US20190076436A1 - Macrocyclic compounds as ros1 kinase inhibitors - Google Patents

Macrocyclic compounds as ros1 kinase inhibitors Download PDF

Info

Publication number
US20190076436A1
US20190076436A1 US16/199,867 US201816199867A US2019076436A1 US 20190076436 A1 US20190076436 A1 US 20190076436A1 US 201816199867 A US201816199867 A US 201816199867A US 2019076436 A1 US2019076436 A1 US 2019076436A1
Authority
US
United States
Prior art keywords
ros1
cancer
amino acid
alkyl
kinase
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.)
Abandoned
Application number
US16/199,867
Other languages
English (en)
Inventor
Steven W. Andrews
James F. Blake
Julia Haas
Gabrielle R. Kolakowski
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.)
Array Biopharma Inc
Original Assignee
Array Biopharma 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 Array Biopharma Inc filed Critical Array Biopharma Inc
Priority to US16/199,867 priority Critical patent/US20190076436A1/en
Publication of US20190076436A1 publication Critical patent/US20190076436A1/en
Assigned to ARRAY BIOPHARMA INC. reassignment ARRAY BIOPHARMA INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HAAS, JULIA, BLAKE, JAMES F., ANDREWS, STEVEN W., KOLAKOWSKI, Gabrielle R.
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/529Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim forming part of bridged ring 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/91Transferases (2.)
    • G01N2333/912Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • compositions comprising the compounds and the use of the compounds in therapy. More particularly, provided herein are certain macrocyclic compounds which exhibit ROS1 protein kinase inhibition, and which are useful in the treatment of cancer.
  • ROS1 is a receptor tyrosine kinase that is closely related to ALK, and, like ALK, it undergoes genomic rearrangement that creates fusion proteins in various cancers (Davies K D and Doebele R C (2013) Clin Cancer Res 19: 4040-4045). It is well established that these fusion proteins act as oncogenic drivers and that ROS1 inhibition is anti-proliferative in cells that express ROS1 fusions (Davies K D, Le A T, Theodoro M F, Skokan M C, Aisner D L, et al. (2012) Clin Cancer Res 18: 4570-4579). Thus, it appears that ROS1 targeted therapy will likely soon be the standard of care for this patient population. However, based on the experiences with other kinase inhibitors in various cancers, it is fully expected that acquired resistance to ROS1 inhibition will occur, and this will ultimately limit the treatment options for patients.
  • macrocyclic compounds are inhibitors of ROS1 kinase, and are useful for treating various cancers.
  • Compounds which are inhibitors of ROS1 may be useful in the treatment of multiple types of cancer including cancers exhibiting resistance to ROS1 inhibition.
  • the methods provided include administration of a ROS1 inhibitor, wherein the ROS1 inhibitor is a compound of Formula I
  • ring A, ring B, W, m, D, R 2 , R 2a , R 3 , R 3a , and Z are as defined herein.
  • a compound of Formula I has the general formula:
  • the compound of Formula I is selected from the compounds of Table 1, or a pharmaceutically acceptable salt or solvate thereof. In some embodiments, the compound of Formula I is selected from the group consisting of Example No. 2, 3, 7, 9, 14, 19, 20, 22, 33-A, 33-B, 35, 36, and 45, or a pharmaceutically acceptable salt or solvate thereof.
  • a method for treating a cancer in a patient in need thereof comprising:
  • a method for treating a cancer in a patient in need thereof comprising:
  • Also provided herein is a method for treating cancer in a patient in need thereof, the method comprising administering to a patient identified or diagnosed as having a ROS1-associated cancer a therapeutically effective amount of a ROS1 inhibitor, wherein the ROS1 inhibitor is a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • a method of treating cancer in a patient in need thereof comprising:
  • a method of treating cancer in a patient in need thereof comprising:
  • Also provided herein is a method of treating a subject having a cancer, wherein the method comprises:
  • a method of treating a subject having a cancer comprising:
  • Also provided herein is a method of treating a subject having a cancer, wherein the method comprises:
  • a method of treating a subject having a cancer comprising:
  • Also provided herein is a method of treating a subject having a cancer, wherein the method comprises:
  • a method of treating a subject having a cancer comprising:
  • Also provided herein is a method of treating a patient, the method comprising administering a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt or solvate thereof, to a patient having a clinical record that indicates that the patient has a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same.
  • a method of selecting a treatment for a patient comprising selecting a treatment comprising administration of a compound of Formula I, or a pharmaceutically acceptable salt or solvate thereof, for a patient identified or diagnosed as having a ROS1-associated cancer.
  • a method of selecting a treatment for a patient having a cancer comprising:
  • Also provided herein is a method of selecting a patient for treatment including administration of a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt or solvate thereof, the method comprising:
  • a method of selecting a patient having cancer for treatment including administration of a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt or solvate thereof, the method comprising:
  • ring B is ring B-2 having the structure:
  • ring A is ring A-1 having the structure
  • ring A when represented by structure A-1 include the structures:
  • ring A is ring A-2 having the structure
  • ring A when represented by ring A-2 are the structures:
  • ring A is ring A-3 having the structure
  • ring A when represented by ring A-3 are the structures:
  • W is O.
  • W is NH
  • W is CH 2 .
  • D is carbon
  • R 2 and R 2a are independently H, F, (1-3 C)alkyl or OH (provided that R 2 and R 2a are not both OH)
  • R 3 and R 3a are independently H, (1-3 C)alkyl or hydroxy(1-3 C)alkyl.
  • R 2 and R 2a are independently H, F, methyl or OH, provided that R 2 and R 2a are not both OH.
  • R 2 and R 2a are both H.
  • R 2 is H and R 2a is F.
  • R 2 and R 2a are both F.
  • R 2 is H and R 2a is OH.
  • R 2 is H and R 2a is methyl.
  • R 2 and R 2a are both methyl.
  • R 3 and R 3a are independently H, (1-3C)alkyl or hydroxy(1-3 C)alkyl.
  • R 3a is H. In some embodiments, R 3 is H. In some embodiments, both R 3 and R 3a are H.
  • R 3a is (1-3C)alkyl. Examples include methyl, ethyl, propyl and isopropyl. In some embodiments, R 3 is (1-3C)alkyl. Examples include methyl, ethyl, propyl and isopropyl.
  • R 3a is (1-3C)alkyl and R 3 is H. In some embodiments, R 3a is methyl and R 3 is H.
  • both R 3a and R 3 are (1-3C)alkyl. In some embodiments, R 3a and R 3a are both methyl.
  • R 3 is hydroxy(1-3C)alkyl. Examples include hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl, and 3-hydroxypropyl. In some embodiments, R 3 is hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl, or 3-hydroxypropyl and R 3a is H.
  • D is carbon or nitrogen
  • R 2 and R 3 are absent
  • R 2a and R 3a together with the atoms to which they are attached form a 5-6 membered heteroaryl ring having 1-2 ring heteroatoms.
  • R 2a and R 3a together with the atoms to which they are attached form a 5-6 membered heteroaryl ring having 1-2 ring nitrogen atoms.
  • heteroaryl rings include pyridyl and pyrazolyl rings. Specific examples of heteroaryl rings include the structures:
  • Z is *—NR 4a C( ⁇ O)—.
  • R 4a is H.
  • R 4a is (1-6C)alkyl. Examples include methyl, ethyl, propyl, isopropyl, butyl, and isobutyl.
  • R 4a is fluoro(1-6C)alkyl. Examples include fluoromethyl and 2-fluoroethyl.
  • R 4a is difluoro(1-6C)alkyl.
  • Example include difluoromethyl and 2,2-difluoroethyl.
  • R 4a is trifluoro(1-6C)alkyl. Examples include trifluoromethyl and 2,2,2-trifluoroethyl.
  • R 4a is hydroxy(1-6C alkyl). Examples include hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl and 3-hydroxypropyl.
  • R 4a is dihydroxy(2-6C alkyl).
  • An example includes 2,3-dihydroxypropyl.
  • R 4a is H or (1-6C)alkyl. In some embodiments, R 4a is H or Me.
  • Z is *—NR 4b CH 2 —.
  • R 4b is H.
  • R 4b is selected from (1-6C)alkyl, fluoro(1-6C)alkyl, difluoro(1-6C)alkyl, and trifluoro(1-6C)alkyl.
  • R 4b is (1-6C)alkyl. Examples include methyl, ethyl, propyl, isopropyl, butyl and tert-butyl. In some embodiments, R 4b is methyl.
  • R 4b is fluoro(1-6C)alkyl. Examples include fluoromethyl and 2-fluoroethyl.
  • R 4b is difluoro(1-6C)alkyl.
  • Example include difluoromethyl and 2,2-difluoroethyl.
  • R 4b is trifluoro(1-6C)alkyl. Examples include trifluoromethyl and 2,2,2-trifluoroethyl.
  • R 4b is selected from (1-6C alkyl)C(O)—, (3-6C cycloalkyl)C(O)—, Ar 1 C(O)— and HOCH 2 C(O)—.
  • R 4b is (1-6C alkyl)C(O)—. Examples include CH 3 C(O)—, CH 3 CH 2 C(O)—, CH 3 CH 2 CH 2 C(O)—, and (CH 3 ) 2 CHC(O)—. In some embodiments, R 4 is CH 3 C(O)—.
  • R 4b is (3-6C cycloalkyl)C(O)—. Examples include cyclopropylC(O)—, cyclobutylC(O)—, cyclopentylC(O)— and cyclohexylC(O)—.
  • R 4b is Ar 1 C(O)—.
  • An example is phenylC(O)—.
  • R 4b is HOCH 2 C(O)—.
  • R 4b is selected from (1-6C alkyl)sulfonyl, (3-6C cycloalkyl)sulfonyl, and Ar 2 (SO 2 )—.
  • R 4b is (1-6C alkyl)sulfonyl. Examples include methylsulfonyl, ethylsulfonyl and propylsulfonyl.
  • R 4b is (3-6C cycloalkyl)sulfonyl. Examples include cyclopropylsulfonyl, cyclobutylsulfonyl, cyclopentylsulfonyl and cyclohexylsulfonyl. In some embodiments, R 4 is methylsulfonyl.
  • R 4b is Ar 2 (SO 2 )—.
  • An example is phenylsulfonyl.
  • R 4b is HO 2 CCH 2 —.
  • R 4b is (1-6C alkyl)NH(CO)—. Examples include CH 3 NHC(O)—, CH 3 CH 2 NHC(O)—, CH 3 CH 2 CH 2 NHC(O)—, and (CH 3 ) 2 CHNHC(O)—. In some embodiments, R 4 is CH 3 NHC(O)—.
  • R 4b is selected from H, methyl, —C(O)CH 3 , methylsulfonyl, —C(O)CH 2 OH, —CH 2 COOH and —C(O)NHCH 2 CH 3 .
  • Z is *—OC( ⁇ O)—.
  • ring B is ring B-1:
  • R 5 and R 6 are independently H, F, OH, (1-6C)alkyl or hydroxy(1-6C)alkyl. In some embodiments, R 5 is H and R 6 is H, F, OH, (1-6C)alkyl or hydroxy(1-6C)alkyl.
  • R 5 and R 6 are independently H, F, OH, (1-3C)alkyl or hydroxy(1-3C)alkyl. In some embodiments, R 5 is hydrogen and R 6 is H, F, OH, (1-3C)alkyl or hydroxy(1-3C)alkyl.
  • R 5 and R 6 are independently H, F, OH, methyl, ethyl, HOCH 2 — or HOCH 2 CH 2 —. In some embodiments, R 5 is hydrogen and R 6 is H, F, OH, methyl, ethyl, HOCH 2 — or HOCH 2 CH 2 —.
  • R 5 and R 6 are independently H, F, or methyl. In some embodiments, R 5 is H and R 6 is H, F, or methyl.
  • R 5 is H and R 6 is F.
  • R 5 is H and R 6 is methyl.
  • R 5 and R 6 are both H.
  • R 5 and R 6 are both F.
  • R 5 and R 6 are both methyl.
  • ring B is ring B-1 which is optionally substituted with one or two substituents independently selected from OH and F, provided that two OH substituents are not on the same ring carbon atom.
  • ring B when represented by ring B-1 include the structures:
  • ring B is ring B-2 having the formula:
  • m is 0.
  • m is 1.
  • n is 2.
  • Formula IA includes compounds wherein:
  • X is N. In some embodiments, X is CH.
  • Formula IA includes compounds wherein:
  • Formula IA includes compounds wherein:
  • Formula IA includes compounds wherein:
  • Formula IA includes compounds wherein:
  • X is N. In some such embodiments of Formula IA where ring A is ring A-1, W is O. In some embodiments of Formula IA where ring A is ring A-1, W is CH 2 . In some embodiments of Formula IA where ring A is ring A-1, R 2 and R 2a are H. In some embodiments of Formula IA where ring A is ring A-1, R 2 and R 2a are independently F, (1-3 C)alkyl, or OH. In some embodiments of Formula IA where ring A is ring A-1, R 3 is (1-3 C)alkyl. In some embodiments of Formula IA where ring A is ring A-1, R 3 is H. In some embodiments of Formula IA where ring A is ring A-1, Z is *—NR 4a C( ⁇ O)—. In some embodiments of Formula IA where ring A is ring A-1, R 5 and R 6 are H.
  • Formula IA includes compounds wherein:
  • Y is F.
  • R 2 and R 2a are H.
  • R 2 and R 2a are independently H or (1-3 C)alkyl.
  • R 3 is (1-3 C)alkyl.
  • R 5 and R 6 are H.
  • Formula IA includes compounds wherein:
  • Y is F. In some embodiments of Formula IA where ring A is ring A-3, Y is H. In some embodiments of Formula IA where ring A is ring A-3, R 2 and R 2a are H. In some embodiments of Formula IA where ring A is ring A-3, R 2 and R 2a are independently H or (1-3 C)alkyl. In some embodiments of Formula IA where ring A is ring A-3, R 3 is (1-3 C)alkyl. In some embodiments of Formula IA where ring A is ring A-3, R 3 is H. In some embodiments of Formula IA where ring A is ring A-3, R 5 and R 6 are H.
  • certain compounds as provided herein may contain one or more centers of asymmetry and may therefore be prepared and isolated as a mixture of isomers such as a racemic or diastereomeric mixture, or in an enantiomerically or diastereomerically pure form. It is intended that all stereoisomeric forms of the compounds provided herein, including but not limited to, diastereomers, enantiomers and atropisomers, as well as mixtures thereof such as racemic mixtures, form part of the present disclosure.
  • compounds of the general Formula I wherein Ring B is ring B-1 have the absolute configuration of Formula 1-a:
  • compounds of the general Formula I wherein Ring B is ring B-1 have the absolute configuration of Formula 1-b:
  • stereochemistry of any particular chiral atom is not specified, then all stereoisomers are contemplated and included as the compounds of the disclosure. Where stereochemistry is specified by a solid wedge or dashed line representing a particular configuration, then that stereoisomer is so specified and defined.
  • (1-3C)alkyl and “(1-6C)alkyl” as used herein refer to saturated linear or branched-chain monovalent hydrocarbon radicals of one to three carbon atoms and one to six carbon atoms, respectively. Examples include, but are not limited to, methyl, ethyl, 1-propyl, isopropyl, 1-butyl, isobutyl, sec-butyl, tert-butyl, 2-methyl-2-propyl, pentyl, and hexyl.
  • fluoro(1-6C)alkyl refers to saturated linear or branched-chain monovalent hydrocarbon radicals of one to six carbon atoms as defined herein, wherein one of the hydrogens is replaced by a fluorine atom.
  • difluoro(1-6C)alkyl refers to saturated linear or branched-chain monovalent hydrocarbon radicals of one to six carbon atoms as defined herein, wherein two of the hydrogens are replaced by fluorine atoms.
  • trifluoro(1-6C)alkyl refers to saturated linear or branched-chain monovalent hydrocarbon radicals of one to six carbon atoms as defined herein, wherein three of the hydrogens are replaced by fluorine atoms.
  • hydroxy(1-6Calkyl) refers to saturated linear or branched-chain monovalent hydrocarbon radicals of one to six carbon atoms, wherein one of the hydrogens is replaced by a hydroxy (OH) group.
  • dihydroxy(2-6C alkyl) refers to saturated linear or branched-chain monovalent hydrocarbon radicals of two to six carbon atoms as defined herein, wherein two of the hydrogens are replaced by hydroxy (OH) groups, provided the hydroxy groups are not on the same carbon atom.
  • (1-6C alkyl)sulfonyl refers to a (1-6C alkyl)SO 2 — group, wherein the radical is on the sulfur atom and the (1-6C alkyl) portion is as defined above. Examples include methylsulfonyl (CH 3 SO 2 —) and ethylsulfonyl (CH 3 CH 2 SO 2 —).
  • (3-6C cycloalkyl)sulfonyl refers to a (3-6C cycloalkyl)SO 2 — group, wherein the radical is on the sulfur atom.
  • An example is cyclopropylsulfonyl.
  • (1-3C)alkoxy and “(1-6C)alkoxy”, as used herein refer to saturated linear or branched-chain monovalent alkoxy radicals of one to three carbon atoms or one to six carbon atoms, respectively, wherein the radical is on the oxygen atom. Examples include methoxy, ethoxy, propoxy, isopropoxy, and butoxy.
  • halogen includes fluoro, chloro, bromo and iodo.
  • Non-limiting examples of the compounds of Formula I include those in Table 1.
  • the compounds of Formula I include salts thereof.
  • the salts are pharmaceutically acceptable salts.
  • the compounds of Formula I include other salts of such compounds which are not necessarily pharmaceutically acceptable salts, and which may be useful as intermediates for preparing and/or purifying compounds of Formula I and/or for separating enantiomers of compounds of Formula I.
  • pharmaceutically acceptable indicates that the substance or composition is compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
  • the compounds provided herein may also contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. That is, an atom, in particular when mentioned in relation to a compound according to Formula I, comprises all isotopes and isotopic mixtures of that atom, either naturally occurring or synthetically produced, either with natural abundance or in an isotopically enriched form.
  • the compounds provided herein therefore also comprise compounds with one or more isotopes of one or more atom, and mixtures thereof, including radioactive compounds, wherein one or more non-radioactive atoms has been replaced by one of its radioactive enriched isotopes.
  • Radiolabeled compounds are useful as therapeutic agents, e.g., cancer therapeutic agents, research reagents, e.g., assay reagents, and diagnostic agents, e.g., in vivo imaging agents. All isotopic variations of the compounds of the present disclosure, whether radioactive or not, are intended to be encompassed within the scope of the present disclosure.
  • the compounds of Formula I or a salt thereof as defined herein can be prepared as described in U.S. Pat. No. 8,933,084, which is incorporated by reference in its entirety herein.
  • a process for preparing a compound of Formula I or a salt thereof as defined herein can include:
  • ring B is ring B-1 having the structure:
  • D is carbon
  • R 2 and R 2a are independently H, F, (1-3 C)alkyl or OH (provided that R 2 and R 2a are not both OH)
  • R 3 is H, (1-3 C)alkyl or hydroxy(1-3 C)alkyl
  • ring A, W, m, Z, Y, R 3a , R 5 and R 6 are as defined for Formula I.
  • the cyclization may be performed using conventional amide bond formation conditions, for example by treating the carboxylic acid with an activating agent, followed by addition of the amine in the presence of a base.
  • Suitable activating agents include EDCI, oxalyl chloride, thionyl chloride, HATU, and HOBt.
  • Suitable bases include amine bases, for example triethylamine, diisopropylethylamine, pyridine, or excess ammonia.
  • Suitable solvents include DCM, DCE, THF and DMF.
  • the leaving atoms L 1 and L 2 may be, for example a halogen atom such as Br, Cl or I.
  • L 1 and L 2 can be a leaving group, for example an arylsulfonyloxy group or an alkylsulfonyloxy group, such as a mesylate or a tosylate group.
  • Suitable bases include alkali metal carbonates, such as sodium carbonate, potassium carbonate or cesium carbonate.
  • Convenient solvents include aprotic solvents such as ethers (for example tetrahydrofuran or p-dioxane), DMF, or acetone.
  • the reaction can be conveniently performed at elevated temperatures, for example 50-150° C., for example at 85° C.
  • suitable coupling reagents include HATU, HBTU, TBTU, DCC, DIEC, and any other amide coupling reagents well known to persons skilled in the art.
  • Suitable bases include tertiary amine bases such as DIEA and triethylamine.
  • Convenient solvents include DMF, THF, DCM and DCE.
  • suitable reducing agents include Me 4 N(OAc) 3 BH, Na(OAc) 3 BH and NaCNBH 3 .
  • Suitable solvents include neutral solvents such as acetonitrile, THF and DCE. The reaction can be conveniently performed at ambient temperature.
  • the triphenylphosphine reagent is used in the form of a polystyrene-bound PPh 3 resin (sold as PS-PPh 3 by Biotage Systems).
  • the reaction is conveniently performed at ambient temperature.
  • Suitable solvents include neutral solvents, for example DCM.
  • the leaving atom L 3 may be a halogen, for example Cl or Br.
  • Suitable bases include tertiary amine bases such as diisopropylethylamine and triethylamine. The reaction is conveniently performed at ambient temperature.
  • suitable bases include tertiary amine bases such as DIEA and triethylamine.
  • the reaction is conveniently performed at ambient temperature.
  • the fluorination reagent may be, for example, bis(2-methoxyethyl)amino-sulfur trifluoride (Deoxo-FluorTM) or diethylaminosulfur trifluoride (DAST).
  • Suitable solvents include dichloromethane, chloroform, dichloroethane, and toluene. The reaction is conveniently performed at ambient temperature.
  • base may be, for example, an alkali metal carbonate, such as for example sodium carbonate, potassium carbonate or cesium carbonate.
  • Convenient solvents include aprotic solvents such as ethers (for example tetrahydrofuran or p-dioxane) or toluene.
  • the reaction can be conveniently performed at a temperature between ambient temperature and reflux, for example at 85° C.
  • Amine groups in compounds described in any of the above methods may be protected with any convenient amine protecting group, for example as described in Greene & Wuts, eds., “Protecting Groups in Organic Synthesis”, 2 nd ed. New York; John Wiley & Sons, Inc., 1991.
  • amine protecting groups include acyl and alkoxycarbonyl groups, such as t-butoxycarbonyl (BOC), and [2-(trimethylsilyl)ethoxy]methyl (SEM).
  • carboxyl groups may be protected with any convenient carboxyl protecting group, for example as described in Greene & Wuts, eds., “Protecting Groups in Organic Synthesis”, 2 nd ed.
  • carboxyl protecting groups include (1-6C)alkyl groups, such as methyl, ethyl and t-butyl.
  • Alcohol groups may be protected with any convenient alcohol protecting group, for example as described in Greene & Wuts, eds., “Protecting Groups in Organic Synthesis”, 2 nd ed. New York; John Wiley & Sons, Inc., 1991.
  • Examples of alcohol protecting groups include benzyl, trityl, silyl ethers, and the like.
  • test compounds to act as ROS1 inhibitors may be demonstrated by the assay described in Example A. IC 50 values are shown in Table 17.
  • inhibition of L2026M is similar to, or better than, that observed for wild-type ROS1.
  • inhibition of L2026M is within about 2-fold (e.g., about 5-fold, about 7-fold, about 10-fold) of inhibition of wild-type ROS1 (i.e. the compounds are similarly potent against wild-type ROS1 and L2026M).
  • inhibition of L2026M is about the same as inhibition of wild-type ROS1.
  • inhibition of L2026M is about 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, or greater than inhibition of wild-type ROS1.
  • selectivity for a wildtype or L2026M ROS1 kinase over another kinase is measured in an enzyme assay (e.g., an enzyme assay as provided herein).
  • an enzyme assay e.g., an enzyme assay as provided herein.
  • the compounds provided herein exhibit selective cytotoxicity to ROS1-mutant cells.
  • inhibition of D2033N is similar to, or better than, that observed for wild-type ROS1. In some embodiments, inhibition of D2033N is within about 2-fold (e.g., about 5-fold, about 7-fold, about 10-fold) of inhibition of wild-type ROS1 (i.e. the compounds are similarly potent against wild-type ROS1 and D2033N). In some embodiments, inhibition of D2033N is about the same as inhibition of wild-type ROS1. In some embodiments, inhibition of D2033N is about 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, or greater than inhibition of wild-type ROS1.
  • selectivity for a wildtype or D2033N ROS1 kinase over another kinase is measured in an enzyme assay (e.g., an enzyme assay as provided herein).
  • an enzyme assay e.g., an enzyme assay as provided herein.
  • the compounds provided herein exhibit selective cytotoxicity to ROS1-mutant cells.
  • ROS1-associated diseases and disorders e.g., proliferative disorders such as cancers, including hematological cancers and solid tumors.
  • treat or “treatment” refer to therapeutic or palliative measures.
  • Beneficial or desired clinical results include, but are not limited to, alleviation, in whole or in part, of symptoms associated with a disease or disorder or condition, diminishment of the extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state (e.g., one or more symptoms of the disease), and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • the terms “subject,” “individual,” or “patient,” are used interchangeably, refers to any animal, including mammals such as mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, primates, and humans.
  • the patient is a human.
  • the subject has experienced and/or exhibited at least one symptom of the disease or disorder to be treated and/or prevented.
  • the subject has been identified or diagnosed as having a cancer with a dysregulation of a ROS1 gene, a ROS1 protein, or expression or activity, or level of any of the same (a ROS1-associated cancer) (e.g., as determined using a regulatory agency-approved, e.g., FDA-approved, assay or kit).
  • the assay is a liquid biopsy.
  • the subject has a tumor that is positive for a dysregulation of a ROS1 gene, a ROS1 protein, or expression or activity, or level of any of the same (e.g., as determined using a regulatory agency-approved assay or kit).
  • the subject can be a subject with a tumor(s) that is positive for a dysregulation of a ROS1 gene, a ROS1 protein, or expression or activity, or level of any of the same (e.g., identified as positive using a regulatory agency-approved, e.g., FDA-approved, assay or kit).
  • the assay is a liquid biopsy.
  • the subject can be a subject whose tumors have a dysregulation of a ROS1 gene, a ROS1 protein, or expression or activity, or a level of the same (e.g., where the tumor is identified as such using a regulatory agency-approved, e.g., FDA-approved, kit or assay).
  • the subject is suspected of having a ROS1-associated cancer.
  • the subject has a clinical record indicating that the subject has a tumor that has a dysregulation of a ROS1 gene, a ROS1 protein, or expression or activity, or level of any of the same (and optionally the clinical record indicates that the subject should be treated with any of the compositions provided herein).
  • the patient is a pediatric patient.
  • the term “pediatric patient” as used herein refers to a patient under the age of 21 years at the time of diagnosis or treatment.
  • the term “pediatric” can be further be divided into various subpopulations including: neonates (from birth through the first month of life); infants (1 month up to two years of age); children (two years of age up to 12 years of age); and adolescents (12 years of age through 21 years of age (up to, but not including, the twenty-second birthday)).
  • Berhman R E, Kliegman R, Arvin A M Nelson W E. Nelson Textbook of Pediatrics, 15th Ed. Philadelphia: W.B. Saunders Company, 1996; Rudolph A M, et al. Rudolph's Pediatrics, 21st Ed.
  • a pediatric patient is from birth through the first 28 days of life, from 29 days of age to less than two years of age, from two years of age to less than 12 years of age, or 12 years of age through 21 years of age (up to, but not including, the twenty-second birthday).
  • a pediatric patient is from birth through the first 28 days of life, from 29 days of age to less than 1 year of age, from one month of age to less than four months of age, from three months of age to less than seven months of age, from six months of age to less than 1 year of age, from 1 year of age to less than 2 years of age, from 2 years of age to less than 3 years of age, from 2 years of age to less than seven years of age, from 3 years of age to less than 5 years of age, from 5 years of age to less than 10 years of age, from 6 years of age to less than 13 years of age, from 10 years of age to less than 15 years of age, or from 15 years of age to less than 22 years of age.
  • compounds of Formula I are useful for preventing diseases and disorders as defined herein (for example, cancer).
  • preventing means the prevention of the onset, recurrence or spread, in whole or in part, of the disease or condition as described herein, or a symptom thereof.
  • ROS1-associated disease or disorder refers to diseases or disorders associated with or having a dysregulation of a ROS1 gene, a ROS1 kinase (also called herein ROS1 kinase protein), or the expression or activity or level of any (e.g., one or more) of the same (e.g., any of the types of dysregulation of a ROS1 gene, a ROS1 kinase, a ROS1 kinase domain, or the expression or activity or level of any of the same described herein).
  • a non-limiting example of a ROS1-associated disease or disorder includes cancer.
  • ROS1-associated cancer refers to cancers associated with or having a dysregulation of a ROS1 gene, a ROS1 kinase (also called herein ROS1 kinase protein), or expression or activity, or level of any of the same.
  • ROS1 kinase protein also called herein ROS1 kinase protein
  • Non-limiting examples of a ROS1-associated cancer are described herein.
  • the phrase “dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same” refers to a genetic mutation (e.g., a ROS1 gene translocation that results in the expression of a fusion protein, a deletion in a ROS1 gene that results in the expression of a ROS1 protein that includes a deletion of at least one amino acid as compared to the wild-type ROS1 protein, a mutation in a ROS1 gene that results in the expression of a ROS1 protein with one or more point mutations, or an alternative spliced version of a ROS1 mRNA that results in a ROS1 protein having a deletion of at least one amino acid in the ROS1 protein as compared to the wild-type ROS1 protein) or a ROS1 gene amplification that results in overexpression of a ROS1 protein or an autocrine activity resulting from the overexpression of a ROS1 gene in a cell that results in a
  • a dysregulation of a ROS1 gene, a ROS1 protein, or expression or activity, or level of any of the same can be a mutation in a ROS1 gene that encodes a ROS1 protein that is constitutively active or has increased activity as compared to a protein encoded by a ROS1 gene that does not include the mutation.
  • a dysregulation of a ROS1 gene, a ROS1 protein, or expression or activity, or level of any of the same can be the result of a gene or chromosome translocation which results in the expression of a fusion protein that contains a first portion of ROS1 that includes a functional kinase domain, and a second portion of a partner protein that is not ROS1.
  • dysregulation of a ROS1 gene, a ROS1 protein, or expression or activity or level of any of the same can be a result of a gene translocation of one ROS1 gene with another non-ROS1 gene.
  • Non-limiting examples of fusion proteins are described in Table 2.
  • Non-limiting examples of ROS1 kinase protein point mutations are described in Table 3 and Table 3a. Additional examples of ROS1 kinase protein mutations (e.g., point mutations) are ROS1 inhibitor resistance mutations.
  • Non-limiting examples of ROS1 inhibitor resistance mutations are described in Table 4.
  • wildtype when referring to a ROS1 nucleic acid or protein describes a nucleic acid (e.g., a ROS1 gene or a ROS1 mRNA) or protein (e.g., a ROS1 protein) that is found in a subject that does not have a ROS1-associated disease, e.g., a ROS1-associated cancer (and optionally also does not have an increased risk of developing a ROS1-associated disease and/or is not suspected of having a ROS1-associated disease), or is found in a cell or tissue from a subject that does not have a ROS1-associated disease, e.g., a ROS1-associated cancer (and optionally also does not have an increased risk of developing a ROS1-associated disease and/or is not suspected of having a ROS1-associated disease).
  • a ROS1-associated disease e.g., a ROS1-associated cancer
  • regulatory agency refers to a country's agency for the approval of the medical use of pharmaceutical agents with the country.
  • FDA U.S. Food and Drug Administration
  • a method of treating cancer e.g., a ROS1-associated cancer
  • the method comprising administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof or a pharmaceutical composition thereof.
  • methods for treating a ROS1-associated cancer in a patient in need of such treatment comprising a) detecting a dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same in a sample from the patient; and b) administering a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • the dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same includes one or more fusion proteins.
  • ROS1 gene fusion proteins are described in Table 2.
  • the fusion protein is one of SLC34A2-ROS1, CD74-ROS1, EZR-ROS1, TPM3-ROS1, or SDC4-ROS1.
  • the dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same includes one or more ROS1 kinase protein point mutations, insertions, and/or deletions.
  • ROS1 kinase protein point mutations are described in Table 3 and Table 3a.
  • the ROS1 kinase protein point mutations, insertions, and/or deletions are point mutations selected from the group consisting of A15G, R118N, G1025R, T1735M, R1948H, and R2072N.
  • a compound of Formula I is selected from Example No. 2, 3, 7, 9, 14, 19, 20, 22, 33-A, 33-B, 35, 36, and 45, or a pharmaceutically acceptable salt or solvate thereof.
  • the cancer e.g., ROS1-associated cancer
  • the cancer is a hematological cancer.
  • the cancer e.g., ROS1-associated cancer
  • the cancer e.g., ROS1-associated cancer
  • lung cancer e.g., small cell lung carcinoma or non-small cell lung carcinoma
  • papillary thyroid cancer medullary thyroid cancer, differentiated thyroid cancer, recurrent thyroid cancer, refractory differentiated thyroid cancer, lung adenocarcinoma, bronchioles lung cell carcinoma, multiple endocrine neoplasia type 2A or 2B (MEN2A or MEN2B, respectively), pheochromocytoma, parathyroid hyperplasia, breast cancer, colorectal cancer (e.g., metastatic colorectal cancer), papillary renal cell carcinoma, ganglioneuromatosis of the gastroenteric mucosa, inflammatory myofibroblastic tumor, or cervical cancer.
  • lung cancer e.g., small cell lung carcinoma or non-small cell lung carcinoma
  • papillary thyroid cancer medullary thyroid cancer
  • differentiated thyroid cancer differentiated thyroid cancer
  • recurrent thyroid cancer refractory differentiated thyroid cancer
  • the cancer e.g., ROS1-associated cancer
  • ALL acute lymphoblastic leukemia
  • AML acute myeloid leukemia
  • adrenocortical carcinoma anal cancer
  • appendix cancer astrocytoma, atypical teratoid/rhabdoid tumor
  • basal cell carcinoma bile duct cancer
  • bladder cancer bone cancer
  • brain stem glioma brain tumor, breast cancer, bronchial tumor, Burkitt lymphoma
  • carcinoid tumor unknown primary carcinoma, cardiac tumors, cervical cancer, childhood cancers, chordoma, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myeloproliferative neoplasms, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, bile duct cancer, duct
  • a hematological cancer is selected from the group consisting of leukemias, lymphomas (non-Hodgkin's lymphoma), Hodgkin's disease (also called Hodgkin's lymphoma), and myeloma, for instance, acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), acute promyelocytic leukemia (APL), chronic lymphocytic leukemia (CLL), chronic myeloid leukemia (CML), chronic myelomonocytic leukemia (CMML), chronic neutrophilic leukemia (CNL), acute undifferentiated leukemia (AUL), anaplastic large-cell lymphoma (ALCL), prolymphocytic leukemia (PML), juvenile myelomonocyctic leukemia (JMML), adult T-cell ALL, AML with triline
  • ALL acute lymphocytic leukemia
  • AML acute myeloid leukemia
  • hematological cancers include myeloproliferative disorders (MPD) such as polycythemia vera (PV), essential thrombocytopenia (ET) and idiopathic primary myelofibrosis (IMF/IPF/PMF).
  • MPD myeloproliferative disorders
  • PV polycythemia vera
  • ET essential thrombocytopenia
  • IMF/IPF/PMF idiopathic primary myelofibrosis
  • the hematological cancer e.g., the hematological cancer that is a RET-associated cancer
  • AML or CMML.
  • the cancer is a solid tumor.
  • solid tumors e.g., solid tumors that are ROS1-associated cancers
  • the cancer is selected from the group consisting of lung cancer (including, e.g., non-small-cell lung cancer), colorectal cancer, gastric cancer, adenocarcinoma (including, e.g., small bowel adenocarcinoma), cholangiocarcinoma, glioblastoma, ovarian cancer, angiocarcinoma, congenital gliobastoma multiforme, papillary thyroid carcinoma, inflammatory myofibroblastic tumour, a spitzoid neoplasm, anaplastic large cell lymphoma, diffuse large B cell lymphoma, and B-cell acute lymphoblastic leukemia.
  • lung cancer including, e.g., non-small-cell lung cancer
  • colorectal cancer gastric cancer
  • adenocarcinoma including, e.g., small bowel adenocarcinoma
  • cholangiocarcinoma including, e.g., small bowel a
  • the patient is a human.
  • a method for treating a patient diagnosed with or identified as having a ROS1-associated cancer comprising administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition thereof as defined herein.
  • Dysregulation of a ROS1 kinase, a ROS1 gene, or the expression or activity or level of any (e.g., one or more) of the same can contribute to tumorigenesis.
  • a dysregulation of a ROS1 kinase, a ROS1 gene, or expression or activity or level of any of the same can be a translocation, overexpression, activation, amplification, or mutation of a ROS1 kinase, a ROS1 gene, or a ROS1 kinase domain.
  • a translocation can include a translocation involving the ROS1 kinase domain
  • a mutation can include a mutation involving the ROS1 ligand-binding site
  • an amplification can be of a ROS1 gene.
  • the dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same includes overexpression of wild-type ROS1 kinase (e.g., leading to autocrine activation).
  • the dysregulation of a ROS1 gene, a ROS1 kinase protein, or expression or activity or level of any of the same includes overexpression, activation, amplification, or mutation in a chromosomal segment comprising the ROS1 gene or a portion thereof, including, for example, the kinase domain portion, or a portion capable of exhibiting kinase activity.
  • the dysregulation of a ROS1 gene, a ROS1 kinase protein, or expression or activity or level of any of the same includes one or more chromosome translocations or inversions resulting in a ROS1 gene fusion.
  • the dysregulation of a ROS1 gene, a ROS1 kinase protein, or expression or activity or level of any of the same is a result of genetic translocations in which the expressed protein is a fusion protein containing residues from a non-ROS1 partner protein, and includes a minimum of a functional ROS1 kinase domain.
  • ROS1 fusion proteins are shown in Table 2.
  • Non-small-cell lung cancer 1 SLC34A2 including Non-small-cell lung cancer 1 , Colorectal SLC34A2-ROS(S) 28 , cancer 14 , Gastric cancer 15 , Lung SLC34A2-ROS(L) 28 , and adenocarcinoma 24 SLC34A2-ROS(VS) 28 , SLC34A2-ROS (with a breakpoint at chr6: 117653720, chr4: 25678781) 24 TPM3 Non-small-cell lung cancer 1 SDC4 Non-small-cell lung cancer 1 , Adenocarcinoma 10 EZR Non-small-cell lung cancer 1 LRIG3 Non-small-cell lung cancer 1 KDELR2 Non-small-cell lung cancer 1 CCDC6 Non-small-cell lung cancer 1 FIG (GOPC, PIST) Non-small-cell lung cancer 2 , including FIG-ROS1(L) 29 , Cholangiocarcinoma 5 ,
  • the dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same includes one or more deletions, insertions, or point mutation(s) in a ROS1 kinase. In some embodiments, the dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same, includes a deletion of one or more residues from the ROS1 kinase, resulting in constitutive activity of the ROS1 kinase domain.
  • the dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same includes at least one point mutation in a ROS1 gene that results in the production of a ROS1 kinase that has one or more amino acid substitutions, insertions, or deletions as compared to the wild-type ROS1 kinase (see, for example, the point mutations listed in Table 3).
  • ROS1 Point Mutations ROS1 Mutation ROS1-Associated Cancer
  • Amino acid position 15 Diffuse large B cell lymphoma 1 (e.g., A15G) Amino acid position 118 B-cell acute lymphoblastic (e.g., R118N) leukemia 2 Amino acid position 122 Gastrointestinal stromal tumors (e.g., A122T) (GISTs) 3 Amino acid position 245 Uterine Corpus Endometrioid (e.g., R245I) Carcinoma 4 Amino acid position 1025 B-cell acute lymphoblastic (e.g., G1025R) leukemia 2 Amino acid position 1186 Non-Small-Cell Lung Cancer 5 (e.g., S1186F) Amino acid position 1539 Skin Cutaneous Melanoma 7 (e.g., P1539S) Amino acid position 1735 B-cell acute lymphoblastic (e.g., T17) Am
  • Amino acid position 1186 (e.g., S1186F 11 ) Amino acid position 1935 (e.g., E1935G6 10 ) Amino acid position 1945 (e.g., L1945Q 7 ) Amino acid position 1946 (e.g., T1946S 7 ) Amino acid position 1947 (e.g., L1947R 6, 10 , L1947M 7 ) Amino acid position 1948 (e.g., R1948S 7 ) Amino acid position 1951 (e.g., L1951R 5 , L1951V 7 ) Amino acid position 1958 (e.g., E1958V 7 ) Amino acid position 1959 (e.g., V1959E 7 ) Amino acid position 1961 (e.g., E1961K 7 ) Amino acid position 1962 (e.g., G1962E 7 ) Amino acid position 1971 (e.g., G1971E 6, 10 ) Amino acid position 1945 (e
  • the dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same includes a splice variation in a ROS1 mRNA which results in an expressed protein that is an alternatively spliced variant of ROS1 having at least one residue deleted (as compared to the wild-type ROS1 kinase) resulting in a constitutive activity of a ROS1 kinase domain.
  • the dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same includes a splice variation in a ROS1 mRNA which results in an expressed protein that is an alternatively spliced variant of ROS1 having at least one residue added (as compared to the wild-type ROS1 kinase) resulting in a constitutive activity of a ROS1 kinase domain.
  • a “ROS1 kinase inhibitor” as defined herein includes any compound exhibiting ROS1 inhibition activity.
  • a ROS1 kinase inhibitor is selective for a wild type and/or mutant ROS1 kinase.
  • ROS1 kinase inhibitors can exhibit inhibition activity (IC 50 ) against a ROS1 kinase of less than about 1000 nM, less than about 500 nM, less than about 200 nM, less than about 100 nM, less than about 50 nM, less than about 25 nM, less than about 10 nM, or less than about 1 nM as measured in an assay as described herein.
  • a ROS1 kinase inhibitors can exhibit inhibition activity (IC 50 ) against a ROS1 kinase of less than about 25 nM, less than about 10 nM, less than about 5 nM, or less than about 1 nM as measured in an assay as provided herein.
  • the ROS1 kinase inhibitor is a compound of Formula I.
  • a “first ROS1 kinase inhibitor” or “first ROS1 inhibitor” is a ROS1 kinase inhibitor as defined herein, but which does not include a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as defined herein.
  • a “second ROS1 kinase inhibitor” or a “second ROS1 inhibitor” is a ROS1 kinase inhibitor as defined herein. In some embodiments, a second ROS1 inhibitor does not include a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as defined herein.
  • ROS1 inhibitors When more than one ROS1 inhibitor is present in a method provided herein (e.g., both a first and a second ROS1 inhibitor are present in a method provided herein), the two ROS1 inhibitors are different (e.g., the first and second ROS1 kinase inhibitor are different). As provided herein, different ROS1 inhibitors are structurally distinct from one another.
  • the dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same includes at least one point mutation in a ROS1 gene that results in the production of a ROS1 kinase that has one or more amino acid substitutions or insertions or deletions as compared to the wild-type ROS1 kinase.
  • the resulting ROS1 kinase is more resistant to inhibition of its phosphotransferase activity by one or more first ROS1 kinase inhibitor(s), as compared to a wildtype ROS1 kinase or a ROS1 kinase not including the same mutation.
  • Such mutations optionally, do not decrease the sensitivity of the cancer cell or tumor having the ROS1 kinase to treatment with a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof (e.g., as compared to a cancer cell or a tumor that does not include the particular ROS1 inhibitor resistance mutation).
  • a ROS1 inhibitor resistance mutation can result in a ROS1 kinase that has one or more of an increased V max , a decreased K m for ATP, and an increased K D for a first ROS1 kinase inhibitor, when in the presence of a first ROS1 kinase inhibitor, as compared to a wildtype ROS1 kinase or a ROS1 kinase not having the same mutation in the presence of the same first ROS1 kinase inhibitor.
  • the dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same includes at least one point mutation in a ROS1 gene that results in the production of a ROS1 kinase that has one or more amino acid substitutions as compared to the wild-type ROS1 kinase, and which has increased resistance to a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof, as compared to a wildtype ROS1 kinase or a ROS1 kinase not including the same mutation.
  • a ROS1 inhibitor resistance mutation can result in a ROS1 kinase that has one or more of an increased V max , a decreased K m , and a decreased K D in the presence of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof, as compared to a wildtype ROS1 kinase or a ROS1 kinase not having the same mutation in the presence of the same compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • ROS1 inhibitor resistance mutations can, e.g., include point mutations, insertions, or deletions in and near the ATP binding site in the tertiary structure of ROS1 kinase, including but not limited to the gatekeeper residue, P-loop residues, residues in or near the DFG motif, and ATP cleft solvent front amino acid residues. Additional examples of these types of mutations include changes in residues that may affect enzyme activity and/or drug binding including but are not limited to residues in the activation loop, residues near or interacting with the activation loop, residues contributing to active or inactive enzyme conformations, changes including mutations, deletions, and insertions in the loop proceeding the C-helix and in the C-helix.
  • ROS1 inhibitor resistance mutations include but are not limited to those listed in Table 4 based on the human wildtype ROS1 protein sequence (e.g., SEQ ID NO: 1). Changes to these residues may include single or multiple amino acid changes, insertions within or flanking the sequences, and deletions within or flanking the sequences.
  • compounds of Formula I and pharmaceutically acceptable salts and solvates are useful in treating patients that develop cancers with ROS1 inhibitor resistance mutations (e.g., that result in an increased resistance to a first ROS1 inhibitor, e.g., a substitution at amino acid position 2032 (e.g., G2032R), amino acid position 2026 (e.g., L2026M), amino acid position 2033 (e.g., D2033N), and/or one or more ROS1 inhibitor resistance mutations listed in Table 4) by either dosing in combination or as a follow-up therapy to existing drug treatments (e.g., ALK kinase inhibitors, TRK kinase inhibitors, other ROS1 kinase inhibitors, e.g., first and/or second ROS1 kinase inhibitors).
  • a first ROS1 inhibitor e.g., a substitution at amino acid position 2032 (e.g., G2032R), amino acid position 2026 (e.g., L
  • Exemplary ALK kinase inhibitors are described herein.
  • Exemplary TRK kinase inhibitors are described herein.
  • Exemplary first and second ROS1 kinase inhibitors are described herein.
  • a first or second ROS1 kinase inhibitor can be selected from the group consisting of alectinib, brigatinib, cabozantinib, ceritinib, crizotinib, entrectinib, foretinib, lorlatinib, and mesestinib.
  • compounds of Formula I or pharmaceutically acceptable salts and solvates thereof are useful for treating a cancer that has been identified as having one or more ROS1 inhibitor resistance mutations (that result in an increased resistance to a first or second ROS1 inhibitor, e.g., a substitution at amino acid position 2032 (e.g., G2032R), amino acid position 2026 (e.g., L2026M), amino acid position 2033 (e.g., D2033N)).
  • ROS1 inhibitor resistance mutations are listed in Table 4.
  • Amino acid position 1186 (e.g., S1186F 11 ) Amino acid position 1935 (e.g., E1935G6 10 ) Amino acid position 1945 (e.g., L1945Q 7 ) Amino acid position 1946 (e.g., T1946S 7 ) Amino acid position 1947 (e.g., L1947R 6, 10 , L1947M 7 ) Amino acid position 1948 (e.g., R1948S 7 ) Amino acid position 1951 (e.g., L1951R 5 , L1951V 7 ) Amino acid position 1958 (e.g., E1958V 7 ) Amino acid position 1959 (e.g., V1959E 7 ) Amino acid position 1961 (e.g., E1961K 7 ) Amino acid position 1962 (e.g., G1962E 7 ) Amino acid position 1971 (e.g., G1971E 6, 10 ) Amino acid position 1945 (e
  • ROS1 proto-oncogene is expressed in a variety of tumor types, and belongs to the sevenless subfamily of tyrosine kinase insulin receptor genes.
  • the protein encoded by this gene is a type I integral membrane protein with tyrosine kinase activity.
  • ROS1 shares structural similarity with the anaplastic lymphoma kinase (ALK) protein.
  • ALK anaplastic lymphoma kinase
  • ROS1 fusions have been detected in multiple other tumor histologies, including ovarian carcinoma, sarcoma, cholangiocarcinomas and others.
  • ALK is a receptor tyrosine kinase that belongs to the insulin growth factor receptor superfamily. ALK is believed to play a role in the development of the nervous system.
  • a variety of ALK gene fusions have been described, such as EML4, KIF5B, KLC1, and TRK-fused gene (TFG). Such fusion products result in kinase activation and oncogenesis.
  • NSCLC Non-small-cell lung cancer
  • ALK anaplastic lymphoma kinase gene
  • crizotinib which is an inhibitor of ALK and ROS1.
  • the additional therapeutic agent(s) includes any one of the above listed therapies or therapeutic agents which are standards of care in cancers wherein the cancer has a dysregulation of a ROS1 gene, a ROS1 protein, or expression or activity, or level of any of the same.
  • the additional therapeutic agent(s) includes any one of the above listed therapies or therapeutic agents which are standards of care in cancers wherein the cancer has a dysregulation of an ALK gene, an ALK protein, or expression or activity, or level of any of the same (e.g., an ALK-associated cancer).
  • the additional therapeutic agent(s) includes any one of the above listed therapies or therapeutic agents which are standards of care in cancers wherein the cancer has a dysregulation of a TRK gene, a TRK protein, or expression or activity, or level of any of the same (e.g., a TRK-associated cancer).
  • ALK-associated cancer refers to cancers associated with or having a dysregulation of an ALK gene, an ALK protein, or expression or activity, or level of any of the same. Exemplary ALK-associated cancers are provided herein.
  • the phrase “dysregulation of an ALK gene, an ALK kinase, or the expression or activity or level of any of the same” refers to a genetic mutation (e.g., an ALK gene translocation that results in the expression of a fusion protein, a deletion in an ALK gene that results in the expression of an ALK protein that includes a deletion of at least one amino acid as compared to the wild-type ALK protein, a mutation in an ALK gene that results in the expression of an ALK protein with one or more point mutations, or an alternative spliced version of an ALK mRNA that results in an ALK protein having a deletion of at least one amino acid in the ALK protein as compared to the wild-type ALK protein) or an ALK gene amplification that results in overexpression of an ALK protein or an autocrine activity resulting from the overexpression of an ALK gene in a cell that results in a pathogenic increase in the activity of a kinase domain of an ALK protein (e.g.,
  • a dysregulation of an ALK gene, an ALK protein, or expression or activity, or level of any of the same can be a mutation in an ALK gene that encodes an ALK protein that is constitutively active or has increased activity as compared to a protein encoded by an ALK gene that does not include the mutation.
  • a dysregulation of an ALK gene, an ALK protein, or expression or activity, or level of any of the same can be the result of a gene or chromosome translocation which results in the expression of a fusion protein that contains a first portion of ALK that includes a functional kinase domain, and a second portion of a partner protein that is not ALK.
  • dysregulation of an ALK gene, an ALK protein, or expression or activity or level of any of the same can be a result of a gene translocation of one ALK gene with another non-ALK gene.
  • Non-limiting examples of fusion proteins are described in Table 5. Additional examples of ALK kinase protein mutations (e.g., point mutations) are ALK inhibitor resistance mutations.
  • wildtype when referring to an ALK nucleic acid or protein describes a nucleic acid (e.g., an ALK gene or an ALK mRNA) or protein (e.g., an ALK protein) that is found in a subject that does not have an ALK-associated disease, e.g., an ALK-associated cancer (and optionally also does not have an increased risk of developing an ALK-associated disease and/or is not suspected of having an ALK-associated disease), or is found in a cell or tissue from a subject that does not have an ALK-associated disease, e.g., an ALK-associated cancer (and optionally also does not have an increased risk of developing an ALK-associated disease and/or is not suspected of having an ALK-associated disease).
  • the dysregulation of an ALK gene, an ALK kinase protein, or expression or activity or level of any of the same includes one or more chromosome translocations or inversions resulting in an ALK gene fusion.
  • the dysregulation of an ALK gene, an ALK kinase protein, or expression or activity or level of any of the same is a result of genetic translocations in which the expressed protein is a fusion protein containing residues from a non-ALK partner protein, and includes a minimum of a functional ALK kinase domain.
  • Non-limiting examples of ALK fusion proteins are shown in Table 5.
  • Anaplastic large cell lymphoma 1 Diffuse large B-cell lymphoma 1 , Neuroblastoma 8 , Lung adenocarcinoma 9
  • ALO17/RNF213 Anaplastic large cell lymphoma 1 TFG (e.g., TFGs, Anaplastic large cell lymphoma 1 , TFG L , TFG XL ) 38
  • Non-small cell lung cancer 1 Anaplastic thyroid carcinoma 1 MSN (e.g., MSNa Anaplastic large cell lymphoma 1 and MSNb) 38 TPM3 Anaplastic large cell lymphoma 1 , Inflammatory myofibroblastic tumour 1 , Renal medulla carcinoma/renal cell carcinoma 1 , Spitzoid neoplasm 5 TPM4 (e.g., type 1 Anaplastic large cell lymphoma 1 , and type 2) 38 Inflammatory myofibroblastic tumour 1 , Oesoph
  • the dysregulation of an ALK gene, an ALK kinase, or expression or activity or level of any of the same includes at least one point mutation in an ALK gene that results in the production of an ALK kinase that has one or more amino acid substitutions, insertions, or deletions as compared to the wild-type ALK kinase.
  • an ALK-associated cancer has been identified as having one or more ALK inhibitor resistance mutations (that result in an increased resistance to an ALK inhibitor.
  • Tropomyosin-related kinase is a receptor tyrosine kinase family of neurotrophin receptors that are found in multiple tissues types. Three members of the TRK proto-oncogene family have been described: TrkA, TrkB, and TrkC, encoded by the NTRK1, NTRK2, and NTRK3 genes, respectively.
  • TrkA, TrkB, and TrkC encoded by the NTRK1, NTRK2, and NTRK3 genes, respectively.
  • the TRK receptor family is involved in neuronal development, including the growth and function of neuronal synapses, memory development, and maintenance, and the protection of neurons after ischemia or other types of injury (Nakagawara, Cancer Lett. 169:107-114, 2001).
  • TRK was originally identified from a colorectal cancer cell line as an oncogene fusion containing 5′ sequences from tropomyosin-3 (TPM3) gene and the kinase domain encoded by the 3′ region of the neurotrophic tyrosine kinase, receptor, type 1 gene (NTRK1) (Pulciani et al., Nature 300:539-542, 1982; Martin-Zanca et al., Nature 319:743-748, 1986).
  • TPM3 tropomyosin-3
  • NTRK1 neurotrophic tyrosine kinase, receptor, type 1 gene
  • TRK gene fusions follow the well-established paradigm of other oncogenic fusions, such as those involving ALK and ROS1, which have been shown to drive the growth of tumors and can be successfully inhibited in the clinic by targeted drugs (Shaw et al., New Engl. J. Med. 371:1963-1971, 2014; Shaw et al., New Engl. J. Med. 370:1189-1197, 2014).
  • Oncogenic TRK fusions induce cancer cell proliferation and engage critical cancer-related downstream signaling pathways such as mitogen activated protein kinase (MAPK) and AKT (Vaishnavi et al., Cancer Discov. 5:25-34, 2015).
  • MAPK mitogen activated protein kinase
  • AKT AKT
  • NTRK1 and its related TRK family members NTRK2 and NTRK3 have been described (Vaishnavi et al., Cancer Disc. 5:25-34, 2015; Vaishnavi et al., Nature Med. 19:1469-1472, 2013). Although there are numerous different 5′ gene fusion partners identified, all share an in-frame, intact TRK kinase domain.
  • Trk inhibitors have been developed to treat cancer (see, e.g., U.S. Patent Application Publication No. 62/080,374, International Application Publication Nos.
  • TRK-associated cancer refers to cancers associated with or having a dysregulation of a TRK gene, a TRK protein, or expression or activity, or level of any of the same. Exemplary TRK-associated cancers are provided herein.
  • the phrase “dysregulation of a TRK gene, a TRK kinase, or the expression or activity or level of any of the same” refers to a genetic mutation (e.g., a TRK gene translocation that results in the expression of a fusion protein, a deletion in a TRK gene that results in the expression of a TRK protein that includes a deletion of at least one amino acid as compared to the wild-type TRK protein, a mutation in a TRK gene that results in the expression of a TRK protein with one or more point mutations, or an alternative spliced version of a TRK mRNA that results in a TRK protein having a deletion of at least one amino acid in the TRK protein as compared to the wild-type TRK protein) or a TRK gene amplification that results in overexpression of a TRK protein or an autocrine activity resulting from the overexpression of a TRK gene in a cell that results in a pathogenic increase in the activity of a kinase domain
  • a dysregulation of a TRK gene, a TRK protein, or expression or activity, or level of any of the same can be a mutation in a TRK gene that encodes a TRK protein that is constitutively active or has increased activity as compared to a protein encoded by a TRK gene that does not include the mutation.
  • a dysregulation of a TRK gene, a TRK protein, or expression or activity, or level of any of the same can be the result of a gene or chromosome translocation which results in the expression of a fusion protein that contains a first portion of TRK that includes a functional kinase domain, and a second portion of a partner protein that is not TRK.
  • dysregulation of a TRK gene, a TRK protein, or expression or activity or level of any of the same can be a result of a gene translocation of one TRK gene with another non-TRK gene.
  • Non-limiting examples of fusion proteins are described in Tables 6-8. Additional examples of TRK kinase protein mutations (e.g., point mutations) are TRK inhibitor resistance mutations.
  • wildtype when referring to a TRK nucleic acid or protein describes a nucleic acid (e.g., a TRK gene or a TRK mRNA) or protein (e.g., a TRK protein) that is found in a subject that does not have a TRK-associated disease, e.g., a TRK-associated cancer (and optionally also does not have an increased risk of developing a TRK-associated disease and/or is not suspected of having a TRK-associated disease), or is found in a cell or tissue from a subject that does not have a TRK-associated disease, e.g., a TRK-associated cancer (and optionally also does not have an increased risk of developing a TRK-associated disease and/or is not suspected of having a TRK-associated disease).
  • the dysregulation of a TRK gene, a TRK kinase protein, or expression or activity or level of any of the same includes one or more chromosome translocations or inversions resulting in a TRK gene fusion.
  • the dysregulation of a TRK gene, a TRK kinase protein, or expression or activity or level of any of the same is a result of genetic translocations in which the expressed protein is a fusion protein containing residues from a non-TRK partner protein, and includes a minimum of a functional TRK kinase domain. See, for example, Tables 6-8.
  • TrkA Fusion Proteins and Cancers Non-limiting Exemplary Trk-and Synonyms of Associated Fusion Protein Non-TrkA Fusion Partner Cancer(s) TP53-TrkA 1,11 Tumor Protein P53 Spitzoid Melanoma, Spitz tumors LMNA-TrkA 1, 12 Lamin A/C Spitzoid Melanoma, Spitz tumors, Undifferentiated Sarcoma, Adult Soft Tissue Sarcoma (e.g., Soft Tissue Sarcoma Metastatic to Lung), Soft Tissue Fibrosarcoma, Spindle Cell Sarcoma G , Congenital Infantile Fibrosarcoma H , Pediatric haemangiopericytoma-like sarcoma I , Colorectal Cancer K CD74-TrkA 2 MHC class II invariant chain Non-Small Cell Lung Cancer (NSCLC) Lung adenocarcimona TFG-TrkA (TRK-T3) 3 TRK-Fused Gene Pa
  • the dysregulation of a TRK gene, a TRK kinase, or expression or activity or level of any of the same includes at least one point mutation in a TRK gene that results in the production of a TRK kinase that has one or more amino acid substitutions, insertions, or deletions as compared to the wild-type TRK kinase.
  • a TRK-associated cancer has been identified as having one or more TRK inhibitor resistance mutations (that result in an increased resistance to a TRK inhibitor.
  • methods for treating a patient diagnosed with (or identified as having) a cancer that include administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • methods for treating a patient identified or diagnosed as having a ROS1-associated cancer that include administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof or a pharmaceutical composition thereof.
  • the patient that has been identified or diagnosed as having a ROS1-associated cancer through the use of a regulatory agency-approved, e.g., FDA-approved test or assay for identifying dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same, in a patient or a biopsy sample from the patient or by performing any of the non-limiting examples of assays described herein.
  • the test or assay is provided as a kit.
  • the assay is a liquid biopsy.
  • the cancer is a ROS1-associated cancer.
  • the ROS1-associated cancer can be a cancer that includes one or more ROS1 inhibitor resistance mutations.
  • Some embodiments of these methods further include administering to the subject one or more additional anticancer agents (e.g., a second ROS1 inhibitor, a second compound of Formula I or a pharmaceutically acceptable salt or solvate thereof, an ALK inhibitor, and/or a TRK inhibitor).
  • one or more additional anticancer agents e.g., a second ROS1 inhibitor, a second compound of Formula I or a pharmaceutically acceptable salt or solvate thereof, an ALK inhibitor, and/or a TRK inhibitor
  • one or more additional anticancer agents are administered after a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • one or more additional anticancer agents are administered with a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • the subject was previously treated with a first ROS1 inhibitor or previously treated with another anticancer treatment, e.g., treatment with another anticancer agent, resection of the tumor or radiation therapy.
  • the subject was previously treated with an ALK inhibitor, a TRK inhibitor, or both.
  • the patient is determined to have a ROS1-associated cancer through the use of a regulatory agency-approved, e.g., FDA-approved test or assay for identifying dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same, in a patient or a biopsy sample from the patient or by performing any of the non-limiting examples of assays described herein.
  • the test or assay is provided as a kit.
  • the assay is a liquid biopsy.
  • the cancer is a ROS1-associated cancer.
  • the ROS1-associated cancer can be a cancer that includes one or more ROS1 inhibitor resistance mutations.
  • Also provided are methods of treating a patient that include performing an assay on a sample obtained from the patient to determine whether the patient has a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same, and administering (e.g., specifically or selectively administering) a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof or a pharmaceutical composition thereof to the patient determined to have a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same.
  • Some embodiments of these methods further include administering to the subject one or more additional anticancer agents (e.g., a second ROS1 inhibitor, a second compound of Formula I or a pharmaceutically acceptable salt or solvate thereof, an ALK inhibitor, and/or a TRK inhibitor).
  • one or more additional anticancer agents e.g., a second ROS1 inhibitor, a second compound of Formula I or a pharmaceutically acceptable salt or solvate thereof, an ALK inhibitor, and/or a TRK inhibitor
  • one or more additional anticancer agents e.g., a second ROS1 inhibitor (e.g., a second compound of Formula I or a pharmaceutically acceptable salt or solvate thereof), an ALK inhibitor, and/or a TRK inhibitor) are administered after a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • one or more additional anticancer agents e.g., a second ROS1 inhibitor, a second compound of Formula I or a pharmaceutically acceptable salt or solvate thereof, an ALK inhibitor, and/or a TRK inhibitor
  • the subject was previously treated with a first ROS1 inhibitor or previously treated with another anticancer treatment, e.g., treatment with another anticancer agent, resection of a tumor or radiation therapy.
  • the subject was previously treated with an ALK inhibitor, a TRK inhibitor, or both.
  • the patient is a patient suspected of having a ROS1-associated cancer, a patient presenting with one or more symptoms of a ROS1-associated cancer, or a patient having an elevated risk of developing a ROS1-associated cancer.
  • the assay utilizes next generation sequencing, pyrosequencing, immunohistochemistry, an enzyme-linked immunosorbent assay, and/or fluorescence in situ hybridization (FISH) (e.g., break apart FISH or dual-fusion FISH).
  • FISH fluorescence in situ hybridization
  • the assay is a regulatory agency-approved assay, e.g., FDA-approved kit.
  • the assay is a liquid biopsy. Additional, non-limiting assays that may be used in these methods are described herein. Additional assays are also known in the art.
  • the dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same includes one or more ROS1 inhibitor resistance mutations.
  • an assay e.g., an in vitro assay
  • a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof for the manufacture of a medicament for treating a ROS1-associated cancer in a patient identified or diagnosed as having a ROS1-associated cancer through a step of performing an assay on a sample obtained from the patient to determine whether the patient has a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same where the presence of dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same, identifies that the patient has a ROS1-associated cancer.
  • Some embodiments of any of the methods or uses described herein further include recording in the patient's clinical record (e.g., a computer readable medium) that the patient is determined to have a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same, through the performance of the assay, should be administered a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof or a pharmaceutical composition thereof.
  • the assay utilizes next generation sequencing, pyrosequencing, immunohistochemistry, an enzyme-linked immunosorbent assay, and/or fluorescence in situ hybridization (FISH) (e.g., break apart FISH or dual-fusion FISH).
  • FISH fluorescence in situ hybridization
  • the assay is a regulatory agency-approved assay, e.g., FDA-approved kit. In some embodiments, the assay is a liquid biopsy. In some embodiments, the dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same includes one or more ROS1 inhibitor resistance mutations.
  • a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof for use in the treatment of a cancer in a patient in need thereof or a patient identified or diagnosed as having a ROS1-associated cancer. Also provided is the use of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof for the manufacture of a medicament for treating a cancer in a patient identified or diagnosed as having a ROS1-associated cancer.
  • the cancer is a ROS1-associated cancer, for example, a ROS1-associated cancer having one or more ROS1 inhibitor resistance mutations.
  • a patient is identified or diagnosed as having a ROS1-associated cancer through the use of a regulatory agency-approved, e.g., FDA-approved, kit for identifying dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same, in a patient or a biopsy sample from the sample.
  • a ROS1-associated cancer includes those described herein and known in the art.
  • the patient has been identified or diagnosed as having a cancer with a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same.
  • the patient has a tumor that is positive for a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same.
  • the patient can be a patient with a tumor(s) that is positive for a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same.
  • the patient can be a patient whose tumors have a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same.
  • the patient is suspected of having a ROS1-associated cancer (e.g., a cancer having one or more ROS1 inhibitor resistance mutations).
  • provided herein are methods for treating a ROS1-associated cancer in a patient in need of such treatment, the method comprising a) detecting a dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same in a sample from the patient; and b) administering a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • the dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same includes one or more fusion proteins.
  • Non-limiting examples of ROS1 gene fusion proteins are described in Table 2.
  • the fusion protein is SLC34A2-ROS1, CD74-ROS1, EZR-ROS1, TPM3-ROS1, or SDC4-ROS1.
  • the dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same includes one or more ROS1 kinase protein point mutations, insertions, and/or deletions. Non-limiting examples of ROS1 kinase protein point mutations are described in Table 3 and Table 3a.
  • the dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same includes one or more ROS1 inhibitor resistance mutations.
  • ROS1 inhibitor resistance mutations are described in Table 4.
  • the ROS1 inhibitor resistance mutation is selected from the group consisting of L2026M, G2032R, and D2033N.
  • the cancer with a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same is determined using a regulatory agency-approved, e.g., FDA-approved, assay or kit.
  • the assay is a liquid biopsy.
  • the tumor that is positive for a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same is a tumor positive for one or more ROS1 inhibitor resistance mutations.
  • the tumor with a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same is determined using a regulatory agency-approved, e.g., FDA-approved, assay or kit.
  • the assay is a liquid biopsy.
  • the patient has a clinical record indicating that the patient has a tumor that has a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same (e.g., a tumor having one or more ROS1 inhibitor resistance mutations).
  • the clinical record indicates that the patient should be treated with one or more of the compounds of Formula I or a pharmaceutically acceptable salts or solvates thereof or compositions provided herein.
  • the cancer with a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same is a cancer having one or more ROS1 inhibitor resistance mutations.
  • the cancer with a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same is determined using a regulatory agency-approved, e.g., FDA-approved, assay or kit.
  • the assay is a liquid biopsy.
  • the tumor that is positive for a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same is a tumor positive for one or more ROS1 inhibitor resistance mutations.
  • the tumor with a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same is determined using a regulatory agency-approved, e.g., FDA-approved, assay or kit.
  • the assay is a liquid biopsy.
  • Also provided are methods of treating a patient that include administering a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof to a patient having a clinical record that indicates that the patient has a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same. Also provided is the use of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof for the manufacture of a medicament for treating a ROS1-associated cancer in a patient having a clinical record that indicates that the patient has a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same.
  • Some embodiments of these methods and uses can further include: a step of performing an on a sample obtained from the patient to determine whether the patient has a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same, and recording the information in a patient's clinical file (e.g., a computer readable medium) that the patient has been identified to have a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same.
  • the assay is an in vitro assay.
  • an assay that utilizes next generation sequencing, pyrosequencing, immunohistochemistry, an enzyme-linked immunosorbent assay, and/or fluorescence in situ hybridization (FISH) (e.g., break apart FISH or dual-fusion FISH).
  • FISH fluorescence in situ hybridization
  • the assay is a regulatory agency-approved, e.g., FDA-approved, kit.
  • the assay is a liquid biopsy.
  • the dysregulation of a ROS1 gene, ROS1 kinase, or expression or activity or level of any of the same includes one or more ROS1 inhibitor resistance mutations.
  • the method includes performing an assay on a sample obtained from the subject to determine whether the subject has a dysregulation of a ROS1 gene, a ROS1 protein, or expression or level of any of the same.
  • the method also includes administering to a subject determined to have a dysregulation of a ROS1 gene, a ROS1 protein, or expression or activity, or level of any of the same a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • the dysregulation in a ROS1 gene, a ROS1 kinase protein, or expression or activity of the same is a gene or chromosome translocation that results in the expression of a ROS1 fusion protein (e.g., any of the ROS1 fusion proteins described herein).
  • the ROS1 fusion can be selected from a SLC34A2 fusion, a CD74 fusion, a EZR fusion, a TPM3 fusion, or a SDC4 fusion.
  • the dysregulation in a ROS1 gene, a ROS1 kinase protein, or expression or activity or level of any of the same is one or more point mutation in the ROS1 gene (e.g., any of the one or more of the ROS1 point mutations described herein).
  • the one or more point mutations in a ROS1 gene can result, e.g., in the translation of a ROS1 protein having one or more of the following amino acid substitutions: A15G, R118N, G1025R, T1735M, R1948H, and R2072N.
  • the dysregulation in a ROS1 gene, a ROS1 kinase protein, or expression or activity or level of any of the same is one or more ROS1 inhibitor resistance mutations (e.g., any combination of the one or more ROS1 inhibitor resistance mutations described herein).
  • the one or more point mutations in a ROS1 gene can result, e.g., in the translation of a ROS1 protein having one or more of the following amino acid substitutions: L2026M, G2032R, and D2033N.
  • Some embodiments of these methods further include administering to the subject another anticancer agent (e.g., a second ROS1 inhibitor, a second compound of Formula I or a pharmaceutically acceptable salt or solvate thereof, an ALK inhibitor, and/or a TRK inhibitor).
  • another anticancer agent e.g., a second ROS1 inhibitor, a second compound of Formula I or a pharmaceutically acceptable salt or solvate thereof, an ALK inhibitor, and/or a TRK inhibitor.
  • the selected treatment can include administration of a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • Some embodiments can further include a step of performing an assay on a sample obtained from the patient to determine whether the patient has a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same, and identifying and diagnosing a patient determined to have a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same, as having a ROS1-associated cancer.
  • the cancer is a ROS1-associated cancer having one or more ROS1 inhibitor resistance mutations.
  • the patient has been identified or diagnosed as having a ROS1-associated cancer through the use of a regulatory agency-approved, e.g., FDA-approved, kit for identifying dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same, in a patient or a biopsy sample from the patient.
  • the assay is a liquid biopsy.
  • the ROS1-associated cancers is a cancer described herein or known in the art.
  • the assay is an in vitro assay.
  • an assay that utilizes next generation sequencing, pyrosequencing, immunohistochemistry, an enzyme-linked immunosorbent assay, and/or fluorescence in situ hybridization (FISH) (e.g., break apart FISH or dual-fusion FISH).
  • FISH fluorescence in situ hybridization
  • the assay is a regulatory agency-approved, e.g., FDA-approved, kit.
  • the assay is a liquid biopsy.
  • Also provided herein are methods of selecting a treatment for a patient wherein the methods include a step of performing an assay on a sample obtained from the patient to determine whether the patient has a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same (e.g., one or more ROS1 inhibitor resistance mutations), and identifying or diagnosing a patient determined to have a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same, as having a ROS1-associated cancer. Some embodiments further include administering the selected treatment to the patient identified or diagnosed as having a ROS1-associated cancer.
  • the selected treatment can include administration of a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof to the patient identified or diagnosed as having a ROS1-associated cancer.
  • the assay is an in vitro assay.
  • an assay that utilizes next generation sequencing, pyrosequencing, immunohistochemistry, an enzyme-linked immunosorbent assay, and/or fluorescence in situ hybridization (FISH) (e.g., break apart FISH or dual-fusion FISH).
  • FISH fluorescence in situ hybridization
  • the assay is a regulatory agency-approved, e.g., FDA-approved, kit.
  • the assay is a liquid biopsy.
  • Also provided are methods of selecting a patient for treatment wherein the methods include selecting, identifying, or diagnosing a patient having a ROS1-associated cancer, and selecting the patient for treatment including administration of a therapeutically-effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • identifying or diagnosing a patient as having a ROS1-associated cancer can include a step of performing an assay on a sample obtained from the patient to determine whether the patient has a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same, and identifying or diagnosing a patient determined to have a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same, as having a ROS1-associated cancer.
  • the method of selecting a treatment can be used as a part of a clinical study that includes administration of various treatments of a ROS1-associated cancer.
  • a ROS1-associated cancer is a cancer having one or more ROS1 inhibitor resistance mutations.
  • the assay is an in vitro assay. For example, an assay that utilizes next generation sequencing, pyrosequencing, immunohistochemistry, an enzyme-linked immunosorbent assay, and/or fluorescence in situ hybridization (FISH) (e.g., break apart FISH or dual-fusion FISH).
  • FISH fluorescence in situ hybridization
  • the assay is a regulatory agency-approved, e.g., FDA-approved, kit.
  • the assay is a liquid biopsy.
  • the dysregulation of the ROS1 gene, the ROS1 kinase, or expression or activity or level of any of the same includes one or more ROS1 inhibitor resistance mutations.
  • an assay used to determine whether the patient has a dysregulation of a ROS1 gene, or a ROS1 kinase, or expression or activity or level of any of the same, using a sample from a patient can include, for example, next generation sequencing, pyrosequencing, immunohistochemistry, an enzyme-linked immunosorbent assay, and/or fluorescence in situ hybridization (FISH) (e.g., break apart FISH or dual-fusion FISH), fluorescence microscopy, Southern blotting, Western blotting, FACS analysis, Northern blotting, and PCR-based amplification (e.g., RT-PCR and quantitative real-time RT-PCR).
  • FISH fluorescence in situ hybridization
  • the assays are typically performed, e.g., with at least one labelled nucleic acid probe or at least one labelled antibody or antigen-binding fragment thereof.
  • Assays can utilize other detection methods known in the art for detecting dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or levels of any of the same (see, e.g., the references cited herein).
  • the dysregulation of the ROS1 gene, the ROS1 kinase, or expression or activity or level of any of the same includes one or more ROS1 inhibitor resistance mutations.
  • the sample is a biological sample or a biopsy sample (e.g., a paraffin-embedded biopsy sample) from the patient.
  • the patient is a patient suspected of having a ROS1-associated cancer, a patient having one or more symptoms of a ROS1-associated cancer, and/or a patient that has an increased risk of developing a ROS1-associated cancer).
  • dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same can be identified using a liquid biopsy (variously referred to as a fluid biopsy or fluid phase biopsy).
  • a liquid biopsy (variously referred to as a fluid biopsy or fluid phase biopsy). See, e.g., Karachialiou et al., “Real-time liquid biopsies become a reality in cancer treatment”, Ann. Transl. Med., 3(3):36, 2016.
  • Liquid biopsy methods can be used to detect total tumor burden and/or the dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same.
  • Liquid biopsies can be performed on biological samples obtained relatively easily from a subject (e.g., via a simple blood draw) and are generally less invasive than traditional methods used to detect tumor burden and/or dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same.
  • liquid biopsies can be used to detect the presence of dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same at an earlier stage than traditional methods.
  • the biological sample to be used in a liquid biopsy can include, blood, plasma, urine, cerebrospinal fluid, saliva, sputum, broncho-alveolar lavage, bile, lymphatic fluid, cyst fluid, stool, ascites, and combinations thereof.
  • a liquid biopsy can be used to detect circulating tumor cells (CTCs).
  • CTCs circulating tumor cells
  • a liquid biopsy can be used to detect cell-free DNA.
  • cell-free DNA detected using a liquid biopsy is circulating tumor DNA (ctDNA) that is derived from tumor cells.
  • Analysis of ctDNA can be used to identify dysregulation of a RET gene, a RET kinase, or the expression or activity or level of any of the same.
  • NGS next-generation sequencing
  • RET RET kinase
  • ctDNA derived from a single gene can be detected using a liquid biopsy.
  • ctDNA derived from a plurality of genes e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more, or any number of genes in between these numbers
  • a liquid biopsy e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more, or any number of genes in between these numbers
  • ctDNA derived from a plurality of genes can be detected using any of a variety of commercially-available testing panels (e.g., commercially-available testing panels designed to detect dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same ).
  • Liquid biopsies can be used to detect dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same including, without limitation, point mutations or single nucleotide variants (SNVs), copy number variants (CNVs), genetic fusions (e.g., translocations or rearrangements), insertions, deletions, or any combination thereof.
  • a liquid biopsy can be used to detect a germline mutation. In some embodiments, a liquid biopsy can be used to detect a somatic mutation. In some embodiments, a liquid biopsy can be used to detect a primary genetic mutation (e.g., a primary mutation or a primary fusion that is associated with initial development of a disease, e.g., cancer). In some embodiments, a liquid biopsy can be used to detect a genetic mutation that develops after development of the primary genetic mutation (e.g., a resistance mutation that arises in response to a treatment administered to a subject).
  • a primary genetic mutation e.g., a primary mutation or a primary fusion that is associated with initial development of a disease, e.g., cancer.
  • a liquid biopsy can be used to detect a genetic mutation that develops after development of the primary genetic mutation (e.g., a resistance mutation that arises in response to a treatment administered to a subject).
  • a dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same identified using a liquid biopsy is also present in a cancer cell that is present in the subject (e.g., in a tumor).
  • any of the types of dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same described herein can be detected using a liquid biopsy.
  • a genetic mutation identified via a liquid biopsy can be used to identify the subject as a candidate for a particular treatment.
  • detection of dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same in the subject can indicate that the subject will be responsive to a treatment that includes administration of a compound of Formula I or a pharmaceutically acceptable salt thereof.
  • Liquid biopsies can be performed at multiple times during a course of diagnosis, a course of monitoring, and/or a course of treatment to determine one or more clinically relevant parameters including, without limitation, progression of the disease, efficacy of a treatment, or development of resistance mutations after administering a treatment to the subject.
  • a first liquid biopsy can be performed at a first time point and a second liquid biopsy can be performed at a second time point during a course of diagnosis, a course of monitoring, and/or a course of treatment.
  • the first time point can be a time point prior to diagnosing a subject with a disease (e.g., when the subject is healthy), and the second time point can be a time point after subject has developed the disease (e.g., the second time point can be used to diagnose the subject with the disease).
  • the first time point can be a time point prior to diagnosing a subject with a disease (e.g., when the subject is healthy), after which the subject is monitored, and the second time point can be a time point after monitoring the subject.
  • the first time point can be a time point after diagnosing a subject with a disease, after which a treatment is administered to the subject, and the second time point can be a time point after the treatment is administered; in such cases, the second time point can be used to assess the efficacy of the treatment (e.g., if the genetic mutation(s) detected at the first time point are reduced in abundance or are undetectable) or to determine the presence of a resistance mutation that has arisen as a result of the treatment.
  • a treatment to be administered to a subject can include a compound of Formula I or a pharmaceutically acceptable salt thereof.
  • compositions provided herein may be, for example, surgery, radiotherapy, and chemotherapeutic agents, such as kinase inhibitors, signal transduction inhibitors and/or monoclonal antibodies.
  • chemotherapeutic agents such as kinase inhibitors, signal transduction inhibitors and/or monoclonal antibodies.
  • Compounds of Formula I therefore may also be useful as adjuvants to cancer treatment, that is, they can be used in combination with one or more additional therapies or therapeutic agents, for example a chemotherapeutic agent that works by the same or by a different mechanism of action.
  • the compound of Formula I (or a pharmaceutically acceptable salt or solvate thereof) is administered in combination with a therapeutically effective amount of at least one additional therapeutic agent selected from one or more additional therapies or therapeutic (e.g., chemotherapeutic) agents.
  • Non-limiting examples of additional therapeutic agents include: other ROS1-targeted therapeutic agents (i.e. a first or second ROS1 kinase inhibitor), ALK-targeted therapeutic agents (e.g., ALK kinase inhibitors), receptor tyrosine kinase-targeted therapeutic agents (e.g., TRK kinase inhibitors), kinase targeted therapeutics, signal transduction pathway inhibitors, checkpoint inhibitors, modulators of the apoptosis pathway (e.g. obataclax); cytotoxic chemotherapeutics, angiogenesis-targeted therapies, immune-targeted agents, including immunotherapy, and radiotherapy.
  • ROS1-targeted therapeutic agents i.e. a first or second ROS1 kinase inhibitor
  • ALK-targeted therapeutic agents e.g., ALK kinase inhibitors
  • receptor tyrosine kinase-targeted therapeutic agents e.g
  • the other ROS1-targeted therapeutic is a multikinase inhibitor exhibiting ROS1 inhibition activity. In some embodiments, the other ROS1-targeted therapeutic inhibitor is selective for a ROS1 kinase.
  • Exemplary ROS1 kinase inhibitors can exhibit inhibition activity (IC 50 ) against a ROS1 kinase of less than about 1000 nM, less than about 500 nM, less than about 200 nM, less than about 100 nM, less than about 50 nM, less than about 25 nM, less than about 10 nM, or less than about 1 nM as measured in an assay as described herein.
  • a ROS1 kinase inhibitor can exhibit inhibition activity (IC 50 ) against a ROS1 kinase of less than about 25 nM, less than about 10 nM, less than about 5 nM, or less than about 1 nM as measured in an assay as provided herein.
  • ROS1-targeted therapeutic agents include (E)-5-chloro-2-(2-(1-(4-fluorophenyl)ethylidene)hydrazinyl)-N-(2-(isopropylsulfonyl)phenyl)pyrimidin-4-amine ( Eur. J. Org. Chem.
  • alectinib alectinib; brigatinib; cabozantinib; ceritinib; crizotinib; entrectinib; foretinib; herbimycin A; lorlatinib; lorlatinib des-methyl analogs; merestinib; ASP3026 (NCT01284192; Astellas Pharma); AZD3634 (AstraZeneca); and ASP3026 (Astrellas Pharma).
  • an ALK-targeted therapeutic is a multikinase inhibitor exhibiting ALK inhibition activity.
  • the ALK-targeted therapeutic inhibitor is selective for an ALK kinase.
  • Exemplary ALK kinase inhibitors can exhibit inhibition activity (IC 50 ) against an ALK kinase of less than about 1000 nM, less than about 500 nM, less than about 200 nM, less than about 100 nM, less than about 50 nM, less than about 25 nM, less than about 10 nM, or less than about 1 nM as measured in an assay as described herein.
  • an ALK kinase inhibitor can exhibit inhibition activity (IC 50 ) against an ALK kinase of less than about 25 nM, less than about 10 nM, less than about 5 nM, or less than about 1 nM as measured in an assay.
  • Non-limiting examples of ALK-targeted therapeutic agents include “Amgen 36”; “Amgen 49”; “Cephalon 30”; “Chugai 13d”; 4-arylaminopyrimidine derivatives (see, e.g., Eur. J. Med. Chem.
  • a receptor tyrosine kinase targeted therapeutic is a multikinase inhibitor (e.g., TRK-targeted therapeutic inhibitor) exhibiting TRK inhibition activity.
  • the TRK-targeted therapeutic inhibitor is selective for a TRK kinase.
  • Exemplary TRK kinase inhibitors can exhibit inhibition activity (IC 50 ) against a TRK kinase of less than about 1000 nM, less than about 500 nM, less than about 200 nM, less than about 100 nM, less than about 50 nM, less than about 25 nM, less than about 10 nM, or less than about 1 nM as measured in an assay as described herein.
  • a TRK kinase inhibitor can exhibit inhibition activity (IC 50 ) against a TRK kinase of less than about 25 nM, less than about 10 nM, less than about 5 nM, or less than about 1 nM as measured in an assay.
  • IC 50 inhibition activity against a TRK kinase of less than about 25 nM, less than about 10 nM, less than about 5 nM, or less than about 1 nM as measured in an assay.
  • a TRK inhibitor assay can be any of those provided in U.S. Pat. No. 8,933,084 (e.g., Example A or B).
  • Non-limiting examples of receptor tyrosine kinase (e.g., Trk) targeted therapeutic agents include afatinib, cabozantinib, cetuximab, crizotinib, dabrafenib, entrectinib, erlotinib, gefitinib, imatinib, lapatinib, lestaurtinib, nilotinib, pazopanib, panitumumab, pertuzumab, sunitinib, trastuzumab, 1-((3 S,4R)-4-(3-fluorophenyl)-1-(2-methoxyethyl)pyrrolidin-3-yl)-3-(4-methyl-3-(2-methylpyrimidin-5-yl)-1-phenyl-1H-pyrazol-5-yl)urea, AG 879, AR-772, AR-786, AR-256, AR-618, AZ-23,
  • Additional Trk targeted therapeutic agents include those described in U.S. Pat. Nos. 8,450,322; 8,513,263; 8,933,084; 8,791,123; 8,946,226; 8,450,322; 8,299,057; and 8,912,194; U.S. Publication No. 2016/0137654; 2015/0166564; 2015/0051222; 2015/0283132; and 2015/0306086; International Publication No.
  • Trk inhibitors can be found in U.S. Pat. No. 8,637,516, International Publication No. WO 2012/034091, U.S. Pat. No. 9,102,671, International Publication No. WO 2012/116217, U.S. Publication No. 2010/0297115, International Publication No. WO 2009/053442, U.S. Pat. No. 8,642,035, International Publication No. WO 2009092049, U.S. Pat. No. 8,691,221, International Publication No. WO2006131952, all of which are incorporated by reference in their entireties herein.
  • Exemplary Trk inhibitors include GNF-4256, described in Cancer Chemother. Pharmacol.
  • Trk inhibitors include those disclosed in U.S. Publication No. 2010/0152219, U.S. Pat. No. 8,114,989, and International Publication No. WO 2006/123113, all of which are incorporated by reference in their entireties herein.
  • Exemplary Trk inhibitors include AZ623, described in Cancer 117(6):1321-1391, 2011; AZD6918, described in Cancer Biol. Ther. 16(3):477-483, 2015; AZ64, described in Cancer Chemother. Pharmacol.
  • a Trk inhibitor can include those described in U.S. Pat. Nos. 7,615,383; 7,384,632; 6,153,189; 6,027,927; 6,025,166; 5,910,574; 5,877,016; and 5,844,092, each of which is incorporated by reference in its entirety.
  • Trk inhibitors include CEP-751, described in Int. J. Cancer 72:672-679, 1997; CT327, described in Acta Derm. Venereol. 95:542-548, 2015; compounds described in International Publication No. WO 2012/034095; compounds described in U.S. Pat. No. 8,673,347 and International Publication No. WO 2007/022999; compounds described in U.S. Pat. No. 8,338,417; compounds described in International Publication No. WO 2016/027754; compounds described in U.S. Pat. No. 9,242,977; compounds described in U.S. Publication No.
  • sunitinib N-(2-diethylaminoethyl)-5-[(Z)-(5-fluoro-2-oxo-1H-indol-3-ylidene)methyl]-2,4-dimethyl-1H-pyrrole-3-carboxamide
  • sunitinib N-(2-diethylaminoethyl)-5-[(Z)-(5-fluoro-2-oxo-1H-indol-3-ylidene)methyl]-2,4-dimethyl-1H-pyrrole-3-carboxamide
  • 4-aminopyrazolylpyrimidines e.g., AZ-23 (((S)-5-chloro-N2-(1-(5-fluoropyridin-2-yl)ethyl)-N4-(5-isopropoxy-1H-pyrazol-3-yl)pyrimidine-2,4-diamine)), as described in J. Med. Chem. 51(15):4672-4684, 2008; PHA-739358 (danusertib), as described in Mol. Cancer Ther.
  • Trk inhibitor to act as a TrkA, TrkB, and/or Trk C inhibitor may be tested using the assays described in Examples A and B in U.S. Pat. No. 8,513,263, which is incorporated herein by reference.
  • signal transduction pathway inhibitors include Ras-Raf-MEK-ERK pathway inhibitors (e.g., binimetinib, selumetinib, encorafinib, sorafenib, trametinib, and vemurafenib), PI3K-Akt-mTOR-S6K pathway inhibitors (e.g.
  • everolimus, rapamycin, perifosine, temsirolimus), and other kinase inhibitors such as baricitinib, brigatinib, capmatinib, danusertib, ibrutinib, milciclib, quercetin, regorafenib, ruxolitinib, semaxanib, AP32788, BLU285, BLU554, INCB39110, INCB40093, INCB50465, INCB52793, INCB54828, MGCD265, NMS-088, NMS-1286937, PF 477736 ((R)-amino-N-[5,6-dihydro-2-(1-methyl-1H-pyrazol-4-yl)-6-oxo-1Hpyrrolo[4,3,2-ef][2,3]benzodiazepin-8-yl]-cyclohexaneacetamide), PLX3397, PLX7486, PLX8394
  • Non-limiting examples of checkpoint inhibitors include ipilimumab, tremelimumab, nivolumab, pidilizumab, MPDL3208A, MEDI4736, MSB0010718C, BMS-936559, BMS-956559, BMS-935559 (MDX-1105), AMP-224, and pembrolizumab.
  • cytotoxic chemotherapeutics are selected from arsenic trioxide, bleomycin, cabazitaxel, capecitabine, carboplatin, cisplatin, cyclophosphamide, cytarabine, dacarbazine, daunorubicin, docetaxel, doxorubicin, etoposide, fluorouracil, gemcitabine, irinotecan, lomustine, methotrexate, mitomycin C, oxaliplatin, paclitaxel, pemetrexed, temozolomide, and vincristine.
  • Non-limiting examples of angiogenesis-targeted therapies include aflibercept and bevacizumab.
  • an immunotherapy refers to an agent that modulates the immune system.
  • an immunotherapy can increase the expression and/or activity of a regulator of the immune system.
  • an immunotherapy can decrease the expression and/or activity of a regulator of the immune system.
  • an immunotherapy can recruit and/or enhance the activity of an immune cell.
  • the immunotherapy is a cellular immunotherapy (e.g., adoptive T-cell therapy, dendritic cell therapy, natural killer cell therapy).
  • the cellular immunotherapy is sipuleucel-T (APC8015; ProvengeTM; Plosker (2011) Drugs 71(1): 101-108).
  • the cellular immunotherapy includes cells that express a chimeric antigen receptor (CAR).
  • the cellular immunotherapy is a CAR-T cell therapy.
  • the CAR-T cell therapy is tisagenlecleucel (KymriahTM).
  • the immunotherapy is an antibody therapy (e.g., a monoclonal antibody, a conjugated antibody).
  • the antibody therapy is bevacizumab (MvastiTM, Avastin®), trastuzumab (Herceptin®), avelumab (Bavencio®), rituximab (MabTheraTM, Rituxan®), edrecolomab (Panorex), daratumuab (Darzalex®), olaratumab (LartruvoTM), ofatumumab (Arzerra®), alemtuzumab (Campath®), cetuximab (Erbitux®), oregovomab, pembrolizumab (Keytruda®), dinutiximab (Unituxin®), obinutuzumab (Gazyva®), tremelimumab (CP-675,206), ramucirumab (C
  • the immunotherapy is an antibody-drug conjugate.
  • the antibody-drug conjugate is gemtuzumab ozogamicin (MylotargTM), inotuzumab ozogamicin (Besponsa®), brentuximab vedotin (Adcetris®), ado-trastuzumab emtansine (TDM-1; Kadcyla®), mirvetuximab soravtansine (IMGN853) or anetumab ravtansine
  • the immunotherapy includes blinatumomab (AMG103; Blincyto®) or midostaurin (Rydapt).
  • the immunotherapy includes a toxin. In some embodiments, the immunotherapy is denileukin diftitox (Ontak®).
  • the immunotherapy is a cytokine therapy.
  • the cytokine therapy is an interleukin 2 (IL-2) therapy, an interferon alpha (IFN ⁇ ) therapy, a granulocyte colony stimulating factor (G-CSF) therapy, an interleukin 12 (IL-12) therapy, an interleukin 15 (IL-15) therapy, an interleukin 7 (IL-7) therapy or an erythropoietin-alpha (EPO) therapy.
  • the IL-2 therapy is aldesleukin (Proleukin®).
  • the IFN ⁇ therapy is IntronA® (Roferon-A®).
  • the G-CSF therapy is filgrastim (Neupogen®).
  • the immunotherapy is an immune checkpoint inhibitor. In some embodiments, the immunotherapy includes one or more immune checkpoint inhibitors. In some embodiments, the immune checkpoint inhibitor is a CTLA-4 inhibitor, a PD-1 inhibitor or a PD-L1 inhibitor. In some embodiments, the CTLA-4 inhibitor is ipilimumab (Yervoy®) or tremelimumab (CP-675,206). In some embodiments, the PD-1 inhibitor is pembrolizumab (Keytruda®) or nivolumab (Opdivo®). In some embodiments, the PD-L1 inhibitor is atezolizumab (Tecentriq®), avelumab (Bavencio®) or durvalumab (ImfinziTM).
  • the immunotherapy is mRNA-based immunotherapy.
  • the mRNA-based immunotherapy is CV9104 (see, e.g., Rausch et al. (2014) Human Vaccin Immunother 10(11): 3146-52; and Kubler et al. (2015) J. Immunother Cancer 3:26).
  • the immunotherapy is bacillus Calmette-Guerin (BCG) therapy.
  • the immunotherapy is an oncolytic virus therapy.
  • the oncolytic virus therapy is talimogene alherparepvec (T-VEC; Imlygic®).
  • the immunotherapy is a cancer vaccine.
  • the cancer vaccine is a human papillomavirus (HPV) vaccine.
  • the HPV vaccine is Gardasil®, Gardasil9® or Cervarix®.
  • the cancer vaccine is a hepatitis B virus (HBV) vaccine.
  • the HBV vaccine is Engerix-B®, Recombivax HB® or GI-13020 (Tarmogen®).
  • the cancer vaccine is Twinrix® or Pediarix®.
  • the cancer vaccine is BiovaxID®, Oncophage®, GVAX, ADXS11-001, ALVAC-CEA, PROSTVAC®, Rindopepimut®, CimaVax-EGF, lapuleucel-T (APC8024; NeuvengeTM), GRNVAC1, GRNVAC2, GRN-1201, hepcortespenlisimut-L (Hepko-V5), DCVAX®, SCIB1, BMT CTN 1401, PrCa VBIR, PANVAC, ProstAtak®, DPX-Survivac, or viagenpumatucel-L (HS-110).
  • the immunotherapy is a peptide vaccine.
  • the peptide vaccine is nelipepimut-S(E75) (NeuVaxTM), IMA901, or SurVaxM (SVN53-67).
  • the cancer vaccine is an immunogenic personal neoantigen vaccine (see, e.g., Ott et al. (2017) Nature 547: 217-221; Sahin et al. (2017) Nature 547: 222-226).
  • the cancer vaccine is RGSH4K, or NEO-PV-01.
  • the cancer vaccine is a DNA-based vaccine.
  • the DNA-based vaccine is a mammaglobin-A DNA vaccine (see, e.g., Kim et al. (2016) Oncolmmunology 5(2): e1069940).
  • immune-targeted agents are selected from aldesleukin, interferon alfa-2b, ipilimumab, lambrolizumab, nivolumab, prednisone, and sipuleucel-T.
  • Non-limiting examples of radiotherapy include radioiodide therapy, external-beam radiation, and radium 223 therapy.
  • Additional kinase inhibitors include those described in, for example, U.S. Pat. Nos. 7,514,446; 7,863,289; 8,026,247; 8,501,756; 8,552,002; 8,815,901; 8,912,204; 9,260,437; 9,273,051; U.S. Publication No. US 2015/0018336; International Publication No.
  • kinase inhibitors include those described in, for example, WO 2016/081450; WO 2016/022569; WO 2016/011141; WO 2016/011144; WO 2016/011147; WO 2015/191667; WO 2012/101029; WO 2012/113774; WO 2015/191666; WO 2015/161277; WO 2015/161274; WO 2015/108992; WO 2015/061572; WO 2015/058129; WO 2015/057873; WO 2015/017528; WO/2015/017533; WO 2014/160521; and WO 2014/011900, each of which is hereby incorporated by reference in its entirety.
  • a kinase inhibitor as provided herein may have activity against more than one kinase (i.e. may be a multikinase inhibitor).
  • a mechanism of action e.g., ROS1, ALK, or TRK kinase inhibition
  • each of the compounds recited are structurally distinct from one another (e.g., the ROS1 inhibitor and the TRK inhibitor are not the same compound).
  • a method of treating cancer comprising administering to a patient in need thereof a pharmaceutical combination for treating cancer which comprises (a) a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof, (b) an additional therapeutic agent, and (c) optionally at least one pharmaceutically acceptable carrier for simultaneous, separate or sequential use for the treatment of cancer, wherein the amounts of the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof and the additional therapeutic agent are together effective in treating the cancer.
  • additional therapeutic agents may be administered with one or more doses of the compound of Formula I, or a pharmaceutically acceptable salt or solvate thereof, or pharmaceutical composition thereof, as part of the same or separate dosage forms, via the same or different routes of administration, and/or on the same or different administration schedules according to standard pharmaceutical practice known to one skilled in the art.
  • a pharmaceutical combination for treating a cancer in a patient in need thereof which comprises (a) a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof, (b) at least one additional therapeutic agent (e.g., any of the exemplary additional therapeutic agents described herein or known in the art), and (c) optionally at least one pharmaceutically acceptable carrier for simultaneous, separate or sequential use for the treatment of cancer, wherein the amounts of the compound of Formula I or pharmaceutically acceptable salt or solvate thereof and of the additional therapeutic agent are together effective in treating the cancer; (ii) a pharmaceutical composition comprising such a combination; (iii) the use of such a combination for the preparation of a medicament for the treatment of cancer; and (iv) a commercial package or product comprising such a combination as a combined preparation for simultaneous, separate or sequential use; and to a method of treatment of cancer in a patient in need thereof.
  • the patient is a human.
  • the cancer is a ROS1-associated cancer
  • pharmaceutical combination refers to a pharmaceutical therapy resulting from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients.
  • fixed combination means that a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof and at least one additional therapeutic agent (e.g., a chemotherapeutic agent), are both administered to a patient simultaneously in the form of a single composition or dosage.
  • non-fixed combination means that a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof and at least one additional therapeutic agent (e.g., chemotherapeutic agent) are formulated as separate compositions or dosages such that they may be administered to a patient in need thereof simultaneously, concurrently or sequentially with variable intervening time limits (e.g., 1 hour, 1 day, 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months), wherein such administration provides effective levels of the two or more compounds in the body of the patient.
  • variable intervening time limits e.g., 1 hour, 1 day, 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months
  • a method of treating a cancer comprising administering to a patient in need thereof a pharmaceutical combination for treating cancer which comprises (a) a compound of Formula I or pharmaceutically acceptable salt or solvate thereof, (b) an additional therapeutic agent, and (c) optionally at least one pharmaceutically acceptable carrier for simultaneous, separate or sequential use for the treatment of cancer, wherein the amounts of the compound of Formula I or pharmaceutically acceptable salt or solvate thereof and the additional therapeutic agent are together effective in treating the cancer.
  • the compound of Formula I or pharmaceutically acceptable salt or solvate thereof, and the additional therapeutic agent are administered simultaneously as separate dosages.
  • the compound of Formula I or pharmaceutically acceptable salt or solvate thereof, and the additional therapeutic agent are administered as separate dosages sequentially in any order, in jointly therapeutically effective amounts, e.g. in daily or intermittently dosages.
  • the compound of Formula I or pharmaceutically acceptable salt or solvate thereof, and the additional therapeutic agent are administered simultaneously as a combined dosage.
  • the cancer is a ROS1-associated cancer.
  • a ROS1-associated cancer having one or more ROSlinhibitor resistance mutations.
  • the disease or disorder mediated by ROS1 is a dysregulation of ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same.
  • the dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same includes one or more ROS1 inhibitor resistance mutations.
  • a disease or disorder mediated by ROS1 can include any disease, disorder or condition that is directly or indirectly linked to expression or activity of ROS1, including overexpression and/or abnormal activity levels.
  • the disease is cancer (e.g., a ROS1-associated cancer).
  • the cancer is any of the cancers or ROS1-associated cancers described herein.
  • tumorigenesis may vary between different cancer types, the cellular and molecular mechanisms required for metastasis appear to be similar for all solid tumor types.
  • the cancer cells lose growth inhibitory responses, undergo alterations in adhesiveness and produce enzymes that can degrade extracellular matrix components. This leads to detachment of tumor cells from the original tumor, infiltration into the circulation through newly formed vasculature, migration and extravasation of the tumor cells at favorable distant sites where they may form colonies.
  • a number of genes have been identified as being promoters or suppressors of metastasis.
  • the method comprising administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof or a pharmaceutical composition thereof.
  • Such methods can be used in the treatment of one or more of the cancers described herein. See, e.g., US Publication No. 2013/0029925; International Publication No. WO 2014/083567; and U.S. Pat. No. 8,568,998.
  • the cancer is a ROS1-associated cancer.
  • the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof is used in combination with an additional therapy or another therapeutic agent, including a chemotherapeutic agent, such as a kinase inhibitor, for example, a first or second ROS1 kinase inhibitor.
  • a chemotherapeutic agent such as a kinase inhibitor, for example, a first or second ROS1 kinase inhibitor.
  • tumor is an art known term and means the formation of an additional tumor (e.g., a solid tumor) at a site distant from a primary tumor in a subject or patient, where the additional tumor includes the same or similar cancer cells as the primary tumor.
  • additional tumor e.g., a solid tumor
  • Also provided are methods of decreasing the risk of developing a metastasis or an additional metastasis in a patient having a ROS1-associated cancer that include: selecting, identifying, or diagnosing a patient as having a ROS1-associated cancer, and administering a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof to the patient selected, identified, or diagnosed as having a ROS1-associated cancer. Also provided are methods of decreasing the risk of developing a metastasis or an additional metastasis in a patient having a ROS1-associated cancer that includes administering a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvent thereof to a patient having a ROS1-associated cancer.
  • the decrease in the risk of developing a metastasis or an additional metastasis in a patient having a ROS1-associated cancer can be compared to the risk of developing a metastasis or an additional metastasis in the patient prior to treatment, or as compared to a patient or a population of patients having a similar or the same ROS1-associated cancer that has received no treatment or a different treatment.
  • the ROS1-associated cancer is a ROS1-associated cancer having one or more ROSlinhibitor resistance mutations.
  • risk of developing a metastasis means the risk that a subject or patient having a primary tumor will develop an additional tumor (e.g., a solid tumor) at a site distant from a primary tumor in a subject or patient over a set period of time, where the additional tumor includes the same or similar cancer cells as the primary tumor.
  • additional tumor e.g., a solid tumor
  • risk of developing additional metastases means the risk that a subject or patient having a primary tumor and one or more additional tumors at sites distant from the primary tumor (where the one or more additional tumors include the same or similar cancer cells as the primary tumor) will develop one or more further tumors distant from the primary tumor, where the further tumors include the same or similar cancer cells as the primary tumor. Methods for reducing the risk of developing additional metastasis are described herein.
  • the presence of one or more ROS1 inhibitor resistance mutations in a tumor causes the tumor to be more resistant to treatment with a first ROS1 inhibitor.
  • Methods useful when a ROS1 inhibitor resistance mutation causes the tumor to be more resistant to treatment with a first ROS1 inhibitor are described below.
  • methods of treating a subject having a cancer that include: identifying a subject having a cancer cell that has one or more ROS1 inhibitor resistance mutations; and administering to the identified subject a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof is administered in combination with the first ROS1 inhibitor.
  • the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof is administered in combination with the first ROS1 inhibitor.
  • the one or more ROS1 inhibitor resistance mutations confer increased resistance to a cancer cell or tumor to treatment with the first ROS1 inhibitor.
  • the one or more ROS1 inhibitor resistance mutations include one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • the one or more ROS1 inhibitor resistance mutations can include a substitution at amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • methods for treating a ROS1-associated cancer in a subject in need of such treatment comprising (a) detecting a dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a first ROS1 inhibitor.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (d) administering a compound of Formula I, or a pharmaceutically acceptable salt of solvate thereof as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (e) administering additional doses of the first ROS1 inhibitor of step (b) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • provided herein are methods for treating a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting a dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a first ROS1 inhibitor.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (d) administering a compound of Formula I selected from Example No.
  • provided herein are methods for treating a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting one or more fusion proteins of Table 2 and/or one or more ROS1 kinase protein point mutations, insertions, and/or deletions (e.g., one or more point mutations of Table 3 or Table 3a) in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a first ROS1 inhibitor.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations of Table 4; and (d) administering a compound of Formula I selected from Example No. 2, 3, 7, 9, 14, 19, 20, 22, 33-A, 33-B, 35, 36, and 45, or a pharmaceutically acceptable salt or solvate thereof as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (e) administering additional doses of the first ROS1 inhibitor of step (b) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • provided herein are methods for treating a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting one or more of the fusion proteins SLC34A2-ROS1, CD74-ROS1, EZR-ROS1, TPM3-ROS1, or SDC4-ROS1 in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a first ROS1 inhibitor.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more of the ROS1 inhibitor resistance mutations L2026M, G2032R, or D2033N; and (d) administering a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof selected from the group consisting of a compound of Formula I selected from Example No.
  • a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting a dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a first ROS1 inhibitor, wherein the first ROS1 inhibitor is selected from the group consisting of alectinib, brigatinib, cabozantinib, ceritinib, crizotinib, entrectinib, foretinib, lorlatinib, and mesestinib.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (d) administering a compound of Formula I, or a pharmaceutically acceptable salt of solvate thereof as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (e) administering additional doses of the first ROS1 inhibitor of step (b) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • provided herein are methods for treating a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting a dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a first ROS1 inhibitor, wherein the first ROS1 inhibitor is selected from the group consisting of alectinib, brigatinib, cabozantinib, ceritinib, crizotinib, entrectinib, foretinib, lorlatinib, and mesestinib.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (d) administering a compound of Formula I selected from Example No. 2, 3, 7, 9, 14, 19, 20, 22, 33-A, 33-B, 35, 36, and 45, or a pharmaceutically acceptable salt or solvate thereof as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (e) administering additional doses of the first ROS1 inhibitor of step (b) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • provided herein are methods for treating a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting one or more fusion proteins of Table 2 and/or one or more ROS1 kinase protein point mutations, insertions, and/or deletions (e.g., one or more point mutations of Table 3 or Table 3a) in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a first ROS1 inhibitor, wherein the first ROS1 inhibitor is selected from the group consisting of alectinib, brigatinib, cabozantinib, ceritinib, crizotinib, entrectinib, foretinib, lorlatinib, and mesestinib.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations of Table 4; and (d) administering a compound of Formula I selected from Example No. 2, 3, 7, 9, 14, 19, 20, 22, 33-A, 33-B, 35, 36, and 45, or a pharmaceutically acceptable salt or solvate thereof as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (e) administering additional doses of the first ROS1 inhibitor of step (b) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • provided herein are methods for treating a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting one or more of the fusion proteins SLC34A2-ROS1, CD74-ROS1, EZR-ROS1, TPM3-ROS1, or SDC4-ROSlin a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a first ROS1 inhibitor, wherein the first ROS1 inhibitor is selected from the group consisting of alectinib, brigatinib, cabozantinib, ceritinib, crizotinib, entrectinib, foretinib, lorlatinib, and mesestinib.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more of the ROS1 inhibitor resistance mutations L2026M, G2032R, or D2033N; and (d) administering a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof selected from the group consisting of a compound of Formula I selected from Example No.
  • methods for treating a ROS1-associated cancer in a subject in need of such treatment comprising (a) detecting a dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt of solvate thereof.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (d) administering a second ROS1 inhibitor, as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (e) administering additional doses of the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof of step (b) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • provided herein are methods for treating a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting a dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a compound of Formula I selected from Example No. 2, 3, 7, 9, 14, 19, 20, 22, 33-A, 33-B, 35, 36, and 45, or a pharmaceutically acceptable salt or solvate thereof.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (d) administering a second ROS1 inhibitor, as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (e) administering additional doses of the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof of step (b) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • provided herein are methods for treating a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting one or more fusion proteins of Table 2 and/or one or more ROS1 kinase protein point mutations, insertions, and/or deletions (e.g., one or more of the point mutations of Table 3 or Table 3a) in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a compound of Formula I selected from Example No. 2, 3, 7, 9, 14, 19, 20, 22, 33-A, 33-B, 35, 36, and 45, or a pharmaceutically acceptable salt or solvate thereof.
  • a compound of Formula I selected from Example No. 2, 3, 7, 9, 14, 19, 20, 22, 33-A, 33-B, 35, 36, and 45, or a pharmaceutically acceptable salt or solvate thereof.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations of Table 4; and (d) administering a second ROS1 inhibitor, as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (e) administering additional doses of the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof of step (b) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • provided herein are methods for treating a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting one or more of the fusion proteins SLC34A2-ROS1, CD74-ROS1, EZR-ROS1, TPM3-ROS1, or SDC4-ROS1 in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a compound of Formula I selected Example No. 2, 3, 7, 9, 14, 19, 20, 22, 33-A, 33-B, 35, 36, and 45, or a pharmaceutically acceptable salt or solvate thereof.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more of the ROS1 inhibitor resistance mutations L2026M, G2032R, or D2033N; and (d) administering a second ROS1 inhibitor, as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (e) administering additional doses of the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof of step (b) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • provided herein are methods for treating a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting a dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt of solvate thereof.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (d) administering a second ROS1 inhibitor, wherein the second ROS1 inhibitor is selected from the group consisting of alectinib, brigatinib, cabozantinib, ceritinib, crizotinib, entrectinib, foretinib, lorlatinib, and mesestinib, as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (e) administering additional doses of the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof of step (b) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • the second ROS1 inhibitor is selected from the group consisting of alectinib
  • provided herein are methods for treating a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting a dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a compound of Formula I selected from Example No. 2, 3, 7, 9, 14, 19, 20, 22, 33-A, 33-B, 35, 36, and 45, or a pharmaceutically acceptable salt or solvate thereof.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (d) administering a second ROS1 inhibitor, wherein the second ROS1 inhibitor is selected from the group consisting of alectinib, brigatinib, cabozantinib, ceritinib, crizotinib, entrectinib, foretinib, lorlatinib, and mesestinib, as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (e) administering additional doses of the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof of step (b) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • the second ROS1 inhibitor is selected from the group consisting of alectinib
  • provided herein are methods for treating a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting one or more fusion proteins of Table 2 and/or one or more ROS1 kinase protein point mutations, insertions, and/or deletions (e.g., one or more of the point mutations of Table 3 or Table 3a) in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a compound of Formula I selected from Example No. 2, 3, 7, 9, 14, 19, 20, 22, 33-A, 33-B, 35, 36, and 45, or a pharmaceutically acceptable salt or solvate thereof.
  • a compound of Formula I selected from Example No. 2, 3, 7, 9, 14, 19, 20, 22, 33-A, 33-B, 35, 36, and 45, or a pharmaceutically acceptable salt or solvate thereof.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations of Table 4; and (d) administering a second ROS1 inhibitor, wherein the second ROS1 inhibitor is selected from the group consisting of alectinib, brigatinib, cabozantinib, ceritinib, crizotinib, entrectinib, foretinib, lorlatinib, and mesestinib, as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (e) administering additional doses of the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof of step (b) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • the second ROS1 inhibitor is selected from the group consisting of alect
  • provided herein are methods for treating a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting one or more of the fusion proteins SLC34A2-ROS1, CD74-ROS1, EZR-ROS1, TPM3-ROS1, or SDC4-ROS1 in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a compound of Formula I selected Example No. 2, 3, 7, 9, 14, 19, 20, 22, 33-A, 33-B, 35, 36, and 45, or a pharmaceutically acceptable salt or solvate thereof.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more of the ROS1 inhibitor resistance mutations L2026M, G2032R, or D2033N; and (d) administering a second ROS1 inhibitor, wherein the second ROS1 inhibitor is selected from the group consisting of alectinib, brigatinib, cabozantinib, ceritinib, crizotinib, entrectinib, foretinib, lorlatinib, and mesestinib, as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (e) administering additional doses of the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof of step (b) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting a dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (d) administering additional doses of the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof of step (b) to the subject as a monotherapy or in conjunction with another anticancer agent (e.g., a second ROS1 inhibitor, a second compound of Formula I, an ALK inhibitor, a TRK inhibitor, or a pharmaceutically acceptable salt thereof) or anticancer therapy (e.g., surgery or radiation) if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations.
  • another anticancer agent e.g., a second ROS1 inhibitor, a second compound of Formula I, an ALK inhibitor, a TRK inhibitor, or a pharmaceutically acceptable salt thereof
  • anticancer therapy e.g., surgery or radiation
  • provided herein are methods for treating a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting a dysregulation of a ROS1 gene, a ROS1 kinase, or the expression or activity or level of any of the same in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a compound of Formula I selected from Example No. 2, 3, 7, 9, 14, 19, 20, 22, 33-A, 33-B, 35, 36, and 45, or a pharmaceutically acceptable salt or solvate thereof.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (d) administering additional doses of the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof of step (b) to the subject as a monotherapy or in conjunction with another anticancer agent (e.g., a second ROS1 inhibitor, a second compound of Formula I, an ALK inhibitor, a TRK inhibitor, or a pharmaceutically acceptable salt thereof) or anticancer therapy (e.g., surgery or radiation) if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations.
  • another anticancer agent e.g., a second ROS1 inhibitor, a second compound of Formula I, an ALK inhibitor, a TRK inhibitor, or a pharmaceutically acceptable salt thereof
  • anticancer therapy e.g., surgery or radiation
  • provided herein are methods for treating a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting one or more ROS1 fusion proteins of Table 2 and/or one or more ROS1 kinase protein point mutations, insertions, and/or deletions (e.g., one or more of the point mutations of Table 3 or Table 3a) in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof selected from the group consisting of a compound of Formula I selected from Example No.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations of Table 4; and (d) administering additional doses of the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof of step (b) to the subject as a monotherapy or in conjunction with another anticancer agent (e.g., a second ROS1 inhibitor, a second compound of Formula I, an ALK inhibitor, a TRK inhibitor, or a pharmaceutically acceptable salt thereof) or anticancer therapy (e.g., surgery or radiation) if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations.
  • another anticancer agent e.g., a second ROS1 inhibitor, a second compound of Formula I, an ALK inhibitor, a TRK inhibitor, or a pharmaceutically acceptable salt thereof
  • anticancer therapy e.g., surgery or radiation
  • a second ROS1 inhibitor selected from the group consisting of alectinib, brigatinib, cabozantinib, ceritinib, crizotinib, entrectinib, foretinib, lorlatinib, and mesestinib is administered in step (d).
  • provided herein are methods for treating a ROS1-associated cancer in a subject in need of such treatment, the method comprising (a) detecting one or more of the fusion proteins SLC34A2-ROS1, CD74-ROS1, EZR-ROS1, TPM3-ROS1, or SDC4-ROS1 in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a compound of Formula I selected Example No. 2, 3, 7, 9, 14, 19, 20, 22, 33-A, 33-B, 35, 36, and 45, or a pharmaceutically acceptable salt or solvate thereof.
  • the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has one or more of the ROS1 inhibitor resistance mutations L2026M, G2032R, or D2033N; and (d) administering additional doses of the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof of step (b) to the subject as a monotherapy or in conjunction with another anticancer agent (e.g., a second ROS1 inhibitor, a second compound of Formula I, an ALK inhibitor, a TRK inhibitor, or a pharmaceutically acceptable salt thereof) or anticancer therapy (e.g., surgery or radiation) if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations.
  • another anticancer agent e.g., a second ROS1 inhibitor, a second compound of Formula I, an ALK inhibitor, a TRK inhibitor, or a pharmaceutically acceptable salt thereof
  • anticancer therapy e.g., surgery or radiation
  • a second ROS1 inhibitor selected from the group consisting of alectinib, brigatinib, cabozantinib, ceritinib, crizotinib, entrectinib, foretinib, lorlatinib, and mesestinib is administered in step (d).
  • Also provided are methods of selecting a treatment for a subject having a cancer that include: identifying a subject having a cancer cell that has one or more ROS1 inhibitor resistance mutations; and selecting a treatment that includes administration of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • the one or more ROS1 inhibitor resistance mutations confer increased resistance to a cancer cell or tumor to treatment with a first ROS1 inhibitor.
  • the compound of Formula I or a pharmaceutically acceptable salt or solvate thereof is administered in combination with the first ROS1 inhibitor.
  • Also provided are methods of selecting a treatment for a subject having a cancer that include: selecting a treatment that includes administration of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof for a subject identified as having a cancer cell that has one or more ROS1 inhibitor resistance mutations. Also provided are methods of selecting a subject having a cancer for a treatment that does not include a first ROS1 inhibitor as a monotherapy that include: identifying a subject having a cancer cell that has one or more ROS1 inhibitor resistance mutations; and selecting the identified subject for a treatment that includes a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • Also provided are methods of selecting a subject having a cancer for a treatment that does not include a first ROS1 inhibitor as a monotherapy that include: selecting a subject identified as having a cancer cell that has one or more ROS1 inhibitor resistance mutations for a treatment that includes administration of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • the one or more ROS1 inhibitor resistance mutations include one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • the one or more ROS1 inhibitor resistance mutations can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • Also provided are methods of determining the likelihood that a subject having a cancer e.g., a ROS1-associated cancer
  • determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and determining that a subject having a cancer cell that has one or more ROS1 inhibitor resistance mutations has a decreased likelihood of having a positive response (i.e. an increased likelihood of having a negative response) to treatment with a first ROS1 inhibitor as a monotherapy.
  • Also provided are methods of determining the likelihood that a subject having a cancer e.g., a ROS1-associated cancer
  • determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and determining that a subject not having a cancer cell that has one or more ROS1 inhibitor resistance mutations has an increased likelihood of having a positive response to treatment with a first ROS1 inhibitor as a monotherapy as compared to a subject having a cancer cell that has one or more ROS1 inhibitor resistance mutations.
  • Also provided are methods of predicting the efficacy of treatment with a first ROS1 inhibitor as a monotherapy in a subject having cancer that include: determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and determining that treatment with a first ROS1 inhibitor as a monotherapy is less likely to be effective in a subject having a cancer cell in a sample obtained from the subject that has one or more ROS1 inhibitor resistance mutations.
  • Also provided are methods of predicting the efficacy of treatment with a first ROS1 inhibitor as a monotherapy in a subject having cancer that include: determining that treatment with a first ROS1 inhibitor as a monotherapy is less likely to be effective in a subject having a cancer cell in a sample obtained from the subject that has one or more ROS1 inhibitor resistance mutations.
  • the one or more ROS1 inhibitor resistance mutations confer increased resistance to a cancer cell or tumor to treatment with the first ROS1 inhibitor.
  • the one or more ROS1 inhibitor resistance mutations include one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • the one or more ROS1 inhibitor resistance mutations can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • Also provided are methods of treating a subject having a cancer that include: (a) administering a first ROS1 inhibitor to the subject for a period of time (e.g., 1 month, 2 months, 3 months, 6 months, 9 months, 1 year); (b) after (a), determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (c) administering a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (d) administering additional doses of the first ROS1 inhibitor of step (a) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • the subject can also be administered another anticancer agent (e.g., a second ROS1 inhibitor, an ALK inhibitor, a TRK inhibitor, or a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof).
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent can be another ROS1 inhibitor (e.g., a second ROS1 inhibitor).
  • another anticancer agent can be the first ROS1 inhibitor administered in step (a).
  • the one or more ROS1 inhibitor resistance mutations confer increased resistance to a cancer cell or tumor to treatment with the first ROS1 inhibitor.
  • the one or more ROS1 inhibitor resistance mutations include one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • the one or more ROS1 inhibitor resistance mutations can include a substitution at one or more of amino acid positions 2026, 2032, or 2032, e.g., L2026M, G2032R, or D2033N.
  • Also provided are methods of treating a subject having a cancer that include: (a) administering a first ALK inhibitor to the subject for a period of time (e.g., 1 month, 2 months, 3 months, 6 months, 9 months, 1 year); (b) after (a), determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (c) administering a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has one or more ROS1 inhibitor resistance mutations; or (d) administering additional doses of the first ALK inhibitor of step (a) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • the subject can also be administered another anticancer agent (e.g., a second ALK inhibitor, a first ROS1 inhibitor, a TRK inhibitor, or a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof).
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent can be another ALK inhibitor (e.g., a second ALK inhibitor).
  • another anticancer agent can be the first ALK inhibitor administered in step (a).
  • another anticancer agent can be another ROS1 inhibitor.
  • the ROS1 inhibitor resistance mutation includes one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • a ROS1 inhibitor resistance mutation can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • Also provided are methods of treating a subject having a cancer that include: (a) administering a first TRK inhibitor to the subject for a period of time (e.g., 1 month, 2 months, 3 months, 6 months, 9 months, 1 year); (b) after (a), determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (c) administering a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has one or more ROS1 inhibitor resistance mutations; or (d) administering additional doses of the first TRK inhibitor of step (a) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • the subject can also be administered another anticancer agent (e.g., a second TRK inhibitor, a first ROS1 inhibitor, an ALK inhibitor, or a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof).
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent can be another TRK inhibitor (e.g., a second TRK inhibitor).
  • another anticancer agent can be the first TRK inhibitor administered in step (a).
  • another anticancer agent can be another ROS1 inhibitor.
  • the dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same confers increased resistance to a cancer cell or tumor to treatment with the first TRK inhibitor.
  • the ROS1 inhibitor resistance mutation includes one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • a ROS1 inhibitor resistance mutation can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • Also provided are methods of treating a subject having a cancer that include: (a) administering a first ROS1 inhibitor to the subject for a period of time (e.g., 1 month, 2 months, 3 months, 6 months, 9 months, 1 year); (b) after (a), determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (c) administering a second ROS1 inhibitor as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (d) administering additional doses of the first ROS1 inhibitor step (a) to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • the subject can also be administered another anticancer agent.
  • the one or more ROS1 inhibitor resistance mutations confer increased resistance to a cancer cell or tumor to treatment with the first ROS1 inhibitor.
  • the one or more ROS1 inhibitor resistance mutations include one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • the one or more ROS1 inhibitor resistance mutations can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent can be another ROS1 inhibitor (e.g., a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof).
  • Also provided are methods of treating a subject having a cancer e.g., a ROS1-associated cancer
  • a cancer e.g., a ROS1-associated cancer
  • methods of treating a subject having a cancer that include: (a) determining whether a cancer cell in a sample obtained from a subject having a cancer and previously administered a first ROS1 inhibitor, has one or more ROS1 inhibitor resistance mutations; and (b) administering a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (c) administering additional doses of the first ROS1 inhibitor previously administered to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • a cancer e.g., a ROS1-associated cancer
  • the subject can also be administered another anticancer agent (e.g., a second ROS1 inhibitor, an ALK inhibitor, a TRK inhibitor, or a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof).
  • another anticancer agent e.g., a second ROS1 inhibitor, an ALK inhibitor, a TRK inhibitor, or a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • the one or more ROS1 inhibitor resistance mutations confer increased resistance to a cancer cell or tumor to treatment with the first ROS1 inhibitor.
  • the one or more ROS1 inhibitor resistance mutations include one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • the one or more ROS1 inhibitor resistance mutations can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent can be another ROS1 inhibitor (e.g., a second ROS1 inhibitor).
  • another anticancer agent can be the first ROS1 inhibitor administered in step (a).
  • Also provided are methods of treating a subject having a cancer e.g., a ROS1-associated cancer
  • a cancer e.g., a ROS1-associated cancer
  • methods of treating a subject having a cancer that include: (a) determining whether a cancer cell in a sample obtained from a subject having a cancer and previously administered a first ALK inhibitor has one or more ROS1 inhibitor resistance mutations; and (b) administering a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell having one or more ROS1 inhibitor resistance mutations; or (c) administering additional doses of the first ALK inhibitor previously administered to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • a cancer e.g., a ROS1-associated cancer
  • the subject can also be administered another anticancer agent (e.g., a second ALK inhibitor, a TRK inhibitor, a first ROS1 inhibitor, or a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof).
  • the ROS1 inhibitor resistance mutation includes one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • a ROS1 inhibitor resistance mutation can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent is can be another ALK inhibitor (e.g., a second ALK inhibitor).
  • another anticancer agent can be the first ALK inhibitor administered in step (a).
  • another anticancer agent can be another ROS1 inhibitor.
  • Also provided are methods of treating a subject having a cancer e.g., a ROS1-associated cancer
  • a cancer e.g., a ROS1-associated cancer
  • methods of treating a subject having a cancer that include: (a) determining whether a cancer cell in a sample obtained from a subject having a cancer and previously administered a first TRK inhibitor is associated with a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same; and (b) administering a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (c) administering additional doses of the first TRK inhibitor previously administered to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • a cancer e.g., a ROS1
  • the subject can also be administered another anticancer agent (e.g., a second TRK inhibitor, an ALK inhibitor, a first ROS1 inhibitor, or a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof).
  • the ROS1 inhibitor resistance mutation includes one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • a ROS1 inhibitor resistance mutation can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent is can be another TRK inhibitor (e.g., a second TRK inhibitor).
  • another anticancer agent can be the first TRK inhibitor administered in step (a).
  • another anticancer agent can be another ROS1 inhibitor.
  • Also provided are methods of treating a subject having a cancer that include: (a) determining whether a cancer cell in a sample obtained from a subject having a cancer and previously administered a first ROS1 inhibitor has one or more ROS1 inhibitor resistance mutations; and (b) administering a second ROS1 inhibitor as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (c) administering additional doses of the first ROS1 inhibitor previously administered to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • the subject can also be administered another anticancer agent.
  • the one or more ROS1 inhibitor resistance mutations confer increased resistance to a cancer cell or tumor to treatment with the first ROS1 inhibitor.
  • the one or more ROS1 inhibitor resistance mutations include one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • the one or more ROS1 inhibitor resistance mutations can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent can be another ROS1 inhibitor (e.g., a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof).
  • another anticancer agent can be the first ROS1 inhibitor administered in step (a).
  • Also provided are methods of treating a subject having a cancer that include: (a) determining whether a cancer cell in a sample obtained from a subject having a cancer and previously administered a first ALK inhibitor has one or more ROS1 inhibitor resistance mutations; and (b) administering a ROS1 inhibitor as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (c) administering additional doses of the first ALK inhibitor previously administered to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • the subject can also be administered another anticancer agent.
  • the ROS1 inhibitor resistance mutation includes one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • a ROS1 inhibitor resistance mutation can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent can be a ROS1 inhibitor (e.g., a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof).
  • another anticancer agent can be the first ALK inhibitor administered in step (a).
  • Also provided are methods of treating a subject having a cancer that include: (a) determining whether a cancer cell in a sample obtained from a subject having a cancer and previously administered a first TRK inhibitor is associated with a dysregulation of a ROS1 gene, a ROS1 kinase, or expression or activity or level of any of the same; and (b) administering a ROS1 inhibitor as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (c) administering additional doses of the first TRK inhibitor previously administered to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • the subject can also be administered another anticancer agent.
  • the ROS1 inhibitor resistance mutation includes one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • a ROS1 inhibitor resistance mutation can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent can be a ROS1 inhibitor (e.g., a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof).
  • another anticancer agent can be the first TRK inhibitor administered in step (a).
  • Also provided are methods of selecting a treatment for a subject having a cancer that include (a) administering a first ROS1 inhibitor to the subject for a period of time (e.g., 1 month, 2 months, 3 months, 6 months, 9 months, 1 year); (b) after (a), determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (c) selecting a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy or in conjunction with another anticancer agent for the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (d) selecting additional doses of the first ROS1 inhibitor of step (a) for the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • the method can further include selecting doses of another anticancer agent for the subject.
  • the one or more ROS1 inhibitor resistance mutations confer increased resistance to a cancer cell or tumor to treatment with the first ROS1 inhibitor.
  • the one or more ROS1 inhibitor resistance mutations include one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • the one or more ROS1 inhibitor resistance mutations can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent can be another ROS1 inhibitor (e.g., a second ROS1 inhibitor).
  • another ROS1 inhibitor can be the first ROS1 inhibitor administered in step (a).
  • Also provided are methods of selecting a treatment for a subject having a cancer that include (a) administering a first ALK inhibitor to the subject for a period of time (e.g., 1 month, 2 months, 3 months, 6 months, 9 months, 1 year); (b) after (a), determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (c) selecting a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy or in conjunction with another anticancer agent for the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (d) selecting additional doses of the first ALK inhibitor of step (a) for the subject if the subject has a cancer cell that does not have has one or more ROS1 inhibitor resistance mutations.
  • the method can further include selecting doses of another anticancer agent for the subject.
  • the ROS1 inhibitor resistance mutation includes one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • a ROS1 inhibitor resistance mutation can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent can be another ROS1 inhibitor.
  • another anticancer agent is the first ALK inhibitor administered in step (a).
  • Also provided are methods of selecting a treatment for a subject having a cancer that include (a) administering one or more doses of a first TRK inhibitor to the subject for a period of time (e.g., 1 month, 2 months, 3 months, 6 months, 9 months, 1 year); (b) after (a), determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (c) selecting a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy or in conjunction with another anticancer agent for the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (d) selecting additional doses of the first TRK inhibitor of step (a) for the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • the method can further include selecting doses of another anticancer agent for the subject.
  • the ROS1 inhibitor resistance mutation includes one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • a ROS1 inhibitor resistance mutation can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent can be another ROS1 inhibitor.
  • another anticancer agent is the first TRK inhibitor administered in step (a).
  • Also provided are methods of selecting a treatment for a subject having a cancer that include (a) administering a first ROS1 inhibitor to the subject for a period of time (e.g., 1 month, 2 months, 3 months, 6 months, 9 months, 1 year); (b) after (a), determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and (c) selecting a second ROS1 inhibitor as a monotherapy or in conjunction with another anticancer agent if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (d) selecting additional doses of the first ROS1 inhibitor of step (a) for the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • the method can further include selecting doses of another anticancer agent for the subject.
  • the one or more ROS1 inhibitor resistance mutations confer increased resistance to a cancer cell or tumor to treatment with the first ROS1 inhibitor.
  • the one or more ROS1 inhibitor resistance mutations include one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • the one or more ROS1 inhibitor resistance mutations can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent is another ROS1 inhibitor (e.g., a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof).
  • another ROS1 can be the first ROS1 inhibitor administered in step (a).
  • Also provided are methods of selecting a treatment for a subject having a cancer that include (a) determining whether a cancer cell in a sample obtained from a subject having a cancer and previously administered a first ROS1 inhibitor has one or more ROS1 inhibitor resistance mutations; (b) selecting a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy or in conjunction with another anticancer agent for the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (c) selecting additional doses of the first ROS1 inhibitor previously administered to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • the method can further include selecting doses of another anticancer agent (e.g., a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof) for the subject.
  • another anticancer agent e.g., a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof
  • the one or more ROS1 inhibitor resistance mutations confer increased resistance to a cancer cell or tumor to treatment with the first ROS1 inhibitor.
  • the one or more ROS1 inhibitor resistance mutations include one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • the one or more ROS1 inhibitor resistance mutations can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent is another ROS1 inhibitor (e.g., a second ROS1 inhibitor).
  • another ROS1 inhibitor can be the first ROS1 inhibitor administered in step (a).
  • Also provided are methods of selecting a treatment for a subject having a cancer that include: (a) determining whether a cancer cell in a sample obtained from a subject having a cancer and previously administered a first ALK inhibitor has one or more ROS1 inhibitor resistance mutations; and (b) selecting a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy or in conjunction with another anticancer agent for the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (c) selecting additional doses of the first ALK inhibitor previously administered to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • a cancer e.g., a ROS1-associated cancer
  • the method can further include selecting doses of another anticancer agent (e.g., a second ALK inhibitor, a TRK inhibitor, a first ROS1 inhibitor, or a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof).
  • another anticancer agent e.g., a second ALK inhibitor, a TRK inhibitor, a first ROS1 inhibitor, or a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • the ROS1 inhibitor resistance mutation includes one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • a ROS1 inhibitor resistance mutation can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent is can be another ALK inhibitor (e.g., a second ALK inhibitor).
  • another anticancer agent can be the first ALK inhibitor administered in step (a).
  • another anticancer agent can be another ROS1 inhibitor.
  • Also provided are methods of selecting a treatment for a subject having a cancer that include: (a) determining whether a cancer cell in a sample obtained from a subject having a cancer and previously administered a first TRK inhibitor has one or more ROS1 inhibitor resistance mutations; and (b) selecting a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy or in conjunction with another anticancer agent for the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (c) selecting additional doses of the first TRK inhibitor previously administered to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • a cancer e.g., a ROS1-associated cancer
  • the method can further include selecting doses of another anticancer agent (e.g., a second TRK inhibitor, an ALK inhibitor, a first ROS1 inhibitor, or a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof).
  • another anticancer agent e.g., a second TRK inhibitor, an ALK inhibitor, a first ROS1 inhibitor, or a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof.
  • the ROS1 inhibitor resistance mutation includes one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • a ROS1 inhibitor resistance mutation can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent is can be another TRK inhibitor (e.g., a second TRK inhibitor).
  • another anticancer agent can be the first TRK inhibitor administered in step (a).
  • another anticancer agent can be another ROS1 inhibitor.
  • Also provided are methods of selecting a treatment for a subject having a cancer that include (a) determining whether a cancer cell in a sample obtained from a subject having a cancer and previously administered a first ROS1 inhibitor has one or more ROS1 inhibitor resistance mutations; (b) selecting a second ROS1 inhibitor as a monotherapy or in conjunction with another anticancer agent for the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations; or (c) selecting additional doses of the first ROS1 inhibitor previously administered to the subject if the subject has a cancer cell that does not have one or more ROS1 inhibitor resistance mutations.
  • the method can further include selecting doses of another anticancer agent (e.g., a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof) for the subject.
  • another anticancer agent e.g., a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof
  • the one or more ROS1 inhibitor resistance mutations confer increased resistance to a cancer cell or tumor to treatment with the first ROS1 inhibitor.
  • the one or more ROS1 inhibitor resistance mutations include one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • the one or more ROS1 inhibitor resistance mutations can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent is another ROS1 inhibitor (e.g., a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof).
  • another ROS1 can be the first ROS1 inhibitor administered in step (a).
  • Also provided are methods of determining a subject's risk for developing a cancer that has some resistance to a first ROS1 inhibitor that include: determining whether a cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and identifying a subject having a cell that has one or more ROS1 inhibitor resistance mutations as having an increased likelihood of developing a cancer that has some resistance to the first ROS1 inhibitor. Also provided are methods of determining a subject's risk for developing a cancer that has some resistance to a first ROS1 inhibitor that include: identifying a subject having a cell that has one or more ROS1 inhibitor resistance mutations as having an increased likelihood of developing a cancer that has some resistance to the first ROS1 inhibitor.
  • Also provided are methods of determining the presence of a cancer that has some resistance to a first ROS1 inhibitor that include: determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and determining that the subject having a cancer cell that has one or more ROS1 inhibitor resistance mutations has a cancer that has some resistance to the first ROS1 inhibitor. Also provided are methods of determining the presence of a cancer that has some resistance to a first ROS1 inhibitor in a subject that include: determining that a subject having a cancer cell that has one or more ROS1 inhibitor resistance mutations has a cancer that has some resistance to the first ROS1 inhibitor.
  • the one or more ROS1 inhibitor resistance mutations confer increased resistance to a cancer cell or tumor to treatment with the first ROS1 inhibitor.
  • the one or more ROS1 inhibitor resistance mutations include one or more ROS1 inhibitor resistance mutations listed in Table 4.
  • the one or more ROS1 inhibitor resistance mutations can include a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N.
  • a ROS1 inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with a first ROS1 inhibitor can be any of the ROS1 inhibitor resistance mutations listed in Table 4 (e.g., a substitution at one or more of amino acid positions 2026, 2032, or 2033, e.g., L2026M, G2032R, or D2033N).
  • Also provided are methods of determining the likelihood that a subject having a cancer will have a positive response to treatment with a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy that include: determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and determining that the subject having the cancer cell that has one or more ROS1 inhibitor resistance mutations has an increased likelihood of having a positive response to treatment with a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy.
  • Also provided are methods of determining the likelihood that a subject having cancer will have a positive response to treatment with a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy that include: determining that a subject having a cancer cell that has one or more ROS1 inhibitor resistance mutations has an increased likelihood of having a positive response to treatment with a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy.
  • Also provided are methods of predicting the efficacy of treatment with a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy in a subject having cancer that include: determining whether a cancer cell in a sample obtained from the subject has one or more ROS1 inhibitor resistance mutations; and determining that treatment with a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy is likely to be effective in a subject having a cancer cell in a sample obtained from the subject that has one or more ROS1 inhibitor resistance mutations.
  • Also provided are methods of predicting the efficacy of treatment with a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy in a subject having cancer that include: determining that treatment with a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof as a monotherapy is likely to be effective in a subject having a cancer cell in a sample obtained from the subject that has one or more ROS1 inhibitor resistance mutations.
  • Methods of determining the level of resistance of a cancer cell or a tumor to a ROS1 inhibitor can be determined using methods known in the art.
  • the level of resistance of a cancer cell to a ROS1 inhibitor can be assessed by determining the IC 50 of a ROS1 inhibitor (e.g., any of the ROS1 inhibitors described herein or known in the art) on the viability of a cancer cell.
  • the level of resistance of a cancer cell to a ROS1 inhibitor can be assessed by determining the growth rate of the cancer cell in the presence of a ROS1 inhibitor (e.g., any of the ROS1 inhibitors described herein).
  • the level of resistance of a tumor to a ROS1 inhibitor can be assessed by determining the mass or size of one or more tumors in a subject over time during treatment with a ROS1 inhibitor (e.g., any of the ROS1 inhibitors described herein).
  • the level of resistance of a cancer cell or a tumor to a ROS1 inhibitor can be indirectly assessed by determining the activity of a ROS1 kinase including one or more of the ROS1 inhibitor resistance mutations (i.e., the same ROS1 kinase expressed in a cancer cell or a tumor in a subject).
  • the level of resistance of a cancer cell or tumor having one or more ROS1 inhibitor resistance mutations to a ROS1 inhibitor is relative to the level of resistance in a cancer cell or tumor that does not have one or more ROS1 inhibitor resistance mutations (e.g., a cancer cell or tumor that does not have the same ROS1 inhibitor resistance mutations, a cancer cell or a tumor that does not have any ROS1 inhibitor resistance mutations, or a cancer cell or a tumor that expresses a wildtype ROS1 protein).
  • the determined level of resistance of a cancer cell or a tumor having one or more ROS1 inhibitor resistance mutations can be greater than about 1%, greater than about 2%, greater than about 3%,greater than about 4%, greater than about 5%, greater than about 6%, greater than about 7%, greater than about 8%, greater than about 9%, greater than about 10%, greater than about 11%, greater than about 12%, greater than about 13%, greater than about 14%, greater than about 15%, greater than about 20%, greater than about 25%, greater than about 30%, greater than about 35%, greater than about 40%, greater than about 45%, greater than about 50%, greater than about 60%, greater than about 70%, greater than about 80%, greater than about 90%, greater than about 100%, greater than about 110%, greater than about 120%, greater than about 130%, greater than about 140%, greater than about 150%, greater than about 160%, greater than about 170%, greater than about 180%, greater than about 190%, greater than about 200%, greater than about 210%, greater than about 220%, greater
  • Also provided is a method for inhibiting ROS1 kinase activity in a cell comprising contacting the cell with a compound of Formula I.
  • the contacting is in vitro.
  • the contacting is in vivo.
  • the contacting is in vivo, wherein the method comprises administering an effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof to a subject having a cell having ROS1 kinase activity.
  • the cell is a cancer cell.
  • the cancer cell is any cancer as described herein.
  • the cancer cell is a ROS1-associated cancer cell.
  • a method for inhibiting ROS1 kinase activity in a mammalian cell comprising contacting the cell with a compound of Formula I.
  • the contacting is in vitro.
  • the contacting is in vivo.
  • the contacting is in vivo, wherein the method comprises administering an effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof to a mammal having a cell having ROSlkinase activity.
  • the mammalian cell is a mammalian cancer cell.
  • the mammalian cancer cell is any cancer as described herein.
  • the mammalian cancer cell is a ROS1-associated cancer cell.
  • contacting refers to the bringing together of indicated moieties in an in vitro system or an in vivo system.
  • “contacting” a ROS1 kinase with a compound provided herein includes the administration of a compound provided herein to an individual or patient, such as a human, having a ROS1 kinase, as well as, for example, introducing a compound provided herein into a sample containing a cellular or purified preparation containing the ROS1 kinase.
  • Also provided herein is a method of inhibiting cell proliferation, in vitro or in vivo, the method comprising contacting a cell with an effective amount of a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition thereof as defined herein
  • the phrase “effective amount” means an amount of compound that, when administered to a patient in need of such treatment, is sufficient to (i) treat a ROS1 kinase-associated disease or disorder, (ii) attenuate, ameliorate, or eliminate one or more symptoms of the particular disease, condition, or disorder, or (iii) delay the onset of one or more symptoms of the particular disease, condition, or disorder described herein.
  • the amount of a compound of Formula I that will correspond to such an amount will vary depending upon factors such as the particular compound, disease condition and its severity, the identity (e.g., weight) of the patient in need of treatment, but can nevertheless be routinely determined by one skilled in the art.
  • the compounds of Formula I can be administered in the form of pharmaceutical compositions.
  • These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal), oral or parenteral.
  • Oral administration can include a dosage form formulated for once-daily or twice-daily (BID) administration.
  • Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal intramuscular or injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration.
  • Parenteral administration can be in the form of a single bolus dose, or may be, for example, by a continuous perfusion pump.
  • Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable
  • compositions which contain, as the active ingredient, a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof, in combination with one or more pharmaceutically acceptable carriers (excipients).
  • the composition is suitable for topical administration.
  • the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container.
  • the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
  • the composition is formulated for oral administration.
  • the composition is formulated as a tablet or capsule.
  • compositions comprising a compound of Formula I or a pharmaceutically acceptable salt or solvate thereof can be formulated in a unit dosage form, each dosage containing from about 5 to about 1,000 mg (1 g), more usually about 100 mg to about 500 mg, of the active ingredient.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human subjects and other patients, each unit containing a predetermined quantity of active material (i.e., a compound for Formula I as provided herein) calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • the compositions provided herein contain from about 5 mg to about 50 mg of the active ingredient.
  • the active ingredient contains from about 5 mg to about 50 mg.
  • One having ordinary skill in the art will appreciate that this embodies compounds or compositions containing about 5 mg to about 10 mg, about 10 mg to about 15 mg, about 15 mg to about 20 mg, about 20 mg to about 25 mg, about 25 mg to about 30 mg, about 30 mg to about 35 mg, about 35 mg to about 40 mg, about 40 mg to about 45 mg, or about 45 mg to about 50 mg of the active ingredient.
  • compositions provided herein contain from about 50 mg to about 500 mg of the active ingredient.
  • the active ingredient contains from about 50 mg to about 500 mg of the active ingredient.
  • One having ordinary skill in the art will appreciate that this embodies compounds or compositions containing about 50 mg to about 100 mg, about 100 mg to about 150 mg, about 150 mg to about 200 mg, about 200 mg to about 250 mg, about 250 mg to about 300 mg, about 350 mg to about 400 mg, or about 450 mg to about 500 mg of the active ingredient.
  • the compositions provided herein contain from about 500 mg to about 1,000 mg of the active ingredient.
  • the active ingredient contains from about 500 mg to about 1,000 mg of the active ingredient.
  • this embodies compounds or compositions containing about 500 mg to about 550 mg, about 550 mg to about 600 mg, about 600 mg to about 650 mg, about 650 mg to about 700 mg, about 700 mg to about 750 mg, about 750 mg to about 800 mg, about 800 mg to about 850 mg, about 850 mg to about 900 mg, about 900 mg to about 950 mg, or about 950 mg to about 1,000 mg of the active ingredient.
  • the active compound may be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
  • kits useful for example, in the treatment of RET-associated diseases or disorders, such as cancer or irritable bowel syndrome (IBS), which include one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a compound provided herein.
  • kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art.
  • Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
  • the potency of a compound inhibiting wild type and exemplary mutant ROS1 kinases was determined using CisBio's HTRF Kinease-TK assay technology.
  • the assays contained 5 nM wild type ROS1 (SignalChem—Cat. No. R14-11G), 5 nM G2032R ROS1 (SignalChem—Cat. No. R14-12BG), 5 nM L2026M ROS1 (Array Biopharma, p 1965), or 5 nM D2033N ROS1 (Array Biopharma, p 1994).
  • Each kinase is incubated with 250 nM TK-substrate biotin (CisBio, Cat. No.
  • 62TKOPEC and 1 mM ATP along with test compound in a buffer consisting of 25 mM MOPS [pH 7.4], 5 mM MgCl 2 , 0.005% Triton X-100, and 2% DMSO in a volume of 8 ⁇ L.
  • Compounds were prepared in a four-fold serial dilution in DMSO and added to the assay to give the appropriate final concentration. After a 120-minute incubation at 22° C., the reaction was quenched by adding 8 ⁇ L of quench solution containing 31.3 nM Sa-XL665 and 1 ⁇ TK-Ab-Cryptate in HTRF detection buffer (CisBio, Cat. No. 62TKOPEC).
  • Table 9 provides averaged IC 50 values for compounds tested in this assay.
US16/199,867 2017-03-16 2018-11-26 Macrocyclic compounds as ros1 kinase inhibitors Abandoned US20190076436A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/199,867 US20190076436A1 (en) 2017-03-16 2018-11-26 Macrocyclic compounds as ros1 kinase inhibitors

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201762472185P 2017-03-16 2017-03-16
PCT/US2018/022833 WO2018170381A1 (en) 2017-03-16 2018-03-16 Macrocyclic compounds as ros1 kinase inhibitors
US16/199,867 US20190076436A1 (en) 2017-03-16 2018-11-26 Macrocyclic compounds as ros1 kinase inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/022833 Continuation WO2018170381A1 (en) 2017-03-16 2018-03-16 Macrocyclic compounds as ros1 kinase inhibitors

Publications (1)

Publication Number Publication Date
US20190076436A1 true US20190076436A1 (en) 2019-03-14

Family

ID=61913541

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/199,867 Abandoned US20190076436A1 (en) 2017-03-16 2018-11-26 Macrocyclic compounds as ros1 kinase inhibitors
US16/199,818 Active 2038-04-12 US10966985B2 (en) 2017-03-16 2018-11-26 Macrocyclic compounds as ROS1 kinase inhibitors
US16/199,875 Active US10688100B2 (en) 2017-03-16 2018-11-26 Macrocylic compounds as ROS1 kinase inhibitors

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/199,818 Active 2038-04-12 US10966985B2 (en) 2017-03-16 2018-11-26 Macrocyclic compounds as ROS1 kinase inhibitors
US16/199,875 Active US10688100B2 (en) 2017-03-16 2018-11-26 Macrocylic compounds as ROS1 kinase inhibitors

Country Status (19)

Country Link
US (3) US20190076436A1 (ar)
EP (1) EP3595651A1 (ar)
JP (1) JP2020514356A (ar)
KR (1) KR20190126003A (ar)
CN (1) CN110769820A (ar)
AU (1) AU2018234726A1 (ar)
BR (1) BR112019019101A2 (ar)
CA (1) CA3056754A1 (ar)
CL (1) CL2019002612A1 (ar)
IL (1) IL269216A (ar)
JO (1) JOP20190213A1 (ar)
MA (1) MA49888A (ar)
MX (1) MX2019010988A (ar)
PH (1) PH12019502069A1 (ar)
RU (1) RU2019132666A (ar)
SG (1) SG11201908532UA (ar)
TN (1) TN2019000263A1 (ar)
TW (1) TW201838631A (ar)
WO (1) WO2018170381A1 (ar)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10590139B2 (en) 2008-09-22 2020-03-17 Array Biopharma Inc. Method of treatment using substituted imidazo[1,2b]pyridazine compounds
US10588908B2 (en) 2016-04-04 2020-03-17 Loxo Oncology, Inc. Methods of treating pediatric cancers
US10647730B2 (en) 2010-05-20 2020-05-12 Array Biopharma Inc. Macrocyclic compounds as TRK kinase inhibitors
US10655186B2 (en) 2015-10-26 2020-05-19 Loxo Oncology, Inc. Point mutations in TRK inhibitor-resistant cancer and methods relating to the same
US10668072B2 (en) 2016-04-04 2020-06-02 Loxo Oncology, Inc. Liquid formulations of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
US10688100B2 (en) 2017-03-16 2020-06-23 Array Biopharma Inc. Macrocylic compounds as ROS1 kinase inhibitors
US10758542B2 (en) 2009-07-09 2020-09-01 Array Biopharma Inc. Substituted pyrazolo[l,5-a]pyrimidine compounds as Trk kinase inhibitors
US10774085B2 (en) 2008-10-22 2020-09-15 Array Biopharma Inc. Method of treatment using substituted pyrazolo[1,5-A] pyrimidine compounds
WO2020198379A1 (en) * 2019-03-26 2020-10-01 Ventyx Biosciences, Inc. Tyk2 pseudokinase ligands
US10799505B2 (en) 2014-11-16 2020-10-13 Array Biopharma, Inc. Crystalline form of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-A]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate
CN113004305A (zh) * 2019-12-19 2021-06-22 成都倍特药业股份有限公司 大环化合物及其制备方法和用途
US11091486B2 (en) 2016-10-26 2021-08-17 Array Biopharma, Inc Process for the preparation of pyrazolo[1,5-a]pyrimidines and salts thereof
US11214571B2 (en) 2016-05-18 2022-01-04 Array Biopharma Inc. Process for the preparation of (S)-N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide and salts thereof
WO2022182845A1 (en) * 2021-02-25 2022-09-01 Blossomhill Therapeutics, Inc. Macrocycles and their use
US11753411B2 (en) 2019-11-08 2023-09-12 Ventyx Biosciences, Inc. Substituted pyrazolo[1,5-a]pyrimidines as TYK2 pseudokinase ligands

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW201927790A (zh) * 2017-11-10 2019-07-16 美商安捷克斯製藥公司 作為trk激酶抑制劑之巨環化合物及其用途
CN113329749A (zh) * 2018-10-05 2021-08-31 伊利诺伊大学董事会 用于治疗葡萄膜黑色素瘤的联合疗法
EP3870579A4 (en) 2018-10-22 2022-10-19 Alumis Inc. TYK2 INHIBITORS AND THEIR USES
CN113811534A (zh) * 2019-03-11 2021-12-17 埃斯克疗法股份有限公司 Tyk2抑制剂和其用途
US20220143049A1 (en) 2019-03-21 2022-05-12 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
EP4045645A1 (en) * 2019-10-16 2022-08-24 Health Research, Inc. Combination therapy for treatment of cancers
JP2023500906A (ja) 2019-11-08 2023-01-11 インサーム(インスティテュ ナシオナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシェ メディカル) キナーゼ阻害剤に対する獲得抵抗性を有するがんの処置方法
JP7374532B2 (ja) * 2019-11-18 2023-11-07 クワンチョウ ジョーヨー ファーマテック カンパニー,リミティド 選択性の高いros1阻害剤としての化合物、及びその使用
CN112812128B (zh) * 2019-11-18 2024-04-02 正大天晴药业集团股份有限公司 作为alk和ros调节剂的大环化合物
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
KR20230037564A (ko) * 2020-07-10 2023-03-16 블로썸힐 테라퓨틱스, 인크. 거대고리 화합물 및 그의 용도
CN113072564A (zh) * 2021-03-23 2021-07-06 南京奥利墨斯医药科技有限公司 一种杂芳环类化合物及其用途

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6463922B2 (en) * 2000-01-24 2002-10-15 Graniterie Des Ecorces Wedge intended to be inserted in a cutting slot
US8933084B2 (en) * 2010-05-20 2015-01-13 Array Biopharma Inc. Macrocyclic compounds as Trk kinase inhibitors
US20160032404A1 (en) * 2014-08-01 2016-02-04 Pharmacyclics Llc Biomarkers for predicting response of dlbcl to treatment with a btk inhibitor
US20160367547A1 (en) * 2015-06-19 2016-12-22 Macau University Of Science And Technology Oncogenic ros1 and alk kinase inhibitor

Family Cites Families (386)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1017286A (en) 1909-03-01 1912-02-13 Du Pont Powder Co Apparatus for treating explosive powder.
US1013712A (en) 1911-06-03 1912-01-02 Alfred D Williams Tie and rail-fastener.
EP0009517A1 (en) 1978-10-04 1980-04-16 THE PROCTER & GAMBLE COMPANY Vaginal contraceptive
NZ234143A (en) 1989-06-28 1991-10-25 Mcneil Ppc Inc Aqueous pharmaceutical suspension formulation for administering substantially insoluble pharmaceutical agents
CA2276945C (en) 1993-11-30 2006-08-01 G.D. Searle & Co. Tricyclic-substituted pyrazolyl benzenesulfonamides and pharmaceutical compositions thereof
US5844092A (en) 1994-03-18 1998-12-01 Genentech, Inc. Human TRK receptors and neurotrophic factor inhibitors
US5430021A (en) 1994-03-18 1995-07-04 Pharmavene, Inc. Hydrophobic drug delivery systems
US5877016A (en) 1994-03-18 1999-03-02 Genentech, Inc. Human trk receptors and neurotrophic factor inhibitors
EP0914339B1 (en) 1996-05-08 2006-08-02 Biogen Idec MA Inc. RET LIGAND 3 (RetL3) FOR STIMULATING NEURAL AND RENAL GROWTH
US6677135B1 (en) 1996-05-08 2004-01-13 Biogen, Inc. Ret ligand (RetL) for stimulating neutral and renal growth
CA2206201A1 (en) 1996-05-29 1997-11-29 Yoshiaki Isobe Pyrazole derivatives and their pharmaceutical use
US6682921B1 (en) 1996-08-21 2004-01-27 New York University Crystals of the tyrosine kinase domain of non-insulin receptor tyrosine kinases
JP3898296B2 (ja) 1996-08-28 2007-03-28 ポーラ化成工業株式会社 ピロロピラゾロピリミジン化合物及びこれを有効成分とする医薬
CA2285264A1 (en) 1997-04-25 1998-11-05 Michiyo Gyoten Condensed pyridazine derivatives, their production and use
US6531152B1 (en) 1998-09-30 2003-03-11 Dexcel Pharma Technologies Ltd. Immediate release gastrointestinal drug delivery system
UA74546C2 (en) 1999-04-06 2006-01-16 Boehringer Ingelheim Ca Ltd Macrocyclic peptides having activity relative to hepatitis c virus, a pharmaceutical composition and use of the pharmaceutical composition
AU7103900A (en) 1999-09-01 2001-03-26 Biogen, Inc. Ret ligand 5 (retl5) compositions and uses thereof
US6534085B1 (en) 1999-09-23 2003-03-18 Bioresponse L.L.C. Phytochemicals for promoting weight loss
FI20000403A0 (fi) 2000-02-22 2000-02-22 Hannu Sariola GDNF perhesukuisten yhdisteiden käyttö kivessyövän hoitoon tarkoitettujen tuotteiden valmistamiseksi
AU7142201A (en) 2000-06-22 2002-01-02 Genentech Inc Agonist anti-trk-c monoclonal antibodies
TWI312347B (en) 2001-02-08 2009-07-21 Eisai R&D Man Co Ltd Bicyclic nitrogen-containing condensed ring compounds
CA2448956C (en) 2001-05-30 2017-10-03 Genentech, Inc. Anti-ngf antibodies for the treatment of various disorders
AU2002334355A1 (en) 2001-09-06 2003-03-18 Prochon Biotech Ltd. Protein tyrosine kinase inhibitors
US7101572B2 (en) 2001-12-07 2006-09-05 Unilab Pharmatech, Ltd. Taste masked aqueous liquid pharmaceutical composition
US20030199525A1 (en) 2002-03-21 2003-10-23 Hirst Gavin C. Kinase inhibitors
CA2483306A1 (en) 2002-04-23 2003-11-06 Shionogi & Co., Ltd. Pyrazolo[1,5-a]pyrimidine derivative and nad(p)h oxidase inhibitor containing the same
US7449488B2 (en) 2002-06-04 2008-11-11 Schering Corporation Pyrazolopyrimidines as protein kinase inhibitors
ITMI20021620A1 (it) 2002-07-23 2004-01-23 Novuspharma Spa Composto ad ativita' antitumorale
CA2493000A1 (en) 2002-07-24 2004-01-29 University Of Cincinnati 4-4(methylpiperazin-1-ylmethyl)-n-[4-methyl-3-(pyridin-3-yl)pyrimidin-2-ylamino)phenyl]-benzamide for treating mutated-ret kinase associated diseases
JP4024624B2 (ja) 2002-08-26 2007-12-19 富士通株式会社 半導体装置の製造方法及び製造装置
KR101088922B1 (ko) 2002-09-04 2011-12-01 파마코페이아, 엘엘씨. 사이클린 의존성 키나제 억제제로서의 피라졸로피리미딘
US7196078B2 (en) 2002-09-04 2007-03-27 Schering Corpoartion Trisubstituted and tetrasubstituted pyrazolopyrimidines as cyclin dependent kinase inhibitors
US8580782B2 (en) 2002-09-04 2013-11-12 Merck Sharp & Dohme Corp. Substituted pyrazolo[1,5-a]pyrimidines as cyclin dependent kinase inhibitors
US7119200B2 (en) 2002-09-04 2006-10-10 Schering Corporation Pyrazolopyrimidines as cyclin dependent kinase inhibitors
AU2003298942A1 (en) 2002-12-11 2004-06-30 Merck And Co., Inc. Tyrosine kinase inhibitors
WO2004052315A2 (en) 2002-12-11 2004-06-24 Merck & Co., Inc. Tyrosine kinase inhibitors
ES2325440T3 (es) 2003-02-20 2009-09-04 Smithkline Beecham Corporation Compuestos de pirimidina.
GB0303910D0 (en) 2003-02-20 2003-03-26 Merck Sharp & Dohme Therapeutic agents
WO2004082458A2 (en) 2003-02-21 2004-09-30 The Johns Hopkins University Tyrosine kinome
JP2004277337A (ja) 2003-03-14 2004-10-07 Sumitomo Pharmaceut Co Ltd ピラゾロ[1,5−a]ピリミジン誘導体
EP1608652A1 (en) 2003-03-31 2005-12-28 Vernalis (Cambridge) Limited Pyrazolopyrimidine compounds and their use in medicine
WO2004089415A2 (en) 2003-04-11 2004-10-21 Novo Nordisk A/S COMBINATIONS OF AN 11β-HYDROXYSTEROID DEHYDROGENASE TYPE 1 INHIBITOR AND A GLUCOCORTICOID RECEPTOR AGONIST
US20060094699A1 (en) 2003-04-11 2006-05-04 Kampen Gita Camilla T Combination therapy using an 11beta-hydroxysteroid dehydrogenase type 1 inhibitor and a glucocorticoid receptor agonist to minimize the side effects associated with glucocorticoid receptor agonist therapy
EP1615697A2 (en) 2003-04-11 2006-01-18 Novo Nordisk A/S New pyrazolo[1,5-a] pyrimidine derivatives and pharmaceutical use thereof
CN1795206A (zh) 2003-04-28 2006-06-28 株式会社嘉尔药物 半乳糖凝集素-9诱导因子
PA8603801A1 (es) 2003-05-27 2004-12-16 Janssen Pharmaceutica Nv Derivados de la quinazolina
JP2005008581A (ja) 2003-06-20 2005-01-13 Kissei Pharmaceut Co Ltd 新規なピラゾロ[1,5−a]ピリミジン誘導体、それを含有する医薬組成物およびそれらの用途
EA009517B1 (ru) 2003-06-27 2008-02-28 Байер Кропсайенс Аг Пиразолопиримидины
NZ599196A (en) 2003-07-15 2014-01-31 Amgen Inc Human anti-ngf neutralizing antibodies as selective ngf pathway inhibitors
US7491794B2 (en) 2003-10-14 2009-02-17 Intermune, Inc. Macrocyclic compounds as inhibitors of viral replication
US20090143399A1 (en) 2003-10-14 2009-06-04 Arizona Board Of Regents On Behalf Of The University Of Arizona Protein Kinase Inhibitors
US7169918B2 (en) 2003-10-27 2007-01-30 Genelabs Technologies, Inc. Methods for preparing 7-(2′-substituted-β-D-ribofuranosyl)-4-(NR2R3)-5-(substituted ethyn-1-yl)-pyrrolo[2,3-d]pyrimidine derivatives
MY141220A (en) 2003-11-17 2010-03-31 Astrazeneca Ab Pyrazole derivatives as inhibitors of receptor tyrosine kinases
AU2004295061B2 (en) 2003-11-21 2008-11-20 Novartis Ag 1H-imidazoquinoline derivatives as protein kinase inhibitors
KR20060110307A (ko) 2003-11-28 2006-10-24 노파르티스 아게 단백질 키나아제 의존성 질환의 치료에서의 디아릴 우레아유도체
MXPA06007017A (es) 2003-12-18 2006-08-31 Janssen Pharmaceutica Nv Derivados de pirido y pirimidopirimidina como agentes antiproliferativos.
CA2550361C (en) 2003-12-19 2014-04-29 Prabha Ibrahim Compounds and methods for development of ret modulators
GB0330043D0 (en) 2003-12-24 2004-01-28 Pharmacia Italia Spa Pyrrolo [2,3-b] pyridine derivatives active as kinase inhibitors process for their preparation and pharmaceutical compositions comprising them
GB0330042D0 (en) 2003-12-24 2004-01-28 Pharmacia Italia Spa Pyrrolo [2,3-b] pyridine derivatives active as kinase inhibitors process for their preparation and pharmaceutical compositions them
WO2005068424A1 (en) 2004-01-20 2005-07-28 Cell Therapeutics Europe S.R.L. Indolinone derivatives as receptor tyrosine kinase ihibitors
PE20051089A1 (es) 2004-01-22 2006-01-25 Novartis Ag Derivados de pirazolo [1,5-a] pirimidin-7-il-amina como inhibidores de quinasa de proteina
US20050222171A1 (en) 2004-01-22 2005-10-06 Guido Bold Organic compounds
WO2005099363A2 (en) 2004-03-26 2005-10-27 Whitehead Institute For Biomedical Research Methods of diagnosing, preventing and treating cancer metastasis
UA91677C2 (ru) 2004-03-30 2010-08-25 Интермюн, Инк. Макроциклические соединения как ингибиторы вирусной репликации
GB0512324D0 (en) 2005-06-16 2005-07-27 Novartis Ag Organic compounds
BRPI0514094A (pt) 2004-08-02 2008-05-27 Osi Pharm Inc composto, composição, e, método de tratamento de distúrbio hiperproliferativo
PE20060664A1 (es) 2004-09-15 2006-08-04 Novartis Ag Amidas biciclicas como inhibidores de cinasa
WO2006050076A1 (en) 2004-10-29 2006-05-11 Janssen Pharmaceutica, N.V. Pyrimidinyl substituted fused-pyrrolyl compounds useful in treating kinase disorders
PT1812440E (pt) 2004-11-04 2011-01-25 Vertex Pharma Pirazolo[1,5-a]pirimidinas úteis enquanto inibidores de proteínas cinases
JO3088B1 (ar) 2004-12-08 2017-03-15 Janssen Pharmaceutica Nv مشتقات كوينازولين كبيرة الحلقات و استعمالها بصفتها موانع كينيز متعددة الاهداف
DE102005003687A1 (de) 2005-01-26 2006-07-27 Sphingo Tec Gmbh Immunoassay zur Bestimmung der Freisetzung von Neurotensin in die Zirkulation
GB0501999D0 (en) 2005-02-01 2005-03-09 Sentinel Oncology Ltd Pharmaceutical compounds
AU2006215394B2 (en) 2005-02-16 2009-10-08 Astrazeneca Ab Chemical compounds
CN101119996A (zh) 2005-02-16 2008-02-06 阿斯利康(瑞典)有限公司 化学化合物
CN101257948A (zh) 2005-02-18 2008-09-03 阿特努奥恩公司 嘧啶并二氮䓬衍生物及吲哚并蝶啶化合物
ATE424402T1 (de) 2005-03-21 2009-03-15 Lilly Co Eli Imidazopyridinverbindungen
GB0507575D0 (en) 2005-04-14 2005-05-18 Novartis Ag Organic compounds
WO2006113509A2 (en) 2005-04-15 2006-10-26 Cylene Pharmaceuticals, Inc. Quinobenzoxazine analogs and methods of using thereof
WO2006115452A1 (en) 2005-04-27 2006-11-02 Astrazeneca Ab Use of pyrazolyl-pyrimidine derivatives in the treatment of pain
AU2006248780B2 (en) 2005-05-16 2010-06-03 Astrazeneca Ab Pyrazolylaminopyrimidine derivatives useful as tyrosine kinase inhibitors
WO2006128042A2 (en) 2005-05-26 2006-11-30 The Johns Hopkins University Methods of identifying mutations in nucleic acid
WO2006130613A2 (en) 2005-05-31 2006-12-07 The Pfahl Family Trust (Dated 9 July 1996) Substituted biarylheterocycle derivatives as protein kinase inhibitors for the treatment of cancer and other diseases
US7541367B2 (en) 2005-05-31 2009-06-02 Janssen Pharmaceutica, N.V. 3-benzoimidazolyl-pyrazolopyridines useful in treating kinase disorders
CN100406650C (zh) 2005-06-05 2008-07-30 徐斌 一种抗特大变位的模块式梳型桥梁伸缩缝装置
ITRM20050290A1 (it) 2005-06-07 2006-12-08 Lay Line Genomics Spa Uso di molecole in grado di inibire il legame tra ngf e il suo recettore trka come analgesici ad effetto prolungato.
BRPI0611863B1 (pt) 2005-06-22 2021-11-23 Plexxikon, Inc Composto, bem como composição e kit compreendendo o mesmo, composto intermediário na preparação do mesmo, método para tratamento e uso do mesmo
US20070025540A1 (en) 2005-07-07 2007-02-01 Roger Travis Call center routing based on talkativeness
GB0515026D0 (en) 2005-07-21 2005-08-31 Novartis Ag Organic compounds
JP5249028B2 (ja) 2005-07-25 2013-07-31 インターミューン・インコーポレーテッド C型肝炎ウイルス複製の新規大環状阻害剤
US20100216798A1 (en) 2005-07-29 2010-08-26 Astellas Pharma Inc Fused heterocycles as lck inhibitors
WO2007019058A1 (en) 2005-08-03 2007-02-15 Eastman Chemical Company Tocopheryl polyethylene glycol succinate powder and process for preparing same
EP1926734A1 (en) 2005-08-22 2008-06-04 Amgen Inc. Pyrazolopyridine and pyrazolopyrimidine compounds useful as kinase enzymes modulators
WO2007025090A2 (en) 2005-08-25 2007-03-01 Kalypsys, Inc. Heterobicyclic and - tricyclic inhibitors of mapk/erk kinase
MX2008002524A (es) 2005-08-25 2008-03-14 Creabilis Therapeutics Spa Conjugados polimericos de k-252a y sus derivados.
DE102005042742A1 (de) 2005-09-02 2007-03-08 Schering Ag Substituierte Imidazo[1,2b]pyridazine als Kinase-Inhibitoren, deren Herstellung und Verwendung als Arzneimittel
US20070078136A1 (en) 2005-09-22 2007-04-05 Bristol-Myers Squibb Company Fused heterocyclic compounds useful as kinase modulators
US7645762B2 (en) 2005-10-06 2010-01-12 Schering Corporation Substituted pyrazolo[1,5-a] pyrimidines as protein kinase inhibitors
WO2007044441A2 (en) 2005-10-06 2007-04-19 Schering Corporation Use of pyrazolo [1 , 5 -a] pyrimidine derivatives for inhibiting protein kinases methods for inhibiting protein kinases
EP1948633B1 (en) 2005-10-11 2011-08-10 Centre National De La Recherche Scientifique (Cnrs) 3 -hydroxyflavone derivatives for the detection and the quantification of cell apoptosis
DE602006019323D1 (de) 2005-10-11 2011-02-10 Intermune Inc Verbindungen und verfahren zur inhibierung der replikation des hepatitis-c-virus
CA2626789A1 (en) 2005-10-21 2007-04-26 Exelixis, Inc. Pyrimidinones as casein kinase ii (ck2) modulators
WO2007053776A1 (en) 2005-11-03 2007-05-10 Sgx Pharmaceuticals, Inc. Pyrimidinyl-thiophene kinase modulators
EP1785420A1 (en) 2005-11-14 2007-05-16 4Sc Ag Thiazole analogues and uses thereof
US20070149523A1 (en) 2005-11-14 2007-06-28 Jan Ehlert Thiazole Analogues and Uses Thereof
WO2007057399A2 (en) 2005-11-15 2007-05-24 Boehringer Ingelheim International Gmbh Treatment of cancer with indole derivatives
WO2007057397A1 (en) 2005-11-15 2007-05-24 Boehringer Ingelheim International Gmbh Treatment of cancer
GB0524436D0 (en) 2005-11-30 2006-01-11 Novartis Ag Organic compounds
US7795273B2 (en) 2005-12-08 2010-09-14 Novartis Ag Pyrazolo[1,5-a]pyridine-3-carboxylic acids as EphB and VEGFR2 kinase inhibitors
EP1968579A1 (en) 2005-12-30 2008-09-17 Astex Therapeutics Limited Pharmaceutical compounds
US20080108611A1 (en) 2006-01-19 2008-05-08 Battista Kathleen A Substituted thienopyrimidine kinase inhibitors
EP1978958A4 (en) 2006-01-24 2009-12-02 Merck & Co Inc INHIBITION OF TYROSINE KINASE RET
PT1973910E (pt) 2006-01-27 2013-09-06 Shanghai Hengrui Pharm Co Ltd Inibidores de pirrolo [3,2-c] piridina-4-ona 2-indolinona proteína cinase
KR100846988B1 (ko) 2006-03-06 2008-07-16 제일약품주식회사 신규한 티에노피리미딘 유도체 또는 이의 약학적으로허용가능한 염, 이의 제조방법 및 이를 함유하는 약학조성물
EP2001880A2 (en) 2006-03-07 2008-12-17 Array Biopharma, Inc. Heterobicyclic pyrazole compounds and methods of use
BRPI0710181A2 (pt) 2006-03-16 2011-08-09 Novartis Ag compostos orgánicos
NZ571566A (en) 2006-03-17 2011-07-29 Ambit Biosciences Corp Imidazolothiazole compounds for the treatment of disease
MX2008012422A (es) 2006-03-27 2008-10-09 Nerviano Medical Sciences Srl Derivados de furano y tiofeno, pirrol substituido con pirimidinil y piridil como inhibidores de cinasa.
GB0606805D0 (en) 2006-04-04 2006-05-17 Novartis Ag Organic compounds
CL2007001165A1 (es) 2006-04-26 2008-01-25 Hoffmann La Roche 2-(1h-indazol-4-il)-6-(4-metanosulfonil-piperazin-1-ilmetil)-4-morfolin-4-il-tieno[3,2-d]pirimidina; procedimiento de preparacion; composicion farmaceutica; proceso de preparacion de dicha composicion; kit farmaceutico; y uso para tratar enfermedades tales como cancer, desordenes inmunes y enfermedades cardiovasculares.
AU2007254491A1 (en) 2006-05-15 2007-11-29 Irm Llc Compositions and methods for FGF receptor kinases inhibitors
EP2036557B1 (en) 2006-05-18 2015-10-21 Eisai R&D Management Co., Ltd. Antitumor agent for thyroid cancer
EP1873157A1 (en) 2006-06-21 2008-01-02 Bayer Schering Pharma Aktiengesellschaft Pyrazolopyrimidines and salts thereof, pharmaceutical compositions comprising same, methods of preparing same and uses of same
TWI419889B (zh) 2006-07-05 2013-12-21 Mitsubishi Tanabe Pharma Corp 吡唑并〔1,5-a〕嘧啶化合物
US20100029619A1 (en) 2006-08-04 2010-02-04 Takeda Pharmaceutical Company Limted Fused heterocyclic compound
US7531539B2 (en) 2006-08-09 2009-05-12 Bristol-Myers Squibb Company Pyrrolotriazine kinase inhibitors
US8063225B2 (en) 2006-08-14 2011-11-22 Chembridge Corporation Tricyclic compound derivatives useful in the treatment of neoplastic diseases, inflammatory disorders and immunomodulatory disorders
US20110021513A1 (en) 2006-09-07 2011-01-27 Biogen Idec Ma Inc. Modulators of interleukin-1 receptor-associated kinase
US20110195072A1 (en) 2006-09-12 2011-08-11 Anne Boulay Non-neuroendocrine cancer therapy
CN101553482B (zh) 2006-09-15 2013-11-20 艾科睿制药公司 激酶抑制剂化合物
ATE502943T1 (de) 2006-09-29 2011-04-15 Novartis Ag Pyrazolopyrimidine als pi3k-lipidkinasehemmer
US20120225057A1 (en) 2006-10-11 2012-09-06 Deciphera Pharmaceuticals, Llc Methods and compositions for the treatment of myeloproliferative diseases and other proliferative diseases
EP1918291A1 (en) 2006-10-30 2008-05-07 Novartis AG 3-Aminocarbonyl-substituted fused pyrazolo-derivatives as protein kinase modulators
CN101522682A (zh) 2006-10-30 2009-09-02 诺瓦提斯公司 作为抗炎剂的杂环化合物
KR101546493B1 (ko) 2006-11-06 2015-08-21 톨레로 파마수티컬스, 인크. 이미다조[1,2-b]피리다진 및 피라졸로[1,5-a]피리미딘 유도체 및 단백질 키나제 억제제로서의 이의 용도
DK2848610T3 (da) 2006-11-15 2017-11-06 Ym Biosciences Australia Pty Hæmmere af kinaseaktivitet
WO2008079909A1 (en) 2006-12-21 2008-07-03 Plexxikon, Inc. Pyrrolo [2,3-b] pyridines as kinase modulators
PE20121126A1 (es) 2006-12-21 2012-08-24 Plexxikon Inc Compuestos pirrolo [2,3-b] piridinas como moduladores de quinasa
RU2009122670A (ru) 2006-12-21 2011-01-27 Плекссикон, Инк. (Us) Соединения и способы для модуляции киназ и показания к их применению
US20080199426A1 (en) 2007-01-11 2008-08-21 Sukhatme Vikas P Methods and compositions for the treatment and diagnosis of vascular inflammatory disorders or endothelial cell disorders
JP2010516692A (ja) 2007-01-19 2010-05-20 バイエル・ヘルスケア・エルエルシー 化学療法剤に対し抵抗性を有する癌の処置
US20080234267A1 (en) 2007-03-20 2008-09-25 Karen Elizabeth Lackey Compounds and Methods of Treatment
US20080234262A1 (en) 2007-03-21 2008-09-25 Wyeth Pyrazolopyrimidine analogs and their use as mtor kinase and pi3 kinase inhibitors
WO2008116898A1 (en) 2007-03-28 2008-10-02 Biovitrum Ab (Publ) Pyrazolo [1,5-a]pyrimidines as inhibitors of stearoyl-coa desaturase
PT2137184E (pt) 2007-04-03 2013-08-01 Array Biopharma Inc Compostos imidazo[1,2-a]piridina como inibidores do receptor de tirosina quinase
US20110189167A1 (en) 2007-04-20 2011-08-04 Flynn Daniel L Methods and Compositions for the Treatment of Myeloproliferative Diseases and other Proliferative Diseases
WO2008137794A1 (en) 2007-05-04 2008-11-13 Irm Llc Compounds and compositions as c-kit and pdgfr kinase inhibitors
WO2008138184A1 (fr) 2007-05-14 2008-11-20 Shanghai Hengrui Pharmaceutical Co.Ltd. Dérivés de pyrrolo-azacycles, leur procédé de fabrication et leur utilisation en tant qu'inhibiteurs de protéine kinases
DK2170827T3 (da) 2007-06-21 2013-11-18 Janssen Pharmaceutica Nv Indolin-2-oner og aza-indolin-2-oner
WO2009003136A1 (en) 2007-06-26 2008-12-31 Rigel Pharmaceuticals, Inc. Substituted pyrimidine-2, 4 -diamines for treating cell proliferative disorders
CN101801962A (zh) 2007-07-09 2010-08-11 阿斯利康(瑞典)有限公司 用于治疗增殖疾病的三取代嘧啶衍生物
WO2009012262A1 (en) 2007-07-16 2009-01-22 The Regents Of The University Of California Protein kinase modulating compounds and methods for making and using them
EP2170830B1 (en) 2007-07-17 2014-10-15 Plexxikon, Inc. 2-FLUORO-BENZENESULFONAMIDE COMPOUNDS AS Raf KINASE MODULATORS
KR101552742B1 (ko) 2007-07-19 2015-09-11 머크 샤프 앤드 돔 코포레이션 단백질 키나제 억제제로서의 헤테로사이클릭 아미드 화합물
SI2176231T1 (sl) 2007-07-20 2017-01-31 Nerviano Medical Sciences S.R.L. Substituirani indazolni derivati, aktivni kot kinazni inhibitorji
WO2009017838A2 (en) 2007-08-01 2009-02-05 Exelixis, Inc. Combinations of jak-2 inhibitors and other agents
ES2547229T3 (es) 2007-08-07 2015-10-02 Purdue Research Foundation Inhibidores de cinasa y usos de los mismos
PT2187964E (pt) 2007-08-10 2015-01-14 Regeneron Pharma Anticorpos humanos de elevada afinidade para o factor de crescimento neural humano
EP2025678A1 (en) 2007-08-17 2009-02-18 Oncalis AG Pyrazolo[3,4-d]pyrimidine compounds and their use as modulators of protein kinase
WO2009042646A1 (en) 2007-09-24 2009-04-02 Curis, Inc. Anti-proliferative agents
EP2212333A1 (en) 2007-10-16 2010-08-04 Wyeth LLC Thienopyrimidine and pyrazolopyrimidine compounds and their use as mtor kinase and pi3 kinase inhibitors
JP2011501760A (ja) 2007-10-23 2011-01-13 ノバルティス アーゲー 呼吸器疾患の処置のためのtrkb抗体の使用
US20110046127A1 (en) 2007-11-08 2011-02-24 Paolo Pevarello Imidazopyridazines for Use as Protein Kinase Inhibitors
CA2706946A1 (en) 2007-11-28 2009-06-04 Schering Corporation 2-fluoropyrazolo[1,5-a]pyrimidines as protein kinase inhibitors
US8598172B2 (en) 2007-12-04 2013-12-03 Nerviano Medical Sciences S.R.L. Substituted dihydropteridin-6-one derivatives, process for their preparation and their use as kinase inhibitors
BRPI0906877A2 (pt) 2008-01-17 2015-07-28 Irm Llc Anticorpos anti-trkb aperfeiçoados
JP2009203226A (ja) 2008-01-31 2009-09-10 Eisai R & D Management Co Ltd ピリジン誘導体およびピリミジン誘導体を含有するレセプターチロシンキナーゼ阻害剤
TW200942537A (en) 2008-02-01 2009-10-16 Irm Llc Compounds and compositions as kinase inhibitors
WO2009103076A1 (en) 2008-02-15 2009-08-20 Oxigene, Inc. Methods and compositions for enhancing the efficacy of rtk inhibitors
MX2010010272A (es) 2008-03-19 2011-05-25 Chembridge Corp Nuevos inhibidores de tirosina quinasa.
US8822500B2 (en) 2008-03-19 2014-09-02 Chembridge Corporation Tyrosine kinase inhibitors
JP5767101B2 (ja) 2008-03-28 2015-08-19 ネルビアーノ・メデイカル・サイエンシーズ・エツセ・エルレ・エルレ キナーゼ阻害剤として活性な3,4−ジヒドロ−2h−ピラジノ[1,2−a]インドール−1−オン誘導体、これらの調製方法、およびこれらを含む医薬組成物
US8507488B2 (en) 2008-05-13 2013-08-13 Irm Llc Fused nitrogen containing heterocycles and compositions thereof as kinase inhibitors
US8158636B2 (en) 2008-05-19 2012-04-17 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
PE20091846A1 (es) 2008-05-19 2009-12-16 Plexxikon Inc DERIVADOS DE PIRROLO[2,3-d]-PIRIMIDINA COMO MODULADORES DE CINASAS
EP2282995B1 (en) 2008-05-23 2015-08-26 Novartis AG Derivatives of quinolines and quinoxalines as protein tyrosine kinase inhibitors
AU2009257635A1 (en) 2008-06-10 2009-12-17 Plexxikon, Inc. 5H-Pyrrolo [2,3-b] pyrazine derivatives for kinase modulation, and indications therefor
WO2009155527A2 (en) 2008-06-19 2009-12-23 Progenics Pharmaceuticals, Inc. Phosphatidylinositol 3 kinase inhibitors
US8629135B2 (en) 2008-07-14 2014-01-14 Queen's University At Kingston Pharmaceutical compositions comprising RET inhibitors and methods for the treatment of cancer
ES2532732T3 (es) 2008-07-29 2015-03-31 Nerviano Medical Sciences S.R.L. Uso de un inhibidor de CDK para el tratamiento del glioma
US20100075916A1 (en) 2008-09-05 2010-03-25 Auspex Pharmaceuticals, Inc. Substituted quinazoline inhibitors of growth factor receptor tyrosine kinases
EP2161271A1 (en) 2008-09-08 2010-03-10 Università Degli Studi Di Milano - Bicocca Alpha-carboline inhibitors of NMP-ALK, RET, and Bcr-Abl
JP5746032B2 (ja) 2008-09-19 2015-07-08 ネルビアーノ・メデイカル・サイエンシーズ・エツセ・エルレ・エルレ 3,4−ジヒドロ−2H−ピロロ[1,2−a]ピラジン−1−オン誘導体
CA2738026C (en) 2008-09-22 2017-01-24 Array Biopharma Inc. Substituted imidazo[1,2b]pyridazine compounds as trk kinase inhibitors
EP2331530B8 (en) 2008-09-26 2013-12-25 National Health Research Institutes Fused multicyclic compounds as protein kinase inhibitors
DK3106463T6 (da) 2008-10-22 2020-02-24 Array Biopharma Inc Pyrazolo[1,5-]pyrimidinforbindelse som trk-kinasehæmmer
PE20131197A1 (es) 2008-10-31 2013-11-06 Genentech Inc Compuestos de pirazolopirimidina como inhibidores de jak y composiciones farmaceuticas que los contienen
WO2010054058A1 (en) 2008-11-06 2010-05-14 Ambit Bioscience Corporation Imidazolothiazole compounds as modulators of protein kinase
EP2376084B1 (en) 2008-11-24 2013-07-17 Nerviano Medical Sciences S.r.l. CDK inhibitor for the treatment of mesothelioma
KR101061599B1 (ko) 2008-12-05 2011-09-02 한국과학기술연구원 비정상 세포 성장 질환의 치료를 위한 단백질 키나아제 저해제인 신규 인다졸 유도체, 이의 약학적으로 허용가능한염 및 이를 유효성분으로 함유하는 약학적 조성물
JO3265B1 (ar) 2008-12-09 2018-09-16 Novartis Ag مثبطات بيريديلوكسى اندولات vegf-r2 واستخدامها لعلاج المرض
CA2744236C (en) 2009-02-12 2021-03-16 Cell Signaling Technology, Inc. Mutant ros expression in human cancer
WO2010111527A1 (en) 2009-03-26 2010-09-30 Plexxikon, Inc. Pyrazolo [ 3, 4 -b] pyridines as kinase inhibitors and their medical use
US8492374B2 (en) 2009-04-29 2013-07-23 Industrial Technology Research Institute Azaazulene compounds
EP3009428B1 (en) 2009-05-08 2018-02-21 Astellas Pharma Inc. Diamino heterocyclic carboxamide compound
US8765747B2 (en) 2009-06-12 2014-07-01 Dana-Farber Cancer Institute, Inc. Fused 2-aminothiazole compounds
ES2570756T3 (es) 2009-06-15 2016-05-20 Nerviano Medical Sciences Srl Derivados pirimidinilpirrolopiridinona sustituidos, proceso para su preparación y su uso como inhibidores de cinasa
AR077468A1 (es) 2009-07-09 2011-08-31 Array Biopharma Inc Compuestos de pirazolo (1,5 -a) pirimidina sustituidos como inhibidores de trk- quinasa
KR101256018B1 (ko) 2009-08-20 2013-04-18 한국과학기술연구원 단백질 키나아제 저해활성을 갖는 1,3,6-치환된 인돌 화합물
KR101147550B1 (ko) 2009-10-22 2012-05-17 한국과학기술연구원 단백질 키나아제 저해활성을 가지는 2,7-치환된 티에노[3,2-d]피리미딘 화합물
KR101116756B1 (ko) 2009-10-27 2012-03-13 한국과학기술연구원 단백질 키나아제 저해활성을 갖는 신규의 1,6-치환된 인돌 화합물
EA029273B1 (ru) 2009-10-29 2018-03-30 Джиноско Киназные ингибиторы
US8629132B2 (en) 2009-11-13 2014-01-14 Genosco Kinase inhibitors
KR101094446B1 (ko) 2009-11-19 2011-12-15 한국과학기술연구원 단백질 키나아제 저해활성을 가지는 2,4,7-치환된 티에노[3,2-d]피리미딘 화합물
EP2519517B1 (en) 2009-12-29 2015-03-25 Dana-Farber Cancer Institute, Inc. Type ii raf kinase inhibitors
JP5676650B2 (ja) 2010-01-29 2015-02-25 ハンミ・サイエンス・カンパニー・リミテッドHanmi Science Co., Ltd. プロテインキナーゼ阻害活性を有するチエノ[3,2−d]ピリミジン誘導体
KR101483215B1 (ko) 2010-01-29 2015-01-16 한미약품 주식회사 단백질 키나아제 저해활성을 갖는 비시클릭 헤테로아릴 유도체
EP2528918B1 (en) 2010-01-29 2014-09-10 Nerviano Medical Sciences S.r.l. 6,7-dihydroimidazo[1,5-a]pyrazin-8(5h)-one derivatives as protein kinase modulators
US9161944B2 (en) 2010-02-18 2015-10-20 Inserm (Institut National De La Sante Et De La Sante Et De La Recherche Medicale) Method for preventing cancer metastasis
AU2011237669B2 (en) 2010-04-06 2016-09-08 Caris Life Sciences Switzerland Holdings Gmbh Circulating biomarkers for disease
US8383793B2 (en) 2010-04-15 2013-02-26 St. Jude Children's Research Hospital Methods and compositions for the diagnosis and treatment of cancer resistant to anaplastic lymphoma kinase (ALK) kinase inhibitors
TWI619713B (zh) 2010-04-21 2018-04-01 普雷辛肯公司 用於激酶調節的化合物和方法及其適應症
RS63063B1 (sr) 2010-08-19 2022-04-29 Zoetis Belgium S A Anti-ngf antitela i njihova upotreba
WO2012034095A1 (en) 2010-09-09 2012-03-15 Irm Llc Compounds and compositions as trk inhibitors
US8637516B2 (en) 2010-09-09 2014-01-28 Irm Llc Compounds and compositions as TRK inhibitors
WO2012047017A2 (ko) 2010-10-05 2012-04-12 크리스탈지노믹스(주) 2,3-디히드로-이소인돌-1-온 유도체 및 이를 포함하는 조성물
JP2014005206A (ja) 2010-10-22 2014-01-16 Astellas Pharma Inc アリールアミノヘテロ環カルボキサミド化合物
JP6190723B2 (ja) 2010-12-01 2017-08-30 アルダーバイオ ホールディングス エルエルシー 抗ngf組成物およびその使用
US8618146B2 (en) 2011-01-03 2013-12-31 Dr. Reddy's Laboratories Limited Epothilone compound formulations
EP2668188B1 (en) 2011-01-26 2016-05-18 Nerviano Medical Sciences S.r.l. Tricyclic derivatives, process for their preparation and their use as kinase inhibitors
WO2012101032A1 (en) 2011-01-26 2012-08-02 Nerviano Medical Sciences S.R.L. Tricyclic pyrrolo derivatives, process for their preparation and their use as kinase inhibitors
CN102093421B (zh) 2011-01-28 2014-07-02 北京康辰药业有限公司 一种含磷取代基的喹啉类化合物及其制备方法、以及含有该化合物的药物组合物及其应用
TR201816421T4 (tr) 2011-02-07 2018-11-21 Plexxikon Inc Kinaz modülasyonu için bileşikler ve metotlar ve bunların endikasyonları.
WO2012113774A1 (en) 2011-02-24 2012-08-30 Nerviano Medical Sciences S.R.L. Thiazolylphenyl-benzenesulfonamido derivatives as kinase inhibitors
CA2828219A1 (en) 2011-02-25 2012-08-30 Irm Llc Pyrazolo [1,5-a] pyridines as trk inhibitors
WO2012135631A1 (en) 2011-03-30 2012-10-04 Arrien Pharmaeuticals Llc Substituted 5-(pyrazin-2-yl)-1h-pyrazolo [3, 4-b] pyridine and pyrazolo [3, 4-b] pyridine derivatives as protein kinase inhibitors
WO2012135800A1 (en) 2011-04-01 2012-10-04 University Of Utah Research Foundation Substituted n-phenylpyrimidin-2-amine analogs as inhibitors of the axl kinase
EP2702055A1 (en) 2011-04-11 2014-03-05 Nerviano Medical Sciences S.r.l. Pyrazolyl-pyrimidine derivatives as kinase inhibitors
BR112013026137B1 (pt) 2011-04-19 2020-12-01 Nerviano Medical Sciences S.R.L pirimidinil-pirróis substituídos ativos como inibidores da quinase
MX342509B (es) 2011-05-12 2016-10-03 Nerviano Medical Sciences Srl Compuestos de indazol sustituidos como inhibidores de las cinasas de proteina.
KR101960555B1 (ko) 2011-05-13 2019-03-20 어레이 바이오파마 인크. Trka 키나제 저해제로서의 피롤리디닐 유레아 및 피롤리디닐 티오유레아 화합물
RU2477723C2 (ru) 2011-06-16 2013-03-20 Общество С Ограниченной Ответственностью "Фьюжн Фарма" Ингибиторы протеинкиназ (варианты), их применение для лечения онкологических заболеваний и фармацевтическая композиция на их основе
WO2013016720A2 (en) 2011-07-28 2013-01-31 Gerinda Therapeutics, Inc. Novel substituted biarylheterocycle derivatives as protein kinase inhibitors for the treatment of cancer and other diseases
JP6016915B2 (ja) 2011-07-28 2016-10-26 ネルビアーノ・メデイカル・サイエンシーズ・エツセ・エルレ・エルレ キナーゼ阻害剤として活性なアルキニル置換ピリミジニルピロール
CA2848369A1 (en) 2011-08-04 2013-02-07 National Cancer Center Fusion gene of kif5b gene and ret gene, and method for determining effectiveness of cancer treatment targeting fusion gene
SG2014012728A (en) 2011-08-23 2014-06-27 Foundation Medicine Inc Novel kif5b-ret fusion molecules and uses thereof
CA2846496C (en) 2011-09-02 2020-07-14 The Regents Of The University Of California Substituted pyrazolo[3,4-d]pyrimidines and uses thereof
WO2013036232A2 (en) 2011-09-08 2013-03-14 Deciphera Pharmaceuticals, Llc Methods and compositions for the treatment of myeloproliferative diseases and other proliferative diseases
CN102408411B (zh) 2011-09-19 2014-10-22 北京康辰药业股份有限公司 一种含喹啉基的羟肟酸类化合物及其制备方法、以及含有该化合物的药物组合物及其应用
WO2013042137A1 (en) 2011-09-19 2013-03-28 Aurigene Discovery Technologies Limited Bicyclic heterocycles as irak4 inhibitors
ES2660265T3 (es) 2011-10-07 2018-03-21 Nerviano Medical Sciences S.R.L. Derivados de 3,4-dihidropirrolo[1,2-a]pirazin-1(2h)-ona 4-alquil-sustituidos como inhibidores de cinasa
WO2013050446A1 (en) 2011-10-07 2013-04-11 Nerviano Medical Sciences S.R.L. SUBSTITUTED 3,4-DIHYDROPYRROLO[1,2-a]PYRAZIN-1(2H)-ONE DERIVATIVES AS KINASES INHIBITORS
WO2013059740A1 (en) 2011-10-21 2013-04-25 Foundation Medicine, Inc. Novel alk and ntrk1 fusion molecules and uses thereof
AU2012339753A1 (en) 2011-11-14 2014-06-19 Tesaro, Inc. Modulating certain tyrosine kinases
WO2013081188A1 (ja) 2011-11-30 2013-06-06 独立行政法人国立がん研究センター 誘導悪性幹細胞
WO2013088257A1 (en) 2011-12-12 2013-06-20 Dr. Reddy's Laboratories Ltd. Substituted heterocyclic compounds as tropomyosin receptor kinase a (trka) inhibitors
US8377946B1 (en) 2011-12-30 2013-02-19 Pharmacyclics, Inc. Pyrazolo[3,4-d]pyrimidine and pyrrolo[2,3-d]pyrimidine compounds as kinase inhibitors
RU2705577C2 (ru) 2011-12-30 2019-11-08 Ханми Фарм. Ко., Лтд. ПРОИЗВОДНЫЕ ТИЕНО[3,2-d]ПИРИМИДИНА, ОБЛАДАЮЩИЕ ИНГИБИРУЮЩЕЙ АКТИВНОСТЬЮ В ОТНОШЕНИИ ПРОТЕИНКИНАЗ
JP2015109806A (ja) 2012-03-22 2015-06-18 アステラス製薬株式会社 新規ret融合体の検出法
WO2013161919A1 (ja) 2012-04-26 2013-10-31 小野薬品工業株式会社 Trk阻害化合物
JP2013226108A (ja) 2012-04-27 2013-11-07 Astellas Pharma Inc 新規ntrk2活性化変異の検出法
CA2871540A1 (en) 2012-05-10 2013-11-14 Synta Pharmaceuticals Corp. Treating cancer with hsp90 inhibitory compounds
BR112014028841B1 (pt) 2012-05-23 2021-01-12 Nerviano Medical Sciences S.R.L. Formas cristalinas 1 e 2, composição farmacêutica, uso da forma cristalina 1 para o tratamento de um estado de doença tratável pela inibição de alk e processo para a preparação de n-[5-(3,5-difluoro-benzil)-1h-indazol-3-il]-4-(4-metilpiperazin-1-il)-2-(tetra-hidro-piran-4-ilamino)- benzamida
TWI585088B (zh) 2012-06-04 2017-06-01 第一三共股份有限公司 作爲激酶抑制劑之咪唑并[1,2-b]嗒衍生物
DK2872491T3 (da) 2012-07-11 2021-08-09 Blueprint Medicines Corp Inhibitorer af fibroblastvækstfaktorreceptoren
WO2014017491A1 (ja) 2012-07-26 2014-01-30 独立行政法人国立がん研究センター Cep55遺伝子とret遺伝子との融合遺伝子
EP2689778A1 (en) 2012-07-27 2014-01-29 Pierre Fabre Medicament Derivatives of azaindoles or diazaindoles for treating pain
BR112015002152B1 (pt) 2012-08-02 2021-04-27 Nerviano Medical Sciences S.R.L. Compostos de pirróis substituídos ativos como inibidores de quinases, processo para a preparação de tais compostos, composição farmacêutica, método in vitro para a inibição da atividade de proteínas quinases da família jak e produto compreendendo os referidos compostos
WO2014036387A2 (en) 2012-08-31 2014-03-06 The Regents Of The University Of Colorado Methods for diagnosis and treatment of cancer
CN114129566A (zh) 2012-09-07 2022-03-04 埃克塞里艾克西斯公司 用于治疗肺腺癌的met、vegfr和ret的抑制剂
US20140084039A1 (en) 2012-09-24 2014-03-27 Electro Scientific Industries, Inc. Method and apparatus for separating workpieces
SG10201704588XA (en) 2012-09-25 2017-07-28 Chugai Pharmaceutical Co Ltd Ret inhibitor
JP2014082984A (ja) 2012-10-23 2014-05-12 Astellas Pharma Inc 新規ntrk2活性化変異の検出法
WO2014071358A2 (en) 2012-11-05 2014-05-08 Foundation Medicine, Inc. Novel ntrk1 fusion molecules and uses thereof
WO2014071419A2 (en) 2012-11-05 2014-05-08 Foundation Medicine, Inc. Novel fusion molecules and uses thereof
CN104870446B (zh) 2012-11-07 2019-08-13 内尔维阿诺医学科学有限公司 取代的嘧啶基和吡啶基吡咯并吡啶酮类、其制备方法及其作为激酶抑制剂的用途
EP2917183A1 (en) 2012-11-12 2015-09-16 Ignyta, Inc. Bendamustine derivatives and methods of using same
MX366761B (es) 2012-11-13 2019-07-23 Array Biopharma Inc Compuestos bíciclicos de urea, tiourea, guanidina y cianoguanidina útiles para el tratamiento del dolor.
WO2014078408A1 (en) 2012-11-13 2014-05-22 Array Biopharma Inc. Bicyclic heteroaryl urea, thiourea, guanidine and cyanoguanidine compounds as trka kinase inhibitors
US9790178B2 (en) 2012-11-13 2017-10-17 Array Biopharma Inc. Pyrrolidinyl urea, thiourea, guanidine and cyanoguanidine compounds as TrkA kinase inhibitors
WO2014078325A1 (en) 2012-11-13 2014-05-22 Array Biopharma Inc. N-(monocyclic aryl),n'-pyrazolyl-urea, thiourea, guanidine and cyanoguanidine compounds as trka kinase inhibitors
DK2922844T3 (en) 2012-11-13 2018-03-05 Array Biopharma Inc N-PYRROLIDINYL, N'-PYRAZOLYL-URINE, THIOURINE, GUANIDINE AND CYANOGUANIDE COMPOUNDS AS TRKA-KINASE INHIBITORS
US9828360B2 (en) 2012-11-13 2017-11-28 Array Biopharma Inc. Pyrrolidinyl urea, thiourea, guanidine and cyanoguanidine compounds as TrkA kinase inhibitors
WO2014078322A1 (en) 2012-11-13 2014-05-22 Array Biopharma Inc. Thiazolyl and oxazolyl urea, thiourea, guanidine and cyanoguanidine compounds as trka kinase inhibitors
US9809578B2 (en) 2012-11-13 2017-11-07 Array Biopharma Inc. Pyrazolyl urea, thiourea, guanidine and cyanoguanidine compounds as trkA kinase inhibitors
US9546156B2 (en) 2012-11-13 2017-01-17 Array Biopharma Inc. N-bicyclic aryl,N'-pyrazolyl urea, thiourea, guanidine cyanoguanidine compounds as TrkA kinase inhibitors
WO2014078331A1 (en) 2012-11-13 2014-05-22 Array Biopharma Inc. N-(arylalkyl)-n'-pyrazolyl-urea, thiourea, guanidine and cyanoguanidine compounds as trka kinase inhibitors
KR20150090116A (ko) 2012-11-29 2015-08-05 예다 리서치 앤드 디벨럽먼트 캄파니 리미티드 종양 전이를 예방하고, 암을 치료하고 예후를 예측하며, 추정상의 전이 억제인자 물질을 확인하는 방법
CN103848785B (zh) 2012-12-04 2016-07-13 上海医药集团股份有限公司 一类氘代3-氰基喹啉类化合物、其药用组合物、制备方法及其用途
US9447135B2 (en) 2012-12-14 2016-09-20 University Of Kentucky Research Foundation Semi-synthetic mithramycin derivatives with anti-cancer activity
AU2013371146C1 (en) 2012-12-28 2019-01-17 Crystalgenomics, Inc. 2,3-dihydro-isoindole-1-on derivative as BTK kinase suppressant, and pharmaceutical composition including same
FR3000493A1 (fr) 2012-12-28 2014-07-04 Oribase Pharma Nouveaux inhibiteurs de proteines kinases
FR3000492B1 (fr) 2012-12-28 2015-09-11 Oribase Pharma Nouveaux derives azaindole en tant qu'inhibiteurs multikinases
FR3000494B1 (fr) 2012-12-28 2015-08-21 Oribase Pharma Nouveaux derives d'azaindoles en tant qu'inhibiteurs de proteines kinases
US9127055B2 (en) 2013-02-08 2015-09-08 Astellas Pharma Inc. Method of treating pain with anti-human NGF antibody
BR122017003181A2 (pt) 2013-02-19 2019-09-10 Ono Pharmaceutical Co composto inibidor de trk, composição farmacêutica e medicamento compreendendo dito composto e seu uso para profilaxia e/ou terapia de doença relacionada à trk e/ou para inibir trk
US20160010068A1 (en) 2013-02-22 2016-01-14 Boris C. Bastian Fusion polynucleotides and fusion polypeptides associated with cancer and particularly melanoma and their uses as therapeutic and diagnostic targets
WO2014134096A1 (en) 2013-02-27 2014-09-04 Oregon Health & Science University Methods of treating cancers characterized by aberrent ros1 activity
CN105518151B (zh) 2013-03-15 2021-05-25 莱兰斯坦福初级大学评议会 循环核酸肿瘤标志物的鉴别和用途
WO2014160521A1 (en) 2013-03-15 2014-10-02 Blueprint Medicines Corporation Piperazine derivatives and their use as kit modulators
CN105051027A (zh) 2013-03-15 2015-11-11 葛兰素史密斯克莱知识产权发展有限公司 作为转染期间重排(ret)激酶抑制剂的吡啶衍生物
EP2971094B1 (en) * 2013-03-15 2021-09-15 Novartis AG Biomarkers associated with brm inhibition
JP2016515508A (ja) 2013-03-15 2016-05-30 ザ・トラスティーズ・オブ・コロンビア・ユニバーシティ・イン・ザ・シティ・オブ・ニューヨーク 融合タンパク質及びその方法
WO2014152777A2 (en) 2013-03-15 2014-09-25 Insight Genetics, Inc. Methods and compositions for the diagnosis and treatment of cancers resistant to ros1 inhibitors
US8937071B2 (en) 2013-03-15 2015-01-20 Glaxosmithkline Intellectual Property Development Limited Compounds as rearranged during transfection (RET) inhibitors
US20140315199A1 (en) 2013-04-17 2014-10-23 Life Technologies Corporation Gene fusions and gene variants associated with cancer
US10072298B2 (en) 2013-04-17 2018-09-11 Life Technologies Corporation Gene fusions and gene variants associated with cancer
ES2646019T3 (es) 2013-05-14 2017-12-11 Nerviano Medical Sciences S.R.L. Derivados de 6-amino-7-deaza-purina, proceso para su preparación y su uso como inhibidores de cinasa
SG11201509338QA (en) 2013-05-30 2015-12-30 Plexxikon Inc Compounds for kinase modulation, and indications therefor
EP3019489A4 (en) 2013-07-11 2016-12-28 Betta Pharmaceuticals Co Ltd MODULATORS OF TYROSINE KINASE PROTEIN AND METHODS OF USE
US10705087B2 (en) 2013-07-26 2020-07-07 Japanese Foundation For Cancer Research Detection method for NTRK3 fusion
EP3027655B1 (en) 2013-07-30 2019-08-21 Blueprint Medicines Corporation Ntrk2 fusions
WO2015017528A1 (en) 2013-07-30 2015-02-05 Blueprint Medicines Corporation Pik3c2g fusions
CN105658814A (zh) 2013-08-20 2016-06-08 日本国立癌症研究中心 在肺癌中检测出的新型融合基因
SG11201601341QA (en) 2013-08-30 2016-03-30 Ambit Biosciences Corp Biaryl acetamide compounds and methods of use thereof
WO2015039006A1 (en) 2013-09-16 2015-03-19 The General Hospital Corporation Methods of treating cancer
US9334263B2 (en) 2013-10-17 2016-05-10 Blueprint Medicines Corporation Compositions useful for treating disorders related to kit
EP3409674B1 (en) 2013-10-17 2022-04-06 Blueprint Medicines Corporation Process for preparing compositions useful for treating disorders related to kit
KR20160065986A (ko) 2013-10-24 2016-06-09 조지타운 유니버시티 암 치료용 방법 및 조성물
JP6458023B2 (ja) 2013-10-25 2019-01-23 ブループリント メディシンズ コーポレイション 繊維芽細胞成長因子受容体の阻害剤
JPWO2015064621A1 (ja) 2013-10-29 2017-03-09 公益財団法人がん研究会 新規融合体及びその検出法
GB201321146D0 (en) 2013-11-29 2014-01-15 Cancer Rec Tech Ltd Quinazoline compounds
WO2015082887A2 (en) 2013-12-02 2015-06-11 Bergenbio As Use of kinase inhibitors
US9695165B2 (en) 2014-01-15 2017-07-04 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
EA031863B1 (ru) 2014-01-24 2019-03-29 ТиПи ТЕРАПЬЮТИКС, ИНК. Диарильные макроциклы в качестве модуляторов протеинкиназ
US9346788B2 (en) 2014-02-05 2016-05-24 VM Oncology, LLC TrkA receptor tyrosine kinase antagonists and uses thereof
MX2021001583A (es) 2014-02-14 2023-02-08 Exelixis Inc Formas sólidas cristalinas de n-{4-[(6,7-dimetoxiquinolin-4-il)oxi ] fenil}-n'-(4-fluorofenil)ciclopropan-1,1-dicarboxamida, procesos para elaboración y métodos de uso.
US10231965B2 (en) 2014-02-20 2019-03-19 Ignyta, Inc. Molecules for administration to ROS1 mutant cancer cells
WO2015161274A1 (en) 2014-04-18 2015-10-22 Blueprint Medicines Corporation Pik3ca fusions
WO2015161277A1 (en) 2014-04-18 2015-10-22 Blueprint Medicines Corporation Met fusions
MA40434B1 (fr) 2014-05-15 2019-09-30 Array Biopharma Inc 1-((3s,4r)-4-(3-fluorophényl)-1-(2-méthoxyéthyl)pyrrolidin-3-yl)-3-(4-méthyl-3-(2-méthylpyrimidin-5-yl)-1-phényl-1h-pyrazol-5-yl)urée comme inhibiteur de la kinase trka
WO2015183836A1 (en) 2014-05-27 2015-12-03 Brian Haynes Compositions, methods, and uses related to ntrk2-tert fusions
WO2015183837A1 (en) 2014-05-27 2015-12-03 Brian Haynes Compositions, methods, and uses related to ntrk2-tert fusions
US20170114415A1 (en) 2014-05-30 2017-04-27 The Regents Of The University Of Colorado, A Body Corporate Activating ntrk1 gene fusions predictive of kinase inhibitor therapy
US10246750B2 (en) 2014-06-10 2019-04-02 Blueprint Medicines Corporation Method for detection of a TECR:PKN1 or an ANXA4:PKN1 gene fusion
WO2015191666A2 (en) 2014-06-10 2015-12-17 Blueprint Medicines Corporation Raf1 fusions
EP3169809B1 (en) 2014-07-17 2020-04-29 Blueprint Medicines Corporation Prkc fusions
US10370723B2 (en) 2014-07-17 2019-08-06 Blueprint Medicines Corporation TERT fusions
CN105272930B (zh) 2014-07-17 2018-07-13 广东东阳光药业有限公司 取代脲衍生物及其在药物中的应用
EP3169804B3 (en) 2014-07-17 2019-09-18 Blueprint Medicines Corporation Fgr fusions
WO2016022569A1 (en) 2014-08-04 2016-02-11 Blueprint Medicines Corporation Compositions useful for treating disorders related to kit
TR201908736T4 (tr) 2014-08-18 2019-07-22 Ono Pharmaceutical Co Trk-inhibe eden bileşiğin asit eklenmiş tuzu.
CN107148418A (zh) 2014-09-08 2017-09-08 葛兰素史密斯克莱知识产权发展有限公司 2‑(4‑(4‑乙氧基‑6‑氧代‑1,6‑二氢吡啶‑3‑基)‑2‑氟苯基)‑n‑(5‑(1,1,1‑三氟‑2‑甲基丙‑2‑基)异噁唑‑3‑基)乙酰胺的晶型
EP3191450B1 (en) 2014-09-10 2019-04-10 GlaxoSmithKline Intellectual Property Development Limited Pyridone derivatives as rearranged during transfection (ret) kinase inhibitors
MY187169A (en) 2014-09-10 2021-09-07 Glaxosmithkline Ip Dev Ltd Novel compounds as rearranged during transfection (ret) inhibitors
TWI538914B (zh) 2014-10-03 2016-06-21 國立交通大學 蛋白質激酶之選擇性抑制劑、其醫藥組成物及其用途
CU20170052A7 (es) 2014-10-14 2017-11-07 Dana Farber Cancer Inst Inc Moléculas de anticuerpo que se unen a pd-l1
CN107108631B (zh) 2014-11-14 2020-06-16 内尔维阿诺医学科学有限公司 作为蛋白激酶抑制剂的6-氨基-7-二环-7-脱氮-嘌呤衍生物
DK3699181T3 (da) 2014-11-16 2023-03-20 Array Biopharma Inc Krystallinsk form af (s)-n-(5-((r)-2-(2,5-difluorphenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidin-1-carboxamidhydrogensulfat
US20170356052A1 (en) 2014-11-18 2017-12-14 Blueprint Medicines Corporation Prkacb fusions
RU2708674C2 (ru) 2014-12-15 2019-12-11 СиЭмДжи ФАРМАСЬЮТИКАЛ КО., ЛТД. Конденсированные кольцевые гетероарильные соединения и их применение в качестве ингибиторов trk
WO2016096709A1 (en) 2014-12-16 2016-06-23 Eudendron S.R.L. Heterocyclic derivatives modulating activity of certain protein kinases
US10202365B2 (en) 2015-02-06 2019-02-12 Blueprint Medicines Corporation 2-(pyridin-3-yl)-pyrimidine derivatives as RET inhibitors
KR101675984B1 (ko) 2015-02-23 2016-11-14 한양대학교 에리카산학협력단 티에노디아제핀 유도체 또는 이의 약학적으로 허용가능한 염, 및 이를 유효성분으로 포함하는 약학적 조성물
US20180045727A1 (en) 2015-03-03 2018-02-15 Caris Mpi, Inc. Molecular profiling for cancer
US20160273049A1 (en) 2015-03-16 2016-09-22 Personal Genome Diagnostics, Inc. Systems and methods for analyzing nucleic acid
US10364247B2 (en) 2015-04-21 2019-07-30 Ruijin Hospital Affiliated To Shanghai Jiao Tong University School Of Medicine Preparation and use of novel protein kinase inhibitors
RU2020132040A (ru) 2015-05-20 2020-10-12 Те Брод Инститьют Инк. Общие неоантигены
AU2016270321B2 (en) 2015-05-29 2020-09-10 Ignyta, Inc. Compositions and methods for treating patients with RTK mutant cells
EA201792679A1 (ru) 2015-06-01 2018-06-29 Локсо Онколоджи, Инк. Способы диагностики и лечения злокачественной опухоли
AU2015101722A4 (en) 2015-06-19 2016-05-19 Macau University Of Science And Technology Oncogenic ros1 and alk kinase inhibitor
GB201511546D0 (en) 2015-07-01 2015-08-12 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
CN107735399B (zh) * 2015-07-02 2021-01-26 特普医药公司 作为蛋白质激酶的调节剂的手性二芳基大环
GB201512365D0 (en) 2015-07-15 2015-08-19 King S College London Novel therapy
EP3322706B1 (en) 2015-07-16 2020-11-11 Array Biopharma, Inc. Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
EP3120851A1 (en) 2015-07-21 2017-01-25 Pangaea Biotech S.L. 4-amino-6-(2,6-dichlorophenyl)-8-methyl-2-(phenylamino)-pyrido[2,3-d]pyrimidin-7(8h)-one for treatment of solid cancers
KR101766194B1 (ko) 2015-08-07 2017-08-10 한국과학기술연구원 RET 키나아제 저해제인 신규 3-(이속사졸-3-일)-피라졸로[3,4-d]피리미딘-4-아민 화합물
WO2017027883A1 (en) 2015-08-13 2017-02-16 San Diego State University Research Foundation Atropisomerism for increased kinase inhibitor selectivity
MA41559A (fr) 2015-09-08 2017-12-26 Taiho Pharmaceutical Co Ltd Composé de pyrimidine condensé ou un sel de celui-ci
WO2017049462A1 (zh) 2015-09-22 2017-03-30 合肥中科普瑞昇生物医药科技有限公司 一类新型的flt3激酶抑制剂及其用途
CN105255927B (zh) 2015-09-30 2018-07-27 温州医科大学附属第一医院 一种kiaa1217-ret融合基因
TN2018000138A1 (en) 2015-10-26 2019-10-04 Array Biopharma Inc Point mutations in trk inhibitor-resistant cancer and methods relating to the same
TWI787018B (zh) 2015-11-02 2022-12-11 美商纜圖藥品公司 轉染過程重排之抑制劑
EP3387145A1 (en) 2015-12-08 2018-10-17 Boehringer Ingelheim International GmbH Method using a ret fusion gene as a biomarker to select non-small cell lung cancer (nsclc) and thyroid cancer patients for a cancer treatment
JP6871869B2 (ja) 2016-01-15 2021-05-19 公益財団法人がん研究会 新規融合体及びその検出法
US20170224662A1 (en) 2016-01-22 2017-08-10 The Medicines Company Aqueous Formulations and Methods of Preparation and Use Thereof
TWI620748B (zh) 2016-02-05 2018-04-11 National Health Research Institutes 氨基噻唑化合物及其用途
WO2017145050A1 (en) 2016-02-23 2017-08-31 Glaxosmithkline Intellectual Property Development Limited Pyridylpyridone derivative useful as a ret kinase inhibitor in the treatment of ibs and cancer
PT3269370T (pt) 2016-02-23 2020-03-05 Taiho Pharmaceutical Co Ltd Novo composto de pirimidina condensada ou sal do mesmo
WO2017155018A1 (ja) 2016-03-11 2017-09-14 小野薬品工業株式会社 Trk阻害剤抵抗性の癌治療剤
TW201738228A (zh) 2016-03-17 2017-11-01 藍圖醫藥公司 Ret之抑制劑
PE20181888A1 (es) 2016-04-04 2018-12-11 Loxo Oncology Inc Formulaciones liquidas de (s)-n-(5-((r)-2-(2,5-difluorofenil)-pirrolidin-1-il)-pirazolo[1,5-a]pirimidin-3-il)-3-hidroxipirrolidina-1-carboxamida
US10045991B2 (en) 2016-04-04 2018-08-14 Loxo Oncology, Inc. Methods of treating pediatric cancers
SG10201911665UA (en) 2016-04-15 2020-01-30 Cancer Research Tech Ltd Heterocyclic compounds as ret kinase inhibitors
DK3442535T3 (da) 2016-04-15 2022-09-05 Cancer Research Tech Ltd Heterocykliske stoffer som as ret-kinase-hæmmere
EP3445361A1 (en) 2016-04-19 2019-02-27 Exelixis, Inc. Triple negative breast cancer treatment method
WO2017197051A1 (en) 2016-05-10 2017-11-16 C4 Therapeutics, Inc. Amine-linked c3-glutarimide degronimers for target protein degradation
WO2017201156A1 (en) 2016-05-18 2017-11-23 Duke University Method of treating kras wild-type metastatic colorectal cell carcinoma using cabozantinib plus panitumumab
EP3800189B1 (en) 2016-05-18 2023-06-28 Loxo Oncology, Inc. Preparation of (s)-n-(5-((r)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
WO2017207696A1 (en) 2016-06-01 2017-12-07 F. Hoffmann-La Roche Ag Novel mutations in anaplastic lymphoma kinase predicting response to alk inhibitor therapy in lung cancer patients
WO2018017983A1 (en) 2016-07-22 2018-01-25 Blueprint Medicines Corporation Compounds useful for treating disorders related to ret
TWI704148B (zh) 2016-10-10 2020-09-11 美商亞雷生物製藥股份有限公司 作為ret激酶抑制劑之經取代吡唑并[1,5-a]吡啶化合物
JOP20190092A1 (ar) 2016-10-26 2019-04-25 Array Biopharma Inc عملية لتحضير مركبات بيرازولو[1، 5-a]بيريميدين وأملاح منها
WO2018136796A1 (en) 2017-01-20 2018-07-26 Exelixis, Inc. Combinations of cabozantinib and atezolizumab to treat cancer
CN108456163A (zh) 2017-02-20 2018-08-28 中国科学院上海药物研究所 含邻氨基杂芳环炔基的化合物及其制备方法和用途
JOP20190213A1 (ar) 2017-03-16 2019-09-16 Array Biopharma Inc مركبات حلقية ضخمة كمثبطات لكيناز ros1
WO2019084285A1 (en) 2017-10-26 2019-05-02 Qian Zhao FORMULATIONS OF A MACROCYCLIC TRK KINASE INHIBITOR

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6463922B2 (en) * 2000-01-24 2002-10-15 Graniterie Des Ecorces Wedge intended to be inserted in a cutting slot
US8933084B2 (en) * 2010-05-20 2015-01-13 Array Biopharma Inc. Macrocyclic compounds as Trk kinase inhibitors
US20160032404A1 (en) * 2014-08-01 2016-02-04 Pharmacyclics Llc Biomarkers for predicting response of dlbcl to treatment with a btk inhibitor
US20160367547A1 (en) * 2015-06-19 2016-12-22 Macau University Of Science And Technology Oncogenic ros1 and alk kinase inhibitor

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10590139B2 (en) 2008-09-22 2020-03-17 Array Biopharma Inc. Method of treatment using substituted imidazo[1,2b]pyridazine compounds
US11267818B2 (en) 2008-10-22 2022-03-08 Array Biopharma Inc. Method of treatment using substituted pyrazolo[1,5-a] pyrimidine compounds
US10774085B2 (en) 2008-10-22 2020-09-15 Array Biopharma Inc. Method of treatment using substituted pyrazolo[1,5-A] pyrimidine compounds
US10758542B2 (en) 2009-07-09 2020-09-01 Array Biopharma Inc. Substituted pyrazolo[l,5-a]pyrimidine compounds as Trk kinase inhibitors
US10647730B2 (en) 2010-05-20 2020-05-12 Array Biopharma Inc. Macrocyclic compounds as TRK kinase inhibitors
US10813936B2 (en) 2014-11-16 2020-10-27 Array Biopharma, Inc. Crystalline form of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-YL)-pyrazolo[1,5-A]pyrimidin-3-YL)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate
US10799505B2 (en) 2014-11-16 2020-10-13 Array Biopharma, Inc. Crystalline form of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-A]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate
US10655186B2 (en) 2015-10-26 2020-05-19 Loxo Oncology, Inc. Point mutations in TRK inhibitor-resistant cancer and methods relating to the same
US10724102B2 (en) 2015-10-26 2020-07-28 Loxo Oncology, Inc. Point mutations in TRK inhibitor-resistant cancer and methods relating to the same
US10907215B2 (en) 2015-10-26 2021-02-02 Loxo Oncology, Inc. Point mutations in TRK inhibitor-resistant cancer and methods relating to the same
US11484535B2 (en) 2016-04-04 2022-11-01 Loxo Oncology, Inc. Liquid formulations of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a] pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
US10668072B2 (en) 2016-04-04 2020-06-02 Loxo Oncology, Inc. Liquid formulations of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
US10588908B2 (en) 2016-04-04 2020-03-17 Loxo Oncology, Inc. Methods of treating pediatric cancers
US11191766B2 (en) 2016-04-04 2021-12-07 Loxo Oncology, Inc. Methods of treating pediatric cancers
US11214571B2 (en) 2016-05-18 2022-01-04 Array Biopharma Inc. Process for the preparation of (S)-N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide and salts thereof
US11091486B2 (en) 2016-10-26 2021-08-17 Array Biopharma, Inc Process for the preparation of pyrazolo[1,5-a]pyrimidines and salts thereof
US10966985B2 (en) 2017-03-16 2021-04-06 Array Biopharma Inc. Macrocyclic compounds as ROS1 kinase inhibitors
US10688100B2 (en) 2017-03-16 2020-06-23 Array Biopharma Inc. Macrocylic compounds as ROS1 kinase inhibitors
WO2020198379A1 (en) * 2019-03-26 2020-10-01 Ventyx Biosciences, Inc. Tyk2 pseudokinase ligands
US11780842B2 (en) 2019-03-26 2023-10-10 Ventyx Biosciences, Inc. TYK2 pseudokinase ligands
US11753411B2 (en) 2019-11-08 2023-09-12 Ventyx Biosciences, Inc. Substituted pyrazolo[1,5-a]pyrimidines as TYK2 pseudokinase ligands
CN113004305A (zh) * 2019-12-19 2021-06-22 成都倍特药业股份有限公司 大环化合物及其制备方法和用途
WO2022182845A1 (en) * 2021-02-25 2022-09-01 Blossomhill Therapeutics, Inc. Macrocycles and their use

Also Published As

Publication number Publication date
SG11201908532UA (en) 2019-10-30
US20190151322A1 (en) 2019-05-23
AU2018234726A1 (en) 2019-09-26
WO2018170381A1 (en) 2018-09-20
MA49888A (fr) 2020-06-24
IL269216A (en) 2019-11-28
PH12019502069A1 (en) 2020-09-14
CA3056754A1 (en) 2018-09-20
EP3595651A1 (en) 2020-01-22
RU2019132666A (ru) 2021-04-16
BR112019019101A2 (pt) 2020-04-22
US10966985B2 (en) 2021-04-06
JP2020514356A (ja) 2020-05-21
TN2019000263A1 (en) 2021-01-07
CN110769820A (zh) 2020-02-07
JOP20190213A1 (ar) 2019-09-16
CL2019002612A1 (es) 2020-05-29
MX2019010988A (es) 2020-02-05
US20190076437A1 (en) 2019-03-14
KR20190126003A (ko) 2019-11-07
US10688100B2 (en) 2020-06-23
TW201838631A (zh) 2018-11-01

Similar Documents

Publication Publication Date Title
US10966985B2 (en) Macrocyclic compounds as ROS1 kinase inhibitors
US11851434B2 (en) Substituted pyrazolo[1,5-A]pyrazine compounds as ret kinase inhibitors
JP7337133B2 (ja) TAMおよびMETキナーゼの阻害薬としてのピラゾロ[3,4-b]ピリジン化合物
CA3039760C (en) Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
US11603374B2 (en) Substituted pyrrolo[2,3-d]pyrimidines compounds as ret kinase inhibitors
CA3079012A1 (en) Crystalline forms
US11472802B2 (en) Substituted pyrazolyl[4,3-c]pyridine compounds as RET kinase inhibitors
CA3039912A1 (en) Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
US11524963B2 (en) Substituted pyrazolo[3,4-d]pyrimidines as RET kinase inhibitors
US11964988B2 (en) Fused heterocyclic compounds as RET kinase inhibitors
WO2018136663A1 (en) Ret inhibitors
US20210023086A1 (en) Treatment of trk-associated cancers
US20230365584A1 (en) Heterocyclic pericondensed cdc7 kinase inhibitors for the treatment of cancer

Legal Events

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

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

AS Assignment

Owner name: ARRAY BIOPHARMA INC., COLORADO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ANDREWS, STEVEN W.;BLAKE, JAMES F.;HAAS, JULIA;AND OTHERS;SIGNING DATES FROM 20190510 TO 20190529;REEL/FRAME:049844/0134

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION