl)-3-chlorophenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide
  • In analogy to Example 2, 2-(6-(3-chloro-4-(piperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride (Example 6) was treated with acetyl chloride in the presence of triethylamine to give the title compound as off-white solid. MS: m/e=637.4 ([M+H]+)
  • Example 8
  • Figure US20220135571A1-20220505-C00007
  • 2-(6-(3-Chloro-4-(4-ethylpiperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide
  • In analogy to Example 4, 2-(6-(3-chloro-4-(piperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride (Example 6) was treated with iodoethane in the presence of triethylamine to give the title compound as off-white solid. MS: m/e=623.4 ([M+H]+)
  • Example 9
  • Figure US20220135571A1-20220505-C00008
  • 2-(4,7-Dichloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride Step 1: tert-Butyl (2S)-2-(2-(6-bromo-4,7-dichloro-2H-indazol-2-yl)-3-ethoxy-3-oxopropanoyl)pyrrolidine-1-carboxylate
  • In analogy to Example 1, step 1, (tert-butoxycarbonyl)-L-proline was treated with carbonyldiimidazole to give solution A. Ethyl 2-(6-bromo-4,7-dichloro-2H-indazol-2-yl)acetate was deprotonated with LDA and treated with solution A at −78° C. After stirring at room temperature overnight and workup in analogy to Example 1, step 1, the crude title compound was obtained which was used for the next step without further purification. MS: m/e=550.2 ([M+H]+)
  • Step 2: Ethyl 2-(6-bromo-4,7-dichloro-2H-indazol-2-yl)-2-(3-thioxo-2,5,6,7-tetrahydro-3H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • In analogy to Example 1, step 2, tert-butyl (2S)-2-(2-(6-bromo-4,7-dichloro-2H-indazol-2-yl)-3-ethoxy-3-oxopropanoyl)pyrrolidine-1-carboxylate was deprotected using HCl in dioxane followed by reaction with potassium thiocyanate to give the crude title compound which was used for the next step without further purification. MS: m/e=491.1 ([M+H]+, Br)
  • Step 3: Ethyl 2-(6-bromo-4,7-dichloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • In analogy to Example 1, step 3, ethyl 2-(6-bromo-4,7-dichloro-2H-indazol-2-yl)-2-(3-thioxo-2,5,6,7-tetrahydro-3H-pyrrolo[1,2-c]imidazol-1-yl)acetate was treated with hydrogen peroxide in AcOH to give the title compound as light yellow solid. MS: m/e=457.1 ([M+H]+, Br)
  • Step 4: tert-Butyl 4-(4-(4,7-dichloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-ethoxy-2-oxoethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • In analogy to Example 1, step 4, ethyl 2-(6-bromo-4,7-dichloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate was treated with (4-(4-(tert-butoxycarbonyl)piperazin-1-yl)phenyl)boronic acid in toluene in the presence of cesium carbonate and [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) to give the title compound as light brown solid. MS: m/e=639.5 ([M+H]+)
  • Step 5: tert-Butyl 4-(4-(4,7-dichloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • In analogy to Example 1, step 5, tert-butyl 4-(4-(4,7-dichloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-ethoxy-2-oxoethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate was first treated with LiOH and the resulting acid salt was reacted with thiazol-2-amine in the presence of HATU and Hunig's base to give the title compound as light brown solid. MS: m/e=691.5 ([M−H])
  • Step 6: 2-(4,7-Dichloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride
  • In analogy to Example 1, step 6, a solution of tert-butyl 4-(4-(4,7-dichloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate in dichloromethane and methanol was treated with HCl in dioxane to give the title compound as light yellow solid. MS: m/e=591.4 ([M−H])
  • Example 12
  • Figure US20220135571A1-20220505-C00009
  • 2-(6-(4-(4-Acetylpiperazin-1-yl)phenyl)-4,7-dichloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide
  • In analogy to Example 2, 2-(4,7-dichloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride (Example 9) was treated with acetyl chloride in the presence of triethylamine to give the title compound as light brown solid. MS: m/e=635.3 ([M+H]+)
  • Example 13
  • Figure US20220135571A1-20220505-C00010
  • 2-(4,7-Dichloro-6-(4-(4-ethylpiperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide
  • In analogy to Example 4, 2-(4,7-dichloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride (Example 9) was treated with iodoethane in the presence of triethylamine to give the title compound as light brown solid. MS: m/e=621.3 ([M+H]+)
  • Example 15
  • Figure US20220135571A1-20220505-C00011
  • 2-(4-Chloro-7-fluoro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride Step 1: tert-Butyl (2S)-2-(2-(6-bromo-4-chloro-7-fluoro-2H-indazol-2-yl)-3-ethoxy-3-oxopropanoyl)pyrrolidine-1-carboxylate
  • In analogy to Example 1, step 1, (tert-butoxycarbonyl)-L-proline was treated with carbonyldiimidazole to give solution A. Ethyl 2-(6-bromo-4-chloro-7-fluoro-2H-indazol-2-yl)acetate was deprotonated with LDA and treated with solution A at −78° C. After stirring at room temperature overnight and workup in analogy to Example 1, step 1, the crude title compound was obtained which was used for the next step without further purification.
  • Step 2: Ethyl 2-(6-bromo-4-chloro-7-fluoro-2H-indazol-2-yl)-2-(3-thioxo-2,5,6,7-tetrahydro-3H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • In analogy to Example 1, step 2, tert-butyl (2S)-2-(2-(6-bromo-4-chloro-7-fluoro-2H-indazol-2-yl)-3-ethoxy-3-oxopropanoyl)pyrrolidine-1-carboxylate was deprotected using HCl in dioxane followed by reaction with potassium thiocyanate to give the crude title compound which was used for the next step without further purification. MS: m/e=475.1 ([M+H]+)
  • Step 3: Ethyl 2-(6-bromo-4-chloro-7-fluoro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • In analogy to Example 1, step 3, ethyl 2-(6-bromo-4-chloro-7-fluoro-2H-indazol-2-yl)-2-(3-thioxo-2,5,6,7-tetrahydro-3H-pyrrolo[1,2-c]imidazol-1-yl)acetate was treated with hydrogen peroxide in AcOH to give the title compound as light yellow solid. MS: m/e=443.2 ([M+H]+, Br)
  • Step 4: tert-Butyl 4-(4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-ethoxy-2-oxoethyl)-7-fluoro-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • In analogy to Example 1, step 4, ethyl 2-(6-bromo-4-chloro-7-fluoro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate was treated with (4-(4-(tert-butoxycarbonyl)piperazin-1-yl)phenyl)boronic acid in toluene in the presence of cesium carbonate and [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) to give the title compound as off-white solid. MS: m/e=623.4 ([M+H]+)
  • Step 5: tert-Butyl 4-(4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-7-fluoro-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • In analogy to Example 1, step 5, tert-butyl 4-(4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-ethoxy-2-oxoethyl)-7-fluoro-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate was first treated with LiOH and the resulting acid salt was reacted with thiazol-2-amine in the presence of HATU and Hunig's base to give the title compound as off-white solid. MS: m/e=675.5 ([M−H])
  • Step 6: 2-(4-Chloro-7-fluoro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride
  • In analogy to Example 1, step 6, a solution of tert-butyl 4-(4-(4-chloro-2-(1-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-7-fluoro-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate in dichloromethane and methanol was treated with HCl in dioxane to give the title compound as off-white solid. MS: m/e=575.4 ([M−H])
  • Example 16
  • Figure US20220135571A1-20220505-C00012
  • 2-(4-Chloro-6-(4-(4-ethylpiperazin-1-yl)phenyl)-7-fluoro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide
  • In analogy to Example 4, 2-(4-chloro-7-fluoro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride (Example 15) was treated with iodoethane in the presence of triethylamine to give the title compound as colorless solid. MS: m/e=603.5 ([M−H])
  • Example 18
  • Figure US20220135571A1-20220505-C00013
  • 2-(4-Chloro-7-fluoro-6-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide Step 1: Ethyl 2-(4-chloro-7-fluoro-6-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • In analogy to Example 1, step 4, ethyl 2-(6-bromo-4-chloro-7-fluoro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate (Example 15, step 3) was reacted with 1-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,4-dihydroquinolin-2(1H)-one in toluene in the presence of [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II), cesium carbonate and 10 equivalents of water to give the title compound as off-white solid. MS: m/e=522.3 ([M+H]+)
  • Step 2: 2-(4-Chloro-7-fluoro-6-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide
  • In analogy to Example 1, step 5, ethyl 2-(4-chloro-7-fluoro-6-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate was first treated with LiOH and the resulting acid salt was reacted with thiazol-2-amine in the presence of HATU and Hunig's base to give the title compound as off-white solid. MS: m/e=576.2 ([M+H]+)
  • Example 20
  • Figure US20220135571A1-20220505-C00014
  • 2-[4-Chloro-7-methoxy-6-(1-methyl-2-oxo-3,4-dihydroquinolin-6-yl)indazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide Step 1: tert-Butyl (2S,4R)-2-(2-(6-bromo-4-chloro-7-methoxy-2H-indazol-2-yl)-3-ethoxy-3-oxopropanoyl)-4-fluoropyrrolidine-1-carboxylate
  • In analogy to Example 1, step 1, (2S,4R)-1-(tert-butoxycarbonyl)-4-fluoropyrrolidine-2-carboxylic acid was treated with carbonyldiimidazole to give solution A. Ethyl 2-(6-bromo-4-chloro-7-methoxy-2H-indazol-2-yl)acetate was deprotonated with LDA and treated with solution A at −78° C. After stirring at room temperature for 90 min and workup in analogy to Example 1, step 1, the crude title compound was obtained which was used for the next step without further purification. MS: m/e=564.2 ([M+H]+, Br)
  • Step 2: Ethyl 2-(6-bromo-4-chloro-7-methoxy-2H-indazol-2-yl)-2-((R)-6-fluoro-3-thioxo-2,5,6,7-tetrahydro-3H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • In analogy to Example 1, step 2, tert-butyl (2S,4R)-2-(2-(6-bromo-4-chloro-7-methoxy-2H-indazol-2-yl)-3-ethoxy-3-oxopropanoyl)-4-fluoropyrrolidine-1-carboxylate was deprotected using HCl in dioxane followed by reaction with potassium thiocyanate to give the crude title compound which was used for the next step without further purification. MS: m/e=505.1 ([M+H]+, Br)
  • Step 3: Ethyl 2-(6-bromo-4-chloro-7-methoxy-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • In analogy to Example 1, step 3, ethyl 2-(6-bromo-4-chloro-7-methoxy-2H-indazol-2-yl)-2-((R)-6-fluoro-3-thioxo-2,5,6,7-tetrahydro-3H-pyrrolo[1,2-c]imidazol-1-yl)acetate was treated with hydrogen peroxide in AcOH to give the title compound as light brown solid. MS: m/e=472.9 ([M+H]+, Br)
  • Step 4: Ethyl 2-(4-chloro-7-methoxy-6-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • In analogy to Example 1, step 4, ethyl 2-(6-bromo-4-chloro-7-methoxy-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate (Example 19, step 3) was reacted with 1-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,4-dihydroquinolin-2(1H)-one in toluene in the presence of [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II), cesium carbonate and 10 equivalents of water to give the title compound as off-white solid. MS: m/e=552.3 ([M+H]+)
  • Step 5: 2-[4-Chloro-7-methoxy-6-(1-methyl-2-oxo-3,4-dihydroquinolin-6-yl)indazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide
  • In analogy to Example 1, step 5, ethyl 2-(4-chloro-7-methoxy-6-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate was first treated with LiOH and the resulting acid salt was reacted with thiazol-2-amine in the presence of HATU and Hunig's base to give the title compound as off-white solid. MS: m/e=606.2 ([M+H]+)
  • Example 21
  • Figure US20220135571A1-20220505-C00015
  • 2-[4-Chloro-6-[4-(4-ethylpiperazin-1-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide Step 1: tert-Butyl 4-(4-(4-chloro-2-(2-ethoxy-1-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxoethyl)-7-methoxy-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • In analogy to Example 1, step 4, ethyl 2-(6-bromo-4-chloro-7-methoxy-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate (Example 20, step 3) was reacted with (4-(4-(tert-butoxycarbonyl)piperazin-1-yl)phenyl)boronic acid in toluene in the presence of [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) and cesium carbonate to give the title compound as off-white solid. MS: m/e=653.4 ([M+H]+)
  • Step 2: tert-Butyl 4-(4-(4-chloro-2-(1-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-7-methoxy-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate
  • In analogy to Example 1, step 5, tert-butyl 4-(4-(4-chloro-2-(2-ethoxy-1-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxoethyl)-7-methoxy-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate was first treated with LiOH and the resulting acid salt was reacted with thiazol-2-amine in the presence of HATU and Hunig's base to give the title compound as off-white solid. MS: m/e=705.5 ([M−H])
  • Step 3: 2-(4-Chloro-7-methoxy-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride
  • In analogy to Example 1, step 6, a solution of tert-butyl 4-(4-(4-chloro-2-(1-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-2-oxo-2-(thiazol-2-ylamino)ethyl)-7-methoxy-2H-indazol-6-yl)phenyl)piperazine-1-carboxylate in dichloromethane and methanol was treated with HCl in dioxane to give the title compound as off-white solid. MS: m/e=605.5 ([M−H])
  • Step 4: 2-[4-Chloro-6-[4-(4-ethylpiperazin-1-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide
  • In analogy to Example 4, 2-(4-chloro-7-methoxy-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride was treated with iodoethane in the presence of triethylamine to give the title compound as off-white solid. MS: m/e=633.4 ([M−H])
  • Example 22
  • Figure US20220135571A1-20220505-C00016
  • 2-[4-Chloro-6-[4-(1-cyclopropylpiperidin-4-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide Step 1: Ethyl 2-(4-chloro-6-(4-(1-cyclopropylpiperidin-4-yl)phenyl)-7-methoxy-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • In analogy to Example 1, step 4, ethyl 2-(6-bromo-4-chloro-7-methoxy-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate (Example 20, step 3) was reacted with 1-cyclopropyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine (CAS #2244702-81-4) in THF and water in the presence of [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) and cesium carbonate to give the title compound as light brown solid. MS: m/e=592.4 ([M+H]+)
  • Step 2: 2-[4-Chloro-6-[4-(1-cyclopropylpiperidin-4-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide
  • In analogy to Example 1, step 5, ethyl 2-(4-chloro-6-(4-(1-cyclopropylpiperidin-4-yl)phenyl)-7-methoxy-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate was first treated with LiOH and the resulting acid salt was reacted with thiazol-2-amine in the presence of HATU and Hunig's base to give the title compound as off-white solid. MS: m/e=644.4 ([M−H])
  • Example 23
  • Figure US20220135571A1-20220505-C00017
  • 2-(4-Chloro-7-fluoro-6-(1-isopropyl-2-oxo-1,2-dihydropyridin-4-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide Step 1: Ethyl 2-(4-chloro-7-fluoro-6-(1-isopropyl-2-oxo-1,2-dihydropyridin-4-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate
  • In analogy to Example 1, step 4, ethyl 2-(6-bromo-4-chloro-7-fluoro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate (Example 15, step 3) was reacted with 1-isopropyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2(1H)-one in toluene in the presence of [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II), cesium carbonate and equivalents of water to give the title compound as off-white solid. MS: m/e=498.4 ([M+H]+)
  • Step 2: 2-(4-Chloro-7-fluoro-6-(1-isopropyl-2-oxo-1,2-dihydropyridin-4-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide
  • In analogy to Example 1, step 5, ethyl 2-(4-chloro-7-fluoro-6-(1-isopropyl-2-oxo-1,2-dihydropyridin-4-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)acetate was first treated with LiOH and the resulting acid salt was reacted with thiazol-2-amine in the presence of HATU and Hunig's base to give the title compound as off-white solid. MS: m/e=550.4 ([M−H]).

Claims (8)

We claim:
1. A compound selected from the group consisting of
2-(4-chloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(6-(4-(4-acetylpiperazin-1-yl)phenyl)-4-chloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4-chloro-6-(3-chloro-4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(4-chloro-6-(3-chloro-4-(4-ethylpiperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(6-(3-chloro-4-(piperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(6-(4-(4-acetylpiperazin-1-yl)-3-chlorophenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(6-(3-chloro-4-(4-ethylpiperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4,7-dichloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(6-(4-(4-acetylpiperazin-1-yl)phenyl)-4,7-dichloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4,7-dichloro-6-(4-(4-ethylpiperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4-chloro-7-fluoro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(4-chloro-6-(4-(4-ethylpiperazin-1-yl)phenyl)-7-fluoro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4-chloro-7-fluoro-6-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-[4-chloro-7-methoxy-6-(1-methyl-2-oxo-3,4-dihydroquinolin-6-yl)indazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide;
2-[4-chloro-6-[4-(4-ethylpiperazin-1-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide;
2-[4-chloro-6-[4-(1-cyclopropylpiperidin-4-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide; and
2-(4-chloro-7-fluoro-6-(1-isopropyl-2-oxo-1,2-dihydropyridin-4-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
or a pharmaceutically acceptable salt thereof.
2. The compound of claim 1 selected from the group consisting of
2-(4-chloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(6-(4-(4-acetylpiperazin-1-yl)phenyl)-4-chloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4-chloro-6-(3-chloro-4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(4-chloro-6-(3-chloro-4-(4-ethylpiperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(6-(3-chloro-4-(piperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(6-(4-(4-acetylpiperazin-1-yl)-3-chlorophenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(6-(3-chloro-4-(4-ethylpiperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4,7-dichloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(6-(4-(4-acetylpiperazin-1-yl)phenyl)-4,7-dichloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4,7-dichloro-6-(4-(4-ethylpiperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4-chloro-7-fluoro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(4-chloro-6-(4-(4-ethylpiperazin-1-yl)phenyl)-7-fluoro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4-chloro-7-fluoro-6-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-[4-chloro-7-methoxy-6-(1-methyl-2-oxo-3,4-dihydroquinolin-6-yl)indazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide;
2-[4-chloro-6-[4-(4-ethylpiperazin-1-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide;
2-[4-chloro-6-[4-(1-cyclopropylpiperidin-4-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide; and
2-(4-chloro-7-fluoro-6-(1-isopropyl-2-oxo-1,2-dihydropyridin-4-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide.
3. A pharmaceutical composition comprising a compound of claim 1 or a pharmaceutically acceptable salt thereof and a therapeutically inert carrier.
4. The pharmaceutical composition of claim 3, wherein the compound is selected from the group consisting of
2-(4-chloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(6-(4-(4-acetylpiperazin-1-yl)phenyl)-4-chloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4-chloro-6-(3-chloro-4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(4-chloro-6-(3-chloro-4-(4-ethylpiperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(6-(3-chloro-4-(piperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(6-(4-(4-acetylpiperazin-1-yl)-3-chlorophenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(6-(3-chloro-4-(4-ethylpiperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4,7-dichloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(6-(4-(4-acetylpiperazin-1-yl)phenyl)-4,7-dichloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4,7-dichloro-6-(4-(4-ethylpiperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4-chloro-7-fluoro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(4-chloro-6-(4-(4-ethylpiperazin-1-yl)phenyl)-7-fluoro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4-chloro-7-fluoro-6-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-[4-chloro-7-methoxy-6-(1-methyl-2-oxo-3,4-dihydroquinolin-6-yl)indazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide;
2-[4-chloro-6-[4-(4-ethylpiperazin-1-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide;
2-[4-chloro-6-[4-(1-cyclopropylpiperidin-4-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide; and
2-(4-chloro-7-fluoro-6-(1-isopropyl-2-oxo-1,2-dihydropyridin-4-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide.
5. A method for the treatment or prophylaxis of cancer, which method comprises administering an effective amount of a compound according to claim 1 or a pharmaceutically acceptable salt thereof.
6. The method of claim 5 wherein the compound is selected from the group consisting of
2-(4-chloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(6-(4-(4-acetylpiperazin-1-yl)phenyl)-4-chloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4-chloro-6-(3-chloro-4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(4-chloro-6-(3-chloro-4-(4-ethylpiperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(6-(3-chloro-4-(piperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(6-(4-(4-acetylpiperazin-1-yl)-3-chlorophenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(6-(3-chloro-4-(4-ethylpiperazin-1-yl)phenyl)-4-fluoro-2H-indazol-2-yl)-2-((R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4,7-dichloro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(6-(4-(4-acetylpiperazin-1-yl)phenyl)-4,7-dichloro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4,7-dichloro-6-(4-(4-ethylpiperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4-chloro-7-fluoro-6-(4-(piperazin-1-yl)phenyl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide hydrochloride;
2-(4-chloro-6-(4-(4-ethylpiperazin-1-yl)phenyl)-7-fluoro-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-(4-chloro-7-fluoro-6-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide;
2-[4-chloro-7-methoxy-6-(1-methyl-2-oxo-3,4-dihydroquinolin-6-yl)indazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide;
2-[4-chloro-6-[4-(4-ethylpiperazin-1-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide;
2-[4-chloro-6-[4-(1-cyclopropylpiperidin-4-yl)phenyl]-7-methoxyindazol-2-yl]-2-[rac-(6R)-6-fluoro-6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl]-N-(1,3-thiazol-2-yl)acetamide; and
2-(4-chloro-7-fluoro-6-(1-isopropyl-2-oxo-1,2-dihydropyridin-4-yl)-2H-indazol-2-yl)-2-(6,7-dihydro-5H-pyrrolo[1,2-c]imidazol-1-yl)-N-(thiazol-2-yl)acetamide.
7. The method of claim 5, wherein the cancer is a non-small cell lung cancer.
8. The method of claim 6, wherein the cancer is a non-small cell lung cancer.
US17/558,053 2019-06-21 2021-12-21 Therapeutic egfr inhibitors Pending US20220135571A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP19181754 2019-06-21
EP19181754.3 2019-06-21
PCT/EP2020/067082 WO2020254565A1 (en) 2019-06-21 2020-06-19 Egfr inhibitors for the treatment of cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2020/067082 Continuation WO2020254565A1 (en) 2019-06-21 2020-06-19 Egfr inhibitors for the treatment of cancer

Publications (1)

Publication Number Publication Date
US20220135571A1 true US20220135571A1 (en) 2022-05-05

Family

ID=67139601

Family Applications (4)

Application Number Title Priority Date Filing Date
US17/620,239 Pending US20220315592A1 (en) 2019-06-21 2020-06-19 Egfr inhibitor for the treatment of cancer
US17/620,242 Pending US20230054473A1 (en) 2019-06-21 2020-06-19 Egfr inhibitors for the treatment of cancer
US17/620,240 Pending US20220315593A1 (en) 2019-06-21 2020-06-19 Egfr inhibitors for the treatment of cancer
US17/558,053 Pending US20220135571A1 (en) 2019-06-21 2021-12-21 Therapeutic egfr inhibitors

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US17/620,239 Pending US20220315592A1 (en) 2019-06-21 2020-06-19 Egfr inhibitor for the treatment of cancer
US17/620,242 Pending US20230054473A1 (en) 2019-06-21 2020-06-19 Egfr inhibitors for the treatment of cancer
US17/620,240 Pending US20220315593A1 (en) 2019-06-21 2020-06-19 Egfr inhibitors for the treatment of cancer

Country Status (28)

Country Link
US (4) US20220315592A1 (en)
EP (4) EP3986895B1 (en)
JP (4) JP2022537191A (en)
KR (1) KR20220024105A (en)
CN (4) CN113993873A (en)
AR (4) AR119210A1 (en)
AU (1) AU2020295703A1 (en)
BR (1) BR112021025445B1 (en)
CA (1) CA3140485A1 (en)
CL (1) CL2021003408A1 (en)
CO (1) CO2021017173A2 (en)
CR (1) CR20210624A (en)
DK (1) DK3986897T3 (en)
ES (1) ES2961187T3 (en)
FI (1) FI3986897T3 (en)
HR (1) HRP20231276T1 (en)
HU (1) HUE063676T2 (en)
IL (1) IL288522A (en)
LT (1) LT3986897T (en)
MA (1) MA56508B1 (en)
MX (1) MX2021015464A (en)
PE (1) PE20220576A1 (en)
PL (1) PL3986897T3 (en)
PT (1) PT3986897T (en)
RS (1) RS64720B1 (en)
SI (1) SI3986897T1 (en)
TW (4) TW202115072A (en)
WO (4) WO2020254568A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023217924A1 (en) * 2022-05-13 2023-11-16 F. Hoffmann-La Roche Ag Combination of allosteric and orthosteric egfr inhibitors for the treatment of cancer
WO2023217923A1 (en) * 2022-05-13 2023-11-16 F. Hoffmann-La Roche Ag New indazole derivatives

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0508715D0 (en) * 2005-04-29 2005-06-08 Astrazeneca Ab Chemical compounds
CN101273033A (en) * 2005-09-20 2008-09-24 阿斯利康(瑞典)有限公司 4- (iH-indazol-s-yl-amino)-quinazoline compounds as ERBB receptor tyrosine kinase inhibitors for the treatment of cancer
US20110212078A1 (en) 2008-06-25 2011-09-01 Reddy Panduranga Adulla P Synthesis and use of heterocyclic antibacterial agents
US8420661B2 (en) 2010-04-13 2013-04-16 Hoffmann-La Roche Inc. Arylethynyl derivatives
WO2015095807A1 (en) * 2013-12-20 2015-06-25 Biomed Valley Discoveries, Inc. Cancer treatments using combinations of egfr and erk inhibitors
WO2016000615A1 (en) * 2014-07-02 2016-01-07 Sunshine Lake Pharma Co., Ltd. Heteroaryl compounds and pharmaceutical applications thereof
JP6639497B2 (en) * 2014-11-10 2020-02-05 ジェネンテック, インコーポレイテッド Bromodomain inhibitors and uses thereof
CN107848964B (en) 2015-05-14 2021-03-19 威斯塔解剖学和生物学研究所 EBNA1 inhibitors and methods of using the same
TW201834651A (en) * 2016-12-22 2018-10-01 瑞士商赫孚孟拉羅股份公司 Compounds
AR113299A1 (en) * 2017-06-02 2020-04-08 Hoffmann La Roche ISOINDOLINE COMPOUNDS
WO2019046668A1 (en) * 2017-08-31 2019-03-07 Kezar Life Sciences Amide substituted thiazoles as protein secretion inhibitors
KR20210025535A (en) * 2018-06-29 2021-03-09 에프. 호프만-라 로슈 아게 compound

Also Published As

Publication number Publication date
EP3986897A1 (en) 2022-04-27
MA56508A (en) 2022-04-27
FI3986897T3 (en) 2023-10-20
TW202115053A (en) 2021-04-16
HRP20231276T1 (en) 2024-02-02
EP3986898B1 (en) 2023-03-15
PE20220576A1 (en) 2022-04-20
TW202115074A (en) 2021-04-16
WO2020254565A1 (en) 2020-12-24
JP2022537769A (en) 2022-08-29
LT3986897T (en) 2023-11-10
WO2020254562A1 (en) 2020-12-24
EP3986898A1 (en) 2022-04-27
AU2020295703A1 (en) 2022-01-06
CN113993872A (en) 2022-01-28
TW202115072A (en) 2021-04-16
TW202115071A (en) 2021-04-16
US20220315593A1 (en) 2022-10-06
JP2022537191A (en) 2022-08-24
CN113993873A (en) 2022-01-28
AR119210A1 (en) 2021-12-01
CO2021017173A2 (en) 2022-01-17
PT3986897T (en) 2023-10-23
SI3986897T1 (en) 2023-12-29
KR20220024105A (en) 2022-03-03
CN113966335A (en) 2022-01-21
RS64720B1 (en) 2023-11-30
CA3140485A1 (en) 2020-12-24
AR119209A1 (en) 2021-12-01
WO2020254568A1 (en) 2020-12-24
ES2961187T3 (en) 2024-03-08
US20220315592A1 (en) 2022-10-06
WO2020254572A1 (en) 2020-12-24
CN114008049A (en) 2022-02-01
JP2022538055A (en) 2022-08-31
MA56508B1 (en) 2023-11-30
BR112021025445A2 (en) 2022-02-01
JP2022538403A (en) 2022-09-02
MX2021015464A (en) 2022-01-24
AR119208A1 (en) 2021-12-01
DK3986897T3 (en) 2023-10-30
EP3986895A1 (en) 2022-04-27
EP3986897B1 (en) 2023-08-30
BR112021025445B1 (en) 2024-02-27
IL288522A (en) 2022-01-01
CR20210624A (en) 2022-02-14
PL3986897T3 (en) 2024-01-22
EP3986895B1 (en) 2023-03-15
HUE063676T2 (en) 2024-01-28
CL2021003408A1 (en) 2022-10-14
AR119206A1 (en) 2021-12-01
EP3986896B1 (en) 2023-03-15
US20230054473A1 (en) 2023-02-23
EP3986896A1 (en) 2022-04-27

Similar Documents

Publication Publication Date Title
US20210079005A1 (en) Compounds
US20220135571A1 (en) Therapeutic egfr inhibitors
US20240059692A1 (en) New indazole derivatives
US20240002390A1 (en) New indazole acetylene derivatives
EP3986567B1 (en) New egfr inhibitors
EP4076664B1 (en) Egfr inhibitors
EP4076665B1 (en) Egfr inhibitors
EP3986565B1 (en) New egfr inhibitors

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: HOFFMANN-LA ROCHE INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:F. HOFFMANN-LA ROCHE AG;REEL/FRAME:062352/0724

Effective date: 20200616

Owner name: F. HOFFMANN-LA ROCHE AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:JAESCHKE, GEORG;STEINER, SANDRA;HEWINGS, DAVID;AND OTHERS;SIGNING DATES FROM 20200212 TO 20200421;REEL/FRAME:062358/0166