US20210139517A1 - Bicyclic heteroaryl compounds and uses thereof - Google Patents

Bicyclic heteroaryl compounds and uses thereof Download PDF

Info

Publication number
US20210139517A1
US20210139517A1 US17/090,342 US202017090342A US2021139517A1 US 20210139517 A1 US20210139517 A1 US 20210139517A1 US 202017090342 A US202017090342 A US 202017090342A US 2021139517 A1 US2021139517 A1 US 2021139517A1
Authority
US
United States
Prior art keywords
membered
alkyl
pharmaceutically acceptable
compound
tautomer
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
US17/090,342
Other languages
English (en)
Inventor
Adrian L. Gill
Andreas BUCKL
Elena S. Koltun
Naing Aay
Arlyn A. Tambo-ong
Severin Thompson
Micah James Gliedt
John E. Knox
James Cregg
Anne V. Edwards
Yang Liu
G. Leslie Burnett
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.)
Revolution Medicines Inc
Original Assignee
Revolution Medicines 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 Revolution Medicines Inc filed Critical Revolution Medicines Inc
Priority to US17/090,342 priority Critical patent/US20210139517A1/en
Assigned to Revolution Medicines, Inc. reassignment Revolution Medicines, Inc. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BURNETT, G. Leslie, CREGG, JAMES, TAMBO-ONG, Arlyn A., AAY, NAING, BUCKL, Andreas, EDWARDS, Anne V., GILL, ADRIAN L., GLIEDT, MICAH JAMES, KNOX, JOHN E., KOLTUN, ELENA S., LIU, YANG, THOMPSON, Severin
Priority to US17/133,340 priority patent/US11168102B1/en
Publication of US20210139517A1 publication Critical patent/US20210139517A1/en
Priority to US18/045,647 priority patent/US20230365605A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6564Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms
    • C07F9/6581Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and nitrogen atoms with or without oxygen or sulfur atoms, as ring hetero atoms
    • C07F9/6584Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and nitrogen atoms with or without oxygen or sulfur atoms, as ring hetero atoms having one phosphorus atom as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6564Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms
    • C07F9/6568Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus atoms as the only ring hetero atoms
    • C07F9/65685Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus atoms as the only ring hetero atoms the ring phosphorus atom being part of a phosphine oxide or thioxide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Definitions

  • the present disclosure relates to inhibitors of SOS1 useful in the treatment of diseases or disorders. Specifically, the present disclosure is concerned with compounds and compositions inhibiting SOS1, methods of treating diseases associated with SOS1, and methods of synthesizing these compounds.
  • RAS-family proteins including KRAS (V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog), NRAS (neuroblastoma RAS viral oncogene homolog) and HRAS (Harvey murine sarcoma virus oncogene) and any mutants thereof are small GTPases that exist in cells in either GTP-bound or GDP-bound states (McCormick et al., J. Mol. Med. (Berl), 2016, 94(3):253-8; Nimnual et al., Sci. STKE., 2002, 2002(145):p 136).
  • the RAS-family proteins have a weak intrinsic GTPase activity and slow nucleotide exchange rates (Hunter et al., Mol. Cancer Res., 2015, 13(9): 1325-35). Binding of GTPase activating proteins (GAPs) such as NF1 increases the GTPase activity of RAS-family proteins.
  • GAPs GTPase activating proteins
  • NF1 NF1
  • GEFs guanine nucleotide exchange factors
  • RAS-family proteins When in the GTP-bound state, RAS-family proteins are active and engage effector proteins including RAF and phosphoinositide 3-kinase (PI3K) to promote the RAF/mitogen or extracellular signal-regulated kinases (MEK/ERK).
  • PI3K phosphoinositide 3-kinase
  • MEK/ERK extracellular signal-regulated kinases
  • SOS1 is also recruited to other phosphorylated cell surface receptors such as the T cell Receptor (TCR), B cell Receptor (BCR) and monocyte colony-stimulating factor receptor (Salojin et al., J. Biol. Chem. 2000, 275(8):5966-75).
  • TCR T cell Receptor
  • BCR B cell Receptor
  • monocyte colony-stimulating factor receptor Salojin et al., J. Biol. Chem. 2000, 275(8):5966-75.
  • SOS1-activation of RAS-family proteins can also be mediated by the interaction of SOS1/Grb2 with the BCR-ABL oncoprotein commonly found in chronic myelogenous leukemia (Kardinal et al., 2001, Blood, 98:1773-81; Sini et al., Nat. Cell Biol., 2004, 6(3):268-74). Furthermore, alterations in SOS1 have been implicated in cancer.
  • hereditary SOS1 mutations are implicated in the pathogenesis of RASopathies like e.g., Noonan syndrome (NS), cardio-facio-cutaneous syndrome (CFC) and hereditary gingival fibromatosis type 1 (Pierre et al., Biochem. Pharmacol., 2011, 82(9):1049-56).
  • SOS1 is also a GEF for the activation of the GTPases RAC1 (Ras-related C3 botulinum toxin substrate 1) (Innocenti et al., J. Cell Biol., 2002, 156(1):125-36).
  • RAC1 Ras-related C3 botulinum toxin substrate 1
  • RAC1 Ras-related C3 botulinum toxin substrate 1
  • RAC1 Ras-related C3 botulinum toxin substrate 1
  • SOS2 Son of Sevenless 2
  • SOS2 Son of Sevenless 2
  • RAS-family proteins Rost al., Biochem. Pharmacol., 2011, 82(9): 1049-56; Buday et al., Biochim. Biophys. Acta., 2008, 1786(2):178-87.
  • SOS1 inhibitor compounds are be expected to consequently inhibit signaling in cells downstream of RAS-family proteins (e.g., ERK phosphorylation).
  • SOS1 inhibitor compounds are be expected to deliver anti-cancer efficacy (e.g., inhibition of proliferation, survival, metastasis, etc.).
  • SOS1 inhibitor compound High potency towards inhibition of SOS1:RAS-family protein binding (nanomolar level IC 50 values) and ERK phosphorylation in cells (nanomolar level IC 50 values) are desirable characteristics for a SOS1 inhibitor compound. Furthermore, a desirable characteristic of a SOS1 inhibitor compound would be the selective inhibition of SOS1 over SOS2. This conclusion is based on the viable phenotype of SOS1 knockout mice and lethality of SOS1/SOS2 double knockout mice, as described above.
  • the present disclosure relates to compounds capable of inhibiting the activity of SOS1.
  • the present disclosure further provides a process for the preparation of compounds, pharmaceutical preparations comprising such compounds and methods of using such compounds and compositions in the management of diseases or disorders associated with the aberrant activity of SOS1.
  • R 1 is selected from the group consisting of optionally substituted 3-6 membered cycloalkyl, optionally substituted 3-6 membered heterocyclyl, optionally substituted 6-membered aryl, and optionally substituted 5-6 membered heteroaryl;
  • R 2 is selected from the group consisting of H, C 1-6 alkyl, halogen, —NHR 2a , —OR 2a , cyclopropyl, and —CN; wherein C 1-6 alkyl is optionally substituted with halogen, —NHR 2a , —OR 2a , or 5-6 membered heterocyclyl, and further wherein R 2a is selected from the group consisting of H, C 1-6 alkyl, 3-6 membered heterocyclyl, and C 1-6 haloalkyl;
  • R 3 is selected from the group consisting of H, C 1-3 alkyl, —OR 3a , cyclopropyl, and 3-6 membered heterocyclyl, wherein each of C 1-3 alkyl, cyclopropyl, and 3-6 membered heterocyclyl is optionally substituted with R 3a , and further wherein R 3a is selected from the group consisting of C 1-3 alkyl, halogen, —OH, or —CN;
  • L 4 is selected from the group consisting of bond, —C(O)—, —C(O)O—, —C(O)NH(CH 2 ) o —, —NH—, —S—, —S(O) 2 —,
  • R 4 is selected from the group consisting of H, C 1-6 alkyl, 3-14 membered cycloalkyl, 3-14 membered cycloalkenyl, 3-14 membered heterocyclyl, 6-10 membered aryl, and 5-10 membered heteroaryl; wherein each C 1-6 alkyl, 3-14 membered cycloalkyl, 3-14 membered cycloalkenyl, 3-14 membered heterocyclyl, 6-10 membered aryl, and 5-10 membered heteroaryl is optionally substituted with C 1-6 alkyl, —R 4a , —OR 4a , —O—C 1-6 alkyl-R 4a , ⁇ O, halogen, —C(O)R 4a , —C(OO)R 4a , —C(O)NR 4b R 4c , —NR 4b C(O)R 4c , —CN, ⁇ NR 4a , —NR 4b R
  • R 2 , R 3 , L 4 , and R 4 are as defined in Formula (I);
  • R 5 , R 6 , R 7 , R 8 , and R 9 are independently selected from the group consisting of H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, 3-14 membered heterocyclyl, —OH, halogen, —NO 2 , —CN, —NR 11 R 12 , —SR 10 , —S(O) 2 NR 11 R 12 , —S(O) 2 R 10 , —NR 10 S(O) 2 NR 11 R 12 , —NR 10 S(O) 2 NR 11 R 12 , —NR 10 S(O) 2 R 11 , —S(O)NR 11 R 12 , —S(O)R 10 , —NR 10 S(O)NR 11 R 12 , —NR 10 S(O)R 11 , —C(O)R 10 , —CO
  • R 10 , R 11 , and R 12 are at each occurrence independently selected from H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, 3-14 membered heterocyclyl, —OR 13 , —SR 13 , halogen, —NR 13 R 14 , —NO 2 , and —CN; and R 13 and R 14 are at each occurrence independently selected from H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, and 3-14 membered heterocyclyl, wherein each C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, and 3-14 membered hetero
  • Another aspect of the present disclosure relates to compounds of Formula (III),
  • R 2 , R 3 , L 4 , and R 4 are as defined in Formula (I);
  • R 5 , R 6 , R 7 , R 8 , and R 9 are independently selected from the group consisting of H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, 3-14 membered heterocyclyl, —OH, halogen, —NO 2 , —CN, —NR 11 R 12 , —SR 10 , —S(O) 2 NR 11 R 12 , —S(O) 2 R 10 , —NR 10 S(O) 2 NR 11 R 12 , —NR 10 S(O) 2 NR 11 R 12 , —NR 10 S(O) 2 R 11 , —S(O)NR 11 R 12 , —S(O)R 10 , —NR 10 S(O)NR 11 R 12 , —NR 10 S(O)R 11 , —C(O)R 10 , —CO
  • R 10 , R 11 , and R 12 are at each occurrence independently selected from H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, 3-14 membered heterocyclyl, —OR 13 , —SR 13 , halogen, —NR 13 R 14 , —NO 2 , and —CN; and
  • R 13 and R 14 are at each occurrence independently selected from H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, and 3-14 membered heterocyclyl, wherein each C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, and 3-14 membered heterocyclyl are independently optionally substituted with —OH, —SH, —NH 2 , —NO 2 , or —CN.
  • Another aspect of the present disclosure relates to compounds of Formula (IV-a), (IV-b), or (IV-c),
  • R 2 , R 3 , L 4 , and R 4 are as defined in Formula (I);
  • R 5 , R 6 , R 7 , R 8 , and R 9 are independently selected from the group consisting of H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, 3-14 membered heterocyclyl, —OH, halogen, —NO 2 , —CN, —NR 11 R 12 , —SR 10 , —S(O) 2 NR 11 R 12 , —S(O) 2 R 10 , —NR 10 S(O) 2 NR 11 R 12 , —NR 10 S(O) 2 NR 11 R 12 , —NR 10 S(O) 2 R 11 , —S(O)NR 11 R 12 , —S(O)R 10 , —NR 10 S(O)NR 11 R 12 , —NR 10 S(O)R 11 , —C(O)R 10 , —CO
  • R 10 , R 11 , and R 12 are at each occurrence independently selected from H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, 3-14 membered heterocyclyl, —OR 13 , —SR 13 , halogen, —NR 13 R 14 , —NO 2 , and —CN; and
  • R 13 and R 14 are at each occurrence independently selected from H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, and 3-14 membered heterocyclyl, wherein each C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, and 3-14 membered heterocyclyl are independently optionally substituted with —OH, —SH, —NH 2 , —NO 2 , or —CN.
  • One aspect of the present disclosure relates to a method of inhibiting SOS1 in a subject in need thereof, comprising administering to the subject a SOS1 inhibitor of the present invention, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or isomer thereof.
  • Another aspect of the present disclosure relates to methods of treating or preventing a disease that is effected by inhibition of the interaction of SOS1 and a RAS-family protein and/or RAC1 in a subject in need thereof, comprising administering to the subject an effective amount of a compound of any one of Formula (I)-(IV), and pharmaceutically acceptable salts, prodrugs, solvates, hydrates, tautomers, and isomers thereof.
  • Another aspect of the present disclosure relates to methods of treating or preventing cancer in a subject in need thereof, comprising administering to the subject an effective amount of a compound of any one of Formula (I)-(IV), and pharmaceutically acceptable salts, prodrugs, solvates, hydrates, tautomers, and isomers thereof.
  • Another aspect of the present disclosure relates to methods of inhibiting SOS1.
  • the method comprises administering to a patient in need thereof, an effective amount of a compound of any one of Formula (I)-(IV), and pharmaceutically acceptable salts, prodrugs, solvates, hydrates, tautomers, and isomers thereof.
  • compositions comprising a compound of any one of Formula (I)-(IV), and pharmaceutically acceptable salts, prodrugs, solvates, hydrates, tautomers, and isomers thereof, and a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier can further comprise an excipient, diluent, or surfactant.
  • the pharmaceutical composition can be effective for treating or preventing a disease associated with SOS1 modulation in a subject in need thereof.
  • the pharmaceutical composition can be effective for treating or preventing a cancer in a subject in need thereof.
  • Another aspect of the present disclosure relates to a compound of any one of Formula (I)-(IV), and pharmaceutically acceptable salts, prodrugs, solvates, hydrates, tautomers, and isomers thereof, for use in treating or preventing a disease associated with SOS1 modulation.
  • Another aspect of the present disclosure relates to a compound of any one of Formula (I)-(IV), and pharmaceutically acceptable salts, prodrugs, solvates, hydrates, tautomers, and isomers thereof, for use in treating or preventing a disease cancer.
  • Another aspect of the present disclosure relates to the use of a compound of any one of Formula (I)-(IV), and pharmaceutically acceptable salts, prodrugs, solvates, hydrates, tautomers, and isomers thereof, in the manufacture of a medicament for treating or preventing a disease associated with SOS1 modulation.
  • Another aspect of the present disclosure relates to the use of a compound of any one of Formula (I)-(IV) and pharmaceutically acceptable salts, prodrugs, solvates, hydrates, tautomers, and isomers thereof, in the manufacture of a medicament for treating or preventing cancer.
  • the present disclosure also provides compounds that are useful in inhibiting SOS1.
  • an element means one element or more than one element.
  • the term “about” is used to indicate that a value includes the standard deviation of error for the device or method being employed to determine the value.
  • the term “about” refers to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of a stated value, unless otherwise stated or otherwise evident from the context (e.g., where such number would exceed 100% of a possible value).
  • an optionally substituted group may be unsubstituted or substituted by one or more (e.g., 0, 1, 2, 3, 4, or 5 or more, or any range derivable therein) of the substituents listed for that group in which said substituents may be the same or different.
  • an optionally substituted group has 1 substituent.
  • an optionally substituted group has 2 substituents.
  • an optionally substituted group has 3 substituents.
  • an optionally substituted group has 4 substituents.
  • an optionally substituted group has 5 substituents.
  • an alkyl group that is optionally substituted can be a fully saturated alkyl chain (i.e., a pure hydrocarbon).
  • the same optionally substituted alkyl group can have substituents different from hydrogen. For instance, it can, at any point along the chain be bonded to a halogen atom, a hydroxyl group, or any other substituent described herein.
  • optionally substituted means that a given chemical moiety has the potential to contain other functional groups, but does not necessarily have any further functional groups.
  • alkyl may mean a straight chain or branched saturated chain having from 1 to 10 carbon atoms.
  • Representative saturated alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, 2-methyl-1-propyl, 2-methyl-2-propyl, 2-methyl-1-butyl, 3-methyl-1-butyl, 2-methyl-3-butyl, 2,2-dimethyl-1-propyl, 2-methyl-1-pentyl, 3-methyl-1-pentyl, 4-methyl-1-pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-1-butyl, 3,3-dimethyl-1-butyl, 2-ethyl-1-butyl, butyl, isobutyl, t-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl and the
  • alkyl group can be unsubstituted or substituted. Alkyl groups containing three or more carbon atoms may be straight or branched. As used herein, “lower alkyl” means an alkyl having from 1 to 6 carbon atoms.
  • heteroalkyl refers to an “alkyl” group (as defined herein), in which at least one carbon atom has been replaced with a heteroatom (e.g., an O, N, or S atom).
  • a heteroatom e.g., an O, N, or S atom.
  • the heteroatom may appear in the middle or at the end of the radical.
  • alkenyl means an aliphatic hydrocarbon group containing a carbon carbon double bond and which may be straight or branched having about 2 to about 6 carbon atoms in the chain. Certain alkenyl groups have 2 to about 4 carbon atoms in the chain. Branched means that one or more lower alkyl groups such as methyl, ethyl, or propyl are attached to a linear alkenyl chain. Exemplary alkenyl groups include ethenyl, propenyl, n-butenyl, and i-butenyl.
  • a C 2 -C 6 alkenyl group is an alkenyl group containing between 2 and 6 carbon atoms.
  • alkynyl means an aliphatic hydrocarbon group containing a carbon-carbon triple bond and which may be straight or branched having about 2 to about 6 carbon atoms in the chain. Certain alkynyl groups have 2 to about 4 carbon atoms in the chain. Branched means that one or more lower alkyl groups such as methyl, ethyl, or propyl are attached to a linear alkynyl chain. Exemplary alkynyl groups include ethynyl, propynyl, n-butynyl, 2-butynyl, 3-methylbutynyl, and n-pentynyl.
  • a C 2 -C 6 alkynyl group is an alkynyl group containing between 2 and 6 carbon atoms.
  • halo or halogen means a fluoro, chloro, bromo, or iodo group.
  • oxo refers to an “ ⁇ O” group.
  • C(O) When an oxo group is bonded to a carbon atom, it can also be abbreviated herein as C(O) or as C ⁇ O.
  • An oxo group can also be bonded to a sulfur atom (e.g., S ⁇ O and S(O) 2 ) or at phosphorous atom (e.g., P ⁇ O, PO 2 , PO 3 , PO 4 , etc.).
  • immine refers to an “ ⁇ N” group. When an imine is bonded to a carbon atom, it can also be abbreviated herein as C ⁇ N. Nitrogen can also be double bonded to sulfur, e.g., S ⁇ N, which is referred to as a thioimine.
  • annular atoms refers to the total number of ring atoms present in the system. “Annular atoms” therefore does not include the atoms present in a substituent attached to the ring. Thus, the number of “annular atoms” includes all atoms present in a fused ring. For example, a 2-indolyl ring,
  • pyridine is considered a 6-membered heteroaryl, and is a heteroaryl containing 6 annular atoms.
  • Cycloalkyl refers to a single saturated all carbon ring having 3 to 20 annular carbon atoms (i.e., C 3 -C 20 cycloalkyl), for example from 3 to 15 annular atoms, for example, from 3 to 12 annular atoms.
  • the cycloalkyl group is either monocyclic (“monocyclic cycloalkyl”) or contains a fused, bridged or spiro ring system such as a bicyclic system (“bicyclic cycloalkyl”) and can be saturated.
  • Cycloalkyl includes ring systems where the cycloalkyl ring, as defined above, is fused with one or more cycloalkyl, cycloalkenyl, heterocyclyl, aryl or heteroaryl groups, wherein the point of attachment is on a cycloalkyl ring, and, in such instances, the number of carbon atoms recited continues to designate the number of carbons in the cycloalkyl ring containing the point of attachment.
  • Examples of cycloalkyl groups include cyclohexyl ccloheptyl, 2-adamantyl
  • cycloalkyl rings can be further characterized by the number of annular atoms.
  • a cyclohexyl ring is a C 6 cycloalkyl ring with 6 annular atoms
  • 2-(2,3-dihydro-1H-indene) is a C 5 cycloalkyl ring with 9 annular atoms.
  • 9-fluorenyl is a C 5 cycloalkyl ring with 13 annular atoms
  • 2-adamantyl is a C 6 cycloalkyl with 10 annular atoms.
  • cycloalkenyl may refer to a partially saturated, monocyclic, fused or spiro polycyclic, all carbon ring having from 3 to 18 carbon atoms per ring and contains at least one double bond.
  • Cycloalkenyl includes ring systems where the cycloalkenyl ring, as defined above, is fused with one or more cycloalkyl, cycloalkenyl, heterocyclyl, aryl or heteroaryl groups, wherein the point of attachment is on a cycloalkenyl ring, and, in such instances, the number of carbon atoms recited continues to designate the number of carbons in the cycloalkenyl ring containing the point of attachment. Cycloalkenyl rings can be further characterized by the number of annular atoms. Examples of cycloalkenyl include 1-cyclohex-1-enyl and cyclopent-1-enyl.
  • aryl refers to a single all carbon aromatic ring or a multiple condensed all carbon ring system wherein at least one of the rings is aromatic.
  • an aryl group has 5 to 20 annular carbon atoms, 5 to 14 annular carbon atoms, or 5 to 12 annular carbon atoms.
  • Aryl also includes multiple condensed ring systems (e.g., ring systems comprising 2, 3 or 4 rings) having about 9 to 20 carbon atoms in which at least one ring is aromatic and wherein the other rings may be aromatic or not aromatic (i.e., cycloalkyl).
  • Aryl includes ring systems where the aryl ring, as defined above, is fused with one or more cycloalkyl, cycloalkenyl, heterocyclyl, aryl or heteroaryl groups, and wherein the point of attachment is on an aryl ring, and, in such instances, the number of carbon atoms recited continues to designate the number of carbon atoms in the aryl ring containing the point of attachment.
  • aryl groups include phenyl and 5-(2,3-dihydro-1H-indene):
  • aryl rings can be further characterized by the number of annular atoms.
  • phenyl is a C 6 aryl with 6 annular atoms
  • 5-(2,3-dihydro-1H-indene) is a C 6 aryl with 9 annular atoms.
  • Heterocyclyl refers to a single saturated or partially unsaturated non-aromatic ring or a non-aromatic multiple ring system (including fused and spiro polycyclic) that has at least one heteroatom in the ring (at least one annular heteroatom selected from oxygen, nitrogen, phosphorus, and sulfur). Unless otherwise specified, a heterocyclyl group has from 5 to about 20 annular atoms, for example from 5 to 15 annular atoms, for example from 5 to 10 annular atoms.
  • the term includes single saturated or partially unsaturated rings (e.g., 3, 4, 5, 6 or 7-membered rings) having from about 1 to 6 annular carbon atoms and from about 1 to 3 annular heteroatoms selected from the group consisting of oxygen, nitrogen, phosphorus, and sulfur in the ring.
  • the term also includes single saturated or partially unsaturated rings (e.g., 5, 6, 7, 8, 9, or 10-membered rings) having from about 4 to 9 annular carbon atoms and from about 1 to 3 annular heteroatoms selected from the group consisting of oxygen, nitrogen, phosphorus, and sulfur in the ring.
  • Heterocyclyl includes ring systems where the heterocyclyl ring, as defined above, is fused with one or more cycloalkyl, cycloalkenyl, heterocyclyl, aryl or heteroaryl groups, wherein the point of attachment is on a heterocyclic ring, and, in such instances, the number of ring members recited continues to designate the number of annular atoms in the heterocyclic ring containing the point of attachment. Heterocyclic rings can be further characterized by the number of annular atoms.
  • heterocyclic groups examples include piperidinyl (6-membered heterocycle with 6 annular atoms), azepanyl (7-membered heterocycle with 7 annular atoms), and 3-chromanyl (6-membered heterocycle with 10 annular atoms)
  • heteroaryl refers to a single aromatic ring that has at least one atom other than carbon in the ring, wherein the atom is selected from the group consisting of oxygen, nitrogen and sulfur; the term also includes multiple condensed ring systems that have at least one such aromatic ring.
  • the term includes single heteroaryl rings of from about 1 to 10 annular carbon atoms and about 1-5 annular heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur in the rings.
  • the sulfur and nitrogen atoms may also be present in an oxidized form provided the ring is aromatic.
  • Heteroaryl includes ring systems where the heteroaryl ring, as defined above, is fused with one or more cycloalkyl, cycloalkenyl, heterocyclyl, aryl or heteroaryl groups, wherein the point of attachment is on a heteroaryl ring, and, in such instances, the number of ring members continues to designate the number of ring members in the heteroaryl ring containing the point of attachment.
  • Heteroaryl rings can be further characterized by the number of annular atoms. For example, pyridine is a 6-membered heteroaryl having 6 annular atoms.
  • compositions comprising an effective amount of a disclosed compound and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable salts include, e.g., water-soluble and water-insoluble salts, such as the acetate, amsonate (4,4-diaminostilbene-2,2-disulfonate), benzenesulfonate, benzonate, bicarbonate, bisulfate, bitartrate, borate, bromide, butyrate, calcium, calcium edetate, camsylate, carbonate, chloride, citrate, clavulariate, dihydrochloride, edetate, edisylate, estolate, esylate, fiunarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexafluorophosphate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, i
  • tautomers refers to a set of compounds that have the same number and type of atoms, but differ in bond connectivity and are in equilibrium with one another.
  • a “tautomer” is a single member of this set of compounds. Typically a single tautomer is drawn but it is understood that this single structure is meant to represent all possible tautomers that might exist. Examples include enol-ketone tautomerism. When a ketone is drawn it is understood that both the enol and ketone forms are part of the present disclosure.
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • Exemplary isotopes that can be incorporated into compounds of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, and iodine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 32 P, 33 P, 35 S, 18 F, 36 Cl, 123 I and 125 I.
  • Isotopically-labeled compounds e.g., those labeled with 3 H and 14 C
  • Tritiated (i.e., 3 H) and carbon-14 (i.e., 14 C) isotopes can be useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements).
  • one or more hydrogen atoms are replaced by 2 H or 3 H, or one or more carbon atoms are replaced by 13 C- or 14 C-enriched carbon.
  • Positron emitting isotopes such as 15 O, 13 N, 11 C, and 18 F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy.
  • isotopically labeled compounds can generally be prepared by following procedures analogous to those disclosed for compounds of the present invention described herein, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
  • prodrug means a compound which is convertible in vivo by metabolic means (e.g., by hydrolysis) to a disclosed compound.
  • a prodrug is a drug which is inactive in the body, but is transformed in the body typically either during absorption or after absorption from the gastrointestinal tract into the active compound.
  • the conversion of the prodrug into the active compound in the body may be done chemically or biologically (i.e., using an enzyme).
  • solvate refers to a complex of variable stoichiometry formed by a solute and solvent. Such solvents for the purpose of the present disclosure may not interfere with the biological activity of the solute. Examples of suitable solvents include, but are not limited to, water, MeOH, EtOH, and AcOH. Solvates wherein water is the solvent molecule are typically referred to as hydrates. Hydrates include compositions containing stoichiometric amounts of water, as well as compositions containing variable amounts of water.
  • the term “isomer” refers to compounds that have the same composition and molecular weight but differ in physical and/or chemical properties. The structural difference may be in constitution (geometric isomers) or in the ability to rotate the plane of polarized light (stereoisomers). With regard to stereoisomers, the compounds herein may have one or more asymmetric carbon atom and may occur as racemates, racemic mixtures and as individual enantiomers or diastereomers.
  • stereoisomers refers to the set of compounds which have the same number and type of atoms and share the same bond connectivity between those atoms, but differ in three dimensional structure.
  • stereoisomer refers to any member of this set of compounds. For instance, a stereoisomer may be an enantiomer or a diastereomer.
  • enantiomers refers to a pair of stereoisomers which are non-superimposable mirror images of one another.
  • enantiomer refers to a single member of this pair of stereoisomers.
  • racemic refers to a 1:1 mixture of a pair of enantiomers.
  • diastereomers refers to the set of stereoisomers which cannot be made superimposable by rotation around single bonds. For example, cis- and trans-double bonds, endo- and exo-substitution on bicyclic ring systems, and compounds containing multiple stereogenic centers with different relative configurations are considered to be diastereomers.
  • diastereomer refers to any member of this set of compounds.
  • the synthetic route may produce a single diastereomer or a mixture of diastereomers.
  • an “effective amount” when used in connection with a compound is an amount effective for treating or preventing a disease in a subject as described herein.
  • carrier encompasses excipients and diluents and means a material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting a pharmaceutical agent from one organ, or portion of the body, to another organ, or portion of the body of a subject.
  • treating refers to improving at least one symptom of the subject's disorder. Treating includes curing, improving, or at least partially ameliorating the disorder.
  • prevent refers to keeping a disease or disorder from afflicting the subject. Preventing includes prophylactic treatment. For instance, preventing can include administering to the subject a compound disclosed herein before a subject is afflicted with a disease and the administration will keep the subject from being afflicted with the disease.
  • inhibiting includes any measurable or complete inhibition to achieve a desired result. For example, there may be a decrease of about, at most about, or at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or more, or any range derivable therein, reduction of activity (e.g., SOS1.Ras-family protein binding activity) compared to normal.
  • activity e.g., SOS1.Ras-family protein binding activity
  • disorder is used in this disclosure to mean, and is used interchangeably with, the terms disease, condition, or illness, unless otherwise indicated.
  • administer refers to either directly administering a disclosed compound or pharmaceutically acceptable salt of the disclosed compound or a composition to a subject, or administering a prodrug derivative or analog of the compound or pharmaceutically acceptable salt of the compound or composition to the subject, which can form an equivalent amount of active compound within the subject's body.
  • a “patient” or “subject” is a mammal, e.g., a human, mouse, rat, guinea pig, dog, cat, horse, cow, pig, or non-human primate, such as a monkey, chimpanzee, baboon or rhesus.
  • the present disclosure relates to compounds of the following formula (A):
  • R 1 is selected from the group consisting of optionally substituted 3-6 membered cycloalkyl, optionally substituted 3-6 membered heterocycloalkyl, optionally substituted 6-membered aryl, and optionally substituted 5-6 membered heteroaryl;
  • R 2 is selected from the group consisting of H, C 1-6 alkyl, halogen, —NHR 2a , —OR 2a , cyclopropyl, and —CN; wherein C 1-6 alkyl is optionally substituted with halogen, —NHR 2a , —OR 2a , or 5-6 membered heterocycloalkyl, and further wherein R 2a is selected from the group consisting of H, C 1-6 alkyl, 3-6 membered heterocyclyl, and C 1-6 haloalkyl;
  • R 3 is selected from the group consisting of H, C 1-3 alkyl, cyclopropyl, and 3-6 membered heterocycloalkyl, wherein each of C 1-3 alkyl, cyclopropyl, and 3-6 membered heterocycloalkyl is optionally substituted with halogen, —OH, or —CN;
  • L 4 is selected from the group consisting of bond, —C(O)—, —C(O)O—, —C(O)NH(CH 2 ) o —, —NH—, —S—, —S(O) 2 —,
  • R 4 is selected from the group consisting of H, C 1-6 alkyl, 3-14 membered cycloalkyl, 3-14 membered cycloalkenyl, 3-14 membered heterocyclyl, 6-10 membered aryl, and 5-10 membered heteroaryl; wherein each C 1-6 alkyl, 3-14 membered cycloalkyl, 3-14 membered cycloalkenyl, 3-14 membered heterocyclyl, 6-10 membered aryl, and 5-10 membered heteroaryl is optionally substituted with C 1-6 alkyl, —OR 4a , ⁇ O, halogen, —C(O)R 4a , —C(OO)R 4a , —C(O)NR 4b R 4c , —CN, —NR 4b R 4c , 3-6 membered cycloalkyl, 3-7 membered heterocyclyl, 6-10 membered aryl, or 5-10 membered heteroaryl
  • R 4a is H, C 1-6 alkyl, C 1-6 haloalkyl, 3-7 membered heterocyclyl, or —(CH 2 ) r OCH 3 , wherein r is 1, 2, or 3;
  • R 4b is H or C 1-6 alkyl
  • R 4c is H or C 1-6 alkyl.
  • R 2 is selected from the group consisting of H, C 1-6 alkyl, halogen, —NHR 2a , —OR 2a , cyclopropyl, and —CN; wherein C 1-6 alkyl is optionally substituted with halogen, —NHR 2a , —OR 2a , or 5-6 membered heterocycloalkyl, and further wherein R 2a is selected from the group consisting of H, C 1-6 alkyl, 3-6 membered heterocyclyl, and C 1-6 haloalkyl;
  • R 3 is selected from the group consisting of H, C 1-3 alkyl, cyclopropyl, and 3-6 membered heterocycloalkyl, wherein each of C 1-3 alkyl, cyclopropyl, and 3-6 membered heterocycloalkyl is optionally substituted with halogen, —OH, or —CN;
  • L 4 is a bond
  • R 4 is selected from the group consisting of H, C 1-6 alkyl, 3-14 membered cycloalkyl, 3-14 membered cycloalkenyl, 3-14 membered heterocyclyl, 6-10 membered aryl, and 5-10 membered heteroaryl; wherein each C 1-6 alkyl, 3-14 membered cycloalkyl, 3-14 membered cycloalkenyl, 3-14 membered heterocyclyl, 6-10 membered aryl, and 5-10 membered heteroaryl is optionally substituted with C 1-6 alkyl, —OR 4a , ⁇ O, halogen, —C(O)R 4a , —C(OO)R 4a , —C(O)NR 4b R 4c , —CN, —NR 4b R 4c , 3-6 membered cycloalkyl, 3-7 membered heterocyclyl, 6-10 membered aryl, or 5-10 membered heteroaryl
  • R 4a is H, C 1-6 alkyl, C 1-6 haloalkyl, 3-7 membered heterocyclyl, or —(CH 2 )OCH 3 , wherein r is 1, 2, or 3;
  • R 4b is H or C 1-6 alkyl
  • R 4c is H or C 1-6 alkyl
  • R 5 , R 6 , R 7 , R 8 , and R 9 are independently selected from the group consisting of H, D, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 4 -C 8 cycloalkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, —OH, halogen, —NO 2 , —CN, —NR 11 R 12 , —SR 10 , —S(O) 2 NR 11 R 12 , —S(O) 2 R 10 , —NR 10 S(O) 2 NR 11 R 12 , —NR 10 S(O) 2 NR 11 R 12 , —NR 10 S(O) 2 R 11 , —S(O)NR 11 R 12 , —S(O)R 10 , —NR 10 S(O)NR 11 R 12 , —NR 10 S(O)R 11 , —C(O)R 10 , —
  • R 13 and R 14 are independently, at each occurrence, H, D, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 4 -C 8 cycloalkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, 3-14 membered heterocyclyl, wherein each alkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkyl, and heterocyclyl is optionally substituted with—OH, —SH, —NH 2 , —NO 2 , or —CN.
  • the present disclosure relates to compounds having the structure of Formula (I),
  • R 1 is selected from the group consisting of optionally substituted 3-6 membered cycloalkyl, optionally substituted 3-6 membered heterocyclyl, optionally substituted 6-membered aryl, and optionally substituted 5-6 membered heteroaryl;
  • R 2 is selected from the group consisting of H, C 1-6 alkyl, halogen, —NHR 2a , —OR 2a , cyclopropyl, and —CN; wherein C 1-6 alkyl is optionally substituted with halogen, —NHR 2a , —OR 2a , or 5-6 membered heterocyclyl, and further wherein R 2a is selected from the group consisting of H, C 1-6 alkyl, 3-6 membered heterocyclyl, and C 1-6 haloalkyl;
  • R 3 is selected from the group consisting of H, C 1-3 alkyl, —OR 3a , cyclopropyl, and 3-6 membered heterocyclyl, wherein each of C 1-3 alkyl, cyclopropyl, and 3-6 membered heterocyclyl is optionally substituted with R 3a , and further wherein R 3a is selected from the group consisting of C 1-3 alkyl, halogen, —OH, or —CN;
  • L 4 is selected from the group consisting of bond, —C(O)—, —C(O)O—, —C(O)NH(CH 2 )O—, —NH—, —S—, —S(O) 2 —,
  • R 4 is selected from the group consisting of H, C 1-6 alkyl, 3-14 membered cycloalkyl, 3-14 membered cycloalkenyl, 3-14 membered heterocyclyl, 6-10 membered aryl, and 5-10 membered heteroaryl; wherein each C 1-6 alkyl, 3-14 membered cycloalkyl, 3-14 membered cycloalkenyl, 3-14 membered heterocyclyl, 6-10 membered aryl, and 5-10 membered heteroaryl is optionally substituted with C 1-6 alkyl, —R 4a , —OR 4a , —O—C 1-6 alkyl-R 4a , ⁇ O, halogen, —C(O)R 4a , —C(OO)R 4a , —C(O)NR 4b R 4c , —NR 4b C(O)R 4c , —CN, ⁇ NR 4a , —NR 4b R
  • R 1 is the optionally substituted 6-membered aryl.
  • the 6-membered aryl has the following structure:
  • R 5 , R 6 , R 7 , R 8 , and R 9 are as defined below in connection with Formula (II)—(IV).
  • R 1 is the optionally substituted 5-6 membered heteroaryl. In some embodiments, R 1 is a 6-membered heteroaryl having any of the following structures:
  • R 5 , R 6 , R 7 , R 8 , and R 9 are as defined below in connection with Formula (II)—(IV).
  • R 1 is the optionally substituted 5-6 membered heteroaryl.
  • R 1 is a 5-membered heteroaryl having the following structure:
  • R 5 , R 6 , and R 7 are as defined below in connection with Formula (II)—(IV).
  • the present disclosure relates to compounds having the structure of Formula (II),
  • R 2 , R 3 , L 4 , and R 4 are as defined in Formula (I);
  • R 5 , R 6 , R 7 , R 8 , and R 9 are independently selected from the group consisting of H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, 3-14 membered heterocyclyl, —OH, halogen, —NO 2 , —CN, —NR 11 R 12 , —SR 10 , —S(O) 2 NR 11 R 12 , —S(O) 2 R 10 , —NR 10 S(O) 2 NR 11 R 12 , —NR 10 S(O) 2 NR 11 R 12 , —NR 10 S(O) 2 R 11 , —S(O)NR 11 R 12 , —S(O)R 10 , —NR 10 S(O)NR 11 R 12 , —NR 10 S(O)R 11 , —C(O)R 10 , —CO
  • R 10 , R 11 , and R 12 are at each occurrence independently selected from H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, 3-14 membered heterocyclyl, —OR 13 , —SR 13 , halogen, —NR 13 R 14 , —NO 2 , and —CN; and R 13 and R 14 are at each occurrence independently selected from H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, and 3-14 membered heterocyclyl, wherein each C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, and 3-14 membered hetero
  • the present disclosure relates to compounds having the structure of Formula (III),
  • R 2 , R 3 , L 4 , and R 4 are as defined Formula (I);
  • R 5 , R 6 , R 7 , R 8 , and R 9 are independently selected from the group consisting of H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, 3-14 membered heterocyclyl, —OH, halogen, —NO 2 , —CN, —NR 11 R 12 , —SR 10 , —S(O) 2 NR 11 R 12 , —S(O) 2 R 10 , —NR 10 S(O) 2 NR 11 R 12 , —NR 10 S(O) 2 NR 11 R 12 , —NR 10 S(O) 2 R 11 , —S(O)NR 11 R 12 , —S(O)R 10 , —NR 10 S(O)NR 11 R 12 , —NR 10 S(O)R 11 , —C(O)R 10 , —CO
  • R 10 , R 11 , and R 12 are at each occurrence independently selected from H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, 3-14 membered heterocyclyl, —OR 13 , —SR 13 , halogen, —NR 13 R 14 , —NO 2 , and —CN; and
  • R 13 and R 14 are at each occurrence independently selected from H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, and 3-14 membered heterocyclyl, wherein each C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, and 3-14 membered heterocyclyl are independently optionally substituted with —OH, —SH, —NH 2 , —NO 2 , or —CN.
  • the present disclosure relates to compounds having the structure of Formula (IV-a), (IV-b), or (IV-c),
  • R 2 , R 3 , L 4 , and R 4 are as defined Formula (I);
  • R 5 , R 6 , R 7 , R 8 , and R 9 are independently selected from the group consisting of H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, 3-14 membered heterocyclyl, —OH, halogen, —NO 2 , —CN, —NR 11 R 12 , —SR 10 , —S(O) 2 NR 11 R 12 , —S(O) 2 R 10 , —NR 10 S(O) 2 NR 11 R 12 , —NR 10 S(O) 2 NR 11 R 12 , —NR 10 S(O) 2 R 11 , —S(O)NR 11 R 12 , —S(O)R 10 , —NR 10 S(O)NR 11 R 12 , —NR 10 S(O)R 11 , —C(O)R 10 , —CO
  • R 10 , R 11 , and R 12 are at each occurrence independently selected from H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, 3-14 membered heterocyclyl, —OR 13 , —SR 13 , halogen, —NR 13 R 14 , —NO 2 , and —CN; and
  • R 13 and R 14 are at each occurrence independently selected from H, D, C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, and 3-14 membered heterocyclyl, wherein each C 1-6 alkyl, C 2-6 alkenyl, 4-8 membered cycloalkenyl, C 2-6 alkynyl, 3-8 membered cycloalkyl, and 3-14 membered heterocyclyl are independently optionally substituted with —OH, —SH, —NH 2 , —NO 2 , or —CN.
  • the present disclosure relates to compounds having the structure of Formula (IV-a) or Formula (IV-b).
  • one to three of R 5 , R 6 , R 7 , R 8 , and R 9 is C 1-6 alkyl, wherein the alkyl is optionally substituted with halogen.
  • one to three of R 5 , R 6 , R 7 , R 8 , and R 9 is C 1-6 alkyl, wherein the alkyl is optionally substituted with halogen or —OH.
  • one to three of R 5 , R 6 , R 7 , R 8 , and R 9 is C 1-6 alkyl, and one to three of R 5 , R 6 , R 7 , R 8 , and R 9 is C 1-6 alkyl optionally substituted with halogen.
  • one to three of R 5 , R 6 , R 7 , R 8 , and R 9 is halogen, and one to three of R 5 , R 6 , R 7 , R 8 , and R 9 is C 1-6 alkyl optionally substituted with halogen.
  • one to three of R 5 , R 6 , R 7 , R 8 , and R 9 is —NH 2 .
  • one of R 5 , R 6 , R 7 , R 8 , and R 9 is —NH 2 ; and one of R 5 , R 6 , R 7 , R 8 , and R 9 is C 1-6 alkyl optionally substituted with halogen.
  • any two adjacent R 5 , R 6 , R 7 , R, and R 9 forms a 3-14 membered fused ring. In some embodiments of compounds of Formula (II)—(IV), any two adjacent R 5 , R 6 , R 7 , R 8 , and R 9 forms a 3-8 membered fused ring. In some embodiments of compounds of Formula (II)—(IV), any two adjacent R 5 , R 6 , R 7 , R 8 , and R 9 forms a 4-8 membered fused ring.
  • any two adjacent R 5 , R 6 , R 7 , R 8 , and R 9 forms a 4-membered fused ring or a 5-membered fused ring.
  • the fused ring is a 3-8 membered heterocyclyl or a 3-8 membered cycloalkyl.
  • the fused ring is a 4-8 membered heterocyclyl or a 4-8 membered cycloalkyl.
  • the fused ring is a 4-membered heterocyclyl or a 5-membered heterocyclyl.
  • the fused ring is a 4-membered cycloalkyl or a 5-membered cycloalkyl. In some embodiments, the fused ring is optionally substituted with —OH, C 1-6 alkyl, halogen, —NO 2 , oxo, —CN, —R 10 , —OR 10 , —NR 11 R 12 , —SR 10 , —S(O) 2 NR 11 R 12 , —S(O) 2 R 10 , —NR 10 S(O) 2 NR 11 R 12 , —NR 10 S(O) 2 NR 11 R 12 , —NR 10 S(O) 2 R 11 , —S(O)NR 11 R 12 , —S(O)R 10 , —NR 10 S(O)NR 11 R 12 , —NR 10 S(O)R 11 , 3-8 membered cycloalkyl, 3-14 membered heterocyclyl, 6-10 membered aryl, or
  • one or more of R 5 , R 6 , R 7 , R 8 , and R 9 is selected from among —CF 3 , —NH 2 , —F, and —CF 2 CH 2 OH.
  • one of R 5 , R 6 , R 7 , R 8 , and R 9 is-CF 3 and one of R 5 , R 6 , R 7 , R 8 , and R 9 is —NH 2 .
  • one of R 5 , R 6 , R 7 , R 8 , and R 9 is —F and one of R 5 , R 6 , R 7 , R 8 , and R 9 is —CF 2 CH 2 OH.
  • R 1 is selected from among:
  • R 1 is selected from among:
  • R 1 is selected from among:
  • R 1 is selected from among:
  • R 2 is H.
  • R 2 is C 1-6 alkyl. In some embodiments of compounds of Formula (I)-(IV), R 2 is —CH 3 .
  • R 2 is C 1-6 alkyl substituted with 5-6 membered heterocycloalkyl. In some embodiments of compounds of Formula (I)-(IV), R 2 is
  • R 2 is C 1-6 alkyl substituted with —NHR 2a , wherein R 2a is C 1-6 alkyl or 3-6 membered heterocyclyl. In some embodiments of compounds of Formula (I)-(IV), R 2 is selected from among
  • R 2 is C 1-6 alkyl substituted with —OR 2a , wherein R 2a is H or C 1-6 alkyl. In some embodiments of compounds of Formula (I)-(IV), R 2 is —CH 2 OH.
  • R 2 is —NHR 2 a, wherein R 2a is C 1-6 alkyl. In some embodiments of compounds of Formula (I)-(IV), R 2 is —NHCH 3 .
  • R 2 is —OR 2a ; wherein R 2a is C 1-6 alkyl. In some embodiments of compounds of Formula (I)-(IV), R 2 is —OCH 3 .
  • R 3 is C 1-3 alkyl. In some embodiments of compounds of Formula (I)-(IV), R 3 is —CH 3 . In some embodiments of compounds of Formula (I)-(IV), R 3 is —CD 3 .
  • R 3 is C 1-3 alkyl substituted with —OH. In some embodiments of compounds of Formula (I)-(IV), R 3 is —CH 2 CH 2 OH.
  • R 3 is H.
  • R 3 is —OR 3a . In some embodiments of compounds of Formula (I)-(IV), R 3 is —OCH 3 .
  • R 3 is cyclopropyl
  • R 3 is 3-6 membered heterocyclyl. In some embodiments of compounds of Formula (I)-(IV), R 3 is
  • L 4 is selected from the group consisting of bond, —C(O)—, —C(O)O—, —C(O)NH(CH 2 ) o —, —NH—, —S—, —S(O) 2 —,
  • L 4 is a bond.
  • L 4 is —C(O)—.
  • L 4 is —(CH 2 ) p —. In some embodiments of compounds of Formula (I), L 4 is —(CH 2 )—.
  • R 4 is selected from the group consisting of H, C 1-6 alkyl, 3-14 membered cycloalkyl, 3-14 membered cycloalkenyl, 3-14 membered heterocyclyl, 6-10 membered aryl, and 5-10 membered heteroaryl; wherein each C 1-6 alkyl, 3-14 membered cycloalkyl, 3-14 membered cycloalkenyl, 3-14 membered heterocyclyl, 6-10 membered aryl, and 5-10 membered heteroaryl is optionally substituted with C 1-6 alkyl, —R 4a , —OR 4a , —O—C 1-6 alkyl-R 4a , ⁇ O, halogen, —C(O)R 4a , —C(OO)R 4a , —C(O)NR 4b R 4c , —NR 4b C(O)R 4c , —CN
  • R 4 is selected from the group consisting of H, C 1-6 alkyl, 3-14 membered cycloalkyl, 3-14 membered cycloalkenyl, 3-14 membered heterocyclyl, 6-10 membered aryl, and 5-10 membered heteroaryl; wherein each C 1-6 alkyl, 3-14 membered cycloalkyl, 3-14 membered cycloalkenyl, 3-14 membered heterocyclyl, 6-10 membered aryl, and 5-10 membered heteroaryl is optionally substituted with C 6 alkyl —OR 4a , ⁇ O, halogen, —C(O)R 4a , —C(OO)R 4a , —C(O)NR 4b R 4c , —CN, —NR 4b R 4c ,3-6 membered cycloalkyl, 3-7 membered heterocyclyl, 6-10 membered ary
  • R 4a is H, C 1-6 alkyl, C 1-6 haloalkyl, —C(O)R 4b , —C(O)NR 4b R 4c , ⁇ O, 3-6 membered cycloalkyl, 6-10 membered aryl optionally substituted with —OR 4b , —CN, ⁇ N-3-6 membered cycloalkyl, 3-7 membered heterocyclyl, —(CH 2 ) r OCH 3 , or —(CH 2 ) r OH, wherein r is 1, 2, or 3; wherein each R 4b is independently H, C 1-6 alkyl; and wherein each R 4c is independently H or C 1-6 alkyl.
  • R 4a is H, C 1-6 alkyl, C 1-6 haloalkyl, —C(O)R 4b , —C(O)NR 4b R 4c , 3-6 membered cycloalkyl, 6-10 membered aryl optionally substituted with —OR 4b , —CN, 3-7 membered heterocyclyl, —(CH 2 )OCH 3 , or —(CH 2 )OH, wherein r is 1, 2, or 3; wherein each R 4 is independently H, C 1-6 alkyl; and wherein each R 4c is independently H or C 1-6 alkyl.
  • R 4 is 3-14 membered heterocyclyl. In some embodiments of compounds of Formula (I)-(IV), R 4 is substituted 3-14 membered heterocyclyl.
  • R 4 is 3-14 membered heterocyclyl substituted with 3-6 membered heterocyclyl.
  • the heterocyclyl substituent is oxetanyl.
  • R 4 is 3-14 membered heterocyclyl substituted with C 1-6 alkyl. In some embodiments of compounds of Formula (I)-(IV), R 4 is 3-14 membered heterocyclyl substituted with —CH 3 . In some embodiments of compounds of Formula (I)-(IV), R 4 is 3-14 membered heterocyclyl substituted with —CH 2 —, i.e., the substituent is a methylene bridge bridging 2 carbon atoms in the heterocyclyl ring.
  • R 4 is 3-14 membered heterocyclyl substituted with 3-6 membered cycloalkyl.
  • the cycloalkyl substituent is cyclopropyl.
  • R 4 is 3-14 membered heterocyclyl substituted with ⁇ O.
  • the R 4 is a heterocyclyl selected from among:
  • the R 4 is a heterocyclyl selected from among:
  • the R 4 is a heterocyclyl selected from among:
  • the R 4 is a heterocyclyl selected from among:
  • the R 4 is a heterocyclyl selected from among:
  • the R 4 is a heterocyclyl selected from among:
  • the R 4 is a heterocyclyl selected from among:
  • the R 4 is a heterocyclyl selected from among:
  • the R 4 is a heterocyclyl selected from among:
  • the R 4 is a heterocyclyl selected from among:
  • the R 4 is a heterocyclyl selected from among:
  • the R 4 is a heterocyclyl selected from among:
  • the R 4 is a heterocyclyl selected from among:
  • the R 4 is a heterocyclyl selected from among:
  • the R 4 is a heterocyclyl selected from among:
  • R 4 is selected from among:
  • R 4 is 3-14 membered cycloalkyl. In some embodiments, R 4 is substituted 3-14 membered cycloalkyl.
  • R 4 is selected from among:
  • R 4 is 6-10 membered aryl. In some embodiments, R 4 is substituted 6-10 membered aryl. In some embodiments, R 4 is phenyl. In some embodiments, R 4 is phenyl substituted with one or two group selected from among —OCH 3 and —CN.
  • R 4 is 5-10 membered heteroaryl. In some embodiments, R 4 is substituted 5-10 membered heteroaryl. In some embodiments, R 4 is selected from among 1H-pyrrole, thiazole, pyridine, pyridazine, pyrimidine, each of which is optionally substituted with a group selected from among —F, —OCH 3 , and —OCH 2 CH 2 OH.
  • R 4 is selected from among:
  • the present disclosure provides a compound, and pharmaceutically acceptable salts, solvates, stereoisomers, and tautomers thereof, selected from the group consisting of compounds of Table A:
  • Example 60 Example 61. Example 62. Example 63. Example 64. Example 64-1. Example 64-2. Example 64-3. Example 65. Example 66. Example 67. Example 68. Example 68-1. Example 68-2. Example 68-3. Example 69. Example 70. Example 71. Example P-1. Example P-2. Example P-3. Example P-4. Example P-5. Example P-6. Example P-7. Example P-8. Example P-9. Example P-10. Example P-11. Example P-12. Example P-13. Example P-14. Example P-15. Example P-16. Example P-17. Example P-18. Example P-19. Example P-20. Example P-21. Note: any compound shown in brackets in Table A above and anywhere herein indicates that the compound is a diastereomer, and the absolute stereochemistry of such diastereomer may not be known.
  • the present disclosure provides a compound, and pharmaceutically acceptable salts, solvates, stereoisomers, and tautomers thereof, selected from the group consisting of compounds of Collection 1:
  • the present disclosure provides a compound, and pharmaceutically acceptable salts, solvates, stereoisomers, and tautomers thereof, selected from the group consisting of compounds of Collection 2:
  • the present disclosure provides a compound, and pharmaceutically acceptable salts, solvates, stereoisomers, and tautomers thereof, selected from the group consisting of compounds of Collection 3:
  • the present disclosure provides a compound, and pharmaceutically acceptable salts, solvates, stereoisomers, and tautomers thereof, selected from the group consisting of compounds of Collection 4:
  • the present disclosure provides a compound, and pharmaceutically acceptable salts, solvates, stereoisomers, and tautomers thereof, selected from the group consisting of compounds of Collection 5.
  • a person of skill in the art may prepare these using conventional methods, including adaptations of synthetic methods disclosed herein.
  • the compounds of the present invention may be made by a variety of methods, including standard chemistry. Suitable synthetic routes are depicted in the schemes given below.
  • the compounds of any of the formulae described herein may be prepared by methods known in the art of organic synthesis as set forth in part by the following synthetic schemes and examples.
  • protecting groups for sensitive or reactive groups are employed where necessary in accordance with general principles or chemistry.
  • Protecting groups are manipulated according to standard methods of organic synthesis (T. W. Greene and P. G. M. Wuts, “Protective Groups in Organic Synthesis”, Third edition, Wiley, New York 1999). These groups are removed at a convenient stage of the compound synthesis using methods that are readily apparent to those skilled in the art.
  • the selection processes, as well as the reaction conditions and order of their execution, shall be consistent with the preparation of compounds of any formula disclosed herein.
  • the present invention includes both possible stereoisomers (unless specified in the synthesis) and includes not only racemic compounds but the individual enantiomers and/or diastereomers as well.
  • a compound When a compound is desired as a single enantiomer or diastereomer, it may be obtained by stereospecific synthesis or by resolution of the final product or any convenient intermediate. Resolution of the final product, an intermediate, or a starting material may be affected by any suitable method known in the art. See, for example, “Stereochemistry of Organic Compounds” by E. L. Eliel, S. H. Wilen, and L. N. Mander (Wiley-Interscience, 1994).
  • the compounds described herein may be made from commercially available starting materials or synthesized using known organic, inorganic, and/or enzymatic processes.
  • the compounds of the present invention can be prepared in a number of ways well known to those skilled in the art of organic synthesis.
  • compounds of the disclosure can be synthesized using the methods described below, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art. These methods include but are not limited to those methods described below.
  • Building block G-1 can be prepared as outlined in scheme 3.
  • Appropriately substituted aryl or heteroaryl bromides G-2 can be transformed into the correspond acetyl hetero)aryls G-3 by metal halogen exchange, followed by addition of an acylating agent.
  • the ketone functionality can be stereoselectively transformed into the chiral amine using Ellman's method.
  • the compounds of the present disclosure may be suitable for treating diseases characterized by excessive or abnormal cell proliferation such as cancer.
  • cancers for example, the following cancers, tumors and other proliferative diseases may be treated with compounds of the present disclosure, without being restricted thereto:
  • cancers/tumors/carcinomas of the head and neck e.g., tumors/carcinomas/cancers of the nasal cavity, paranasal sinuses, nasopharynx, oral cavity (including lip, gum, alveolar ridge, retromolar trigone, floor of mouth, tongue, hard palate, buccal mucosa), oropharynx (including base of tongue, tonsil, tonsillar pilar, soft palate, tonsillar fossa, pharyngeal wall), middle ear, larynx (including supraglottis, glottis, subglottis, vocal cords), hypopharynx, salivary glands (including minor salivary glands); intraocular cancers (e.g., uveal melanoma), and orbital and adnexal cancers;
  • intraocular cancers e.g., uveal melanoma
  • orbital and adnexal cancers e.g
  • cancers/tumors/carcinomas of the lung e.g., non-small cell lung cancer (NSCLC) (squamous cell carcinoma, spindle cell carcinoma, adenocarcinoma, large cell carcinoma, clear cell carcinoma, bronchioalveolar), small cell lung cancer (SCLC) (oat cell cancer, intermediate cell cancer, combined oat cell cancer);
  • NSCLC non-small cell lung cancer
  • SCLC small cell lung cancer
  • neoplasms of the mediastinum e.g., neurogenic tumors (including neurofibroma, neurilemoma, malignant schwannoma, neurosarcoma, ganglioneuroblastoma, ganglioneuroma, neuroblastoma, pheochromocytoma, paraganglioma), germ cell tumors (including seminoma, teratoma, non-seminoma), thymic tumors (including thymoma, thymolipoma, thymic carcinoma, thymic carcinoid), mesenchymal tumors (including fibroma, fibrosarcoma, lipoma, liposarcoma, myxoma, mesothelioma, leiomyoma, leiomyosarcoma, rhabdomyosarcoma, xanthogranuloma, mesenchymoma, hemangiom
  • cancers/tumors/carcinomas of the gastrointestinal (GI) tract e.g., tumors/carcinomas/cancers of the esophagus, stomach (gastric cancer), pancreas, liver and biliary tree (including hepatocellular carcinoma (HCC), e.g., childhood HCC, fibrolamellar HCC, combined HCC, spindle cell HCC, clear cell HCC, giant cell HCC, carcinosarcoma HCC, sclerosing HCC; hepatoblastoma; cholangiocarcinoma; cholangiocellular carcinoma; hepatic cystadenocarcinoma; angiosarcoma, hemangioendothelioma, leiomyosarcoma, malignant schwannoma, fibrosarcoma, Klatskin tumor), gall bladder, extrahepatic bile ducts, small intestine (including duodenum, jejunum, ileum), large intestin
  • cancers/tumors/carcinomas of the testis e.g., seminomas, non-seminomas;
  • gynecologic cancers/tumors/carcinomas e.g., tumors/carcinomas/cancers of the ovary, fallopian tube, peritoneum, cervix, vulva, vagina, uterine body (including endometrium, fundus);
  • cancers/tumors/carcinomas of the breast e.g., mammary carcinoma (infiltrating ductal, colloid, lobular invasive, tubular, adenocystic, papillary, medullary, mucinous), hormone receptor positive breast cancer (estrogen receptor positive breast cancer, progesterone receptor positive breast cancer), HER2 positive breast cancer, triple negative breast cancer, Paget's disease of the breast;
  • cancers/tumors/carcinomas of the endocrine system e.g., tumors/carcinomas/cancers of the endocrine glands, thyroid gland (thyroid carcinomas/tumors; papillary, follicular, anaplastic, medullary), parathyroid gland (parathyroid carcinoma/tumor), adrenal cortex (adrenal cortical carcinoma/tumors), pituitary gland (including prolactinoma, craniopharyngioma), thymus, adrenal glands, pineal gland, carotid body, islet cell tumors, paraganglion, pancreatic endocrine tumors (PET; non-functional PET, PPoma, gastrinoma, insulinoma, VIPoma, glucagonoma, somatostatinoma, GRFoma, ACTHoma), carcinoid tumors;
  • PET pancreatic endocrine tumors
  • sarcomas of the soft tissues e.g., fibrosarcoma, fibrous histiocytoma, liposarcoma, leiomyosarcoma, rhabdomyosarcoma, angiosarcoma, lymphangiosarcoma, Kaposi's sarcoma, glomus tumor, hemangiopericytoma, synovial sarcoma, giant cell tumor of tendon sheath, solitary fibrous tumor of pleura and peritoneum, diffuse mesothelioma, malignant peripheral nerve sheath tumor (MPNST), granular cell tumor, clear cell sarcoma, melanocytic schwannoma, plexosarcoma, neuroblastoma, ganglioneuroblastoma, neuroepithelioma, extraskeletal Ewings sarcoma, paraganglioma, extraskeletal chondrosarcoma, extraskeletal osteosarcoma, mesen
  • sarcomas of the bone e.g., myeloma, reticulum cell sarcoma, chondrosarcoma (including central, peripheral, clear cell, mesenchymal chondrosarcoma), osteosarcoma (including parosteal, periosteal, high-grade surface, small cell, radiation-induced osteosarcoma, Pagets sarcoma), Ewings tumor, malignant giant cell tumor, adamantinoma, (fibrous) histiocytoma, fibrosarcoma, chordoma, small round cell sarcoma, hemangioendothelioma, hemangiopericytoma, osteochondroma, osteoid osteoma, osteoblastoma, eosinophilic granuloma, chondroblastoma;
  • mesothelioma e.g., pleural mesothelioma, peritoneal mesothelioma;
  • cancers of the skin e.g., basal cell carcinoma, squamous cell carcinoma, Merkels cell carcinoma, melanoma (including cutaneous, superficial spreading, lentigo maligna, acral lentiginous, nodular, intraocular melanoma), actinic keratosis, eyelid cancer;
  • neoplasms of the peripheral and central nervous system and brain e.g., astrocytoma (cerebral, cerebellar, diffuse, fibrillary, anaplastic, pilocytic, protoplasmic, gemistocytary), glioblastoma, gliomas, oligodendrogliomas, oligoastrocytomas, ependymomas, ependymoblastomas, choroid plexus tumors, medulloblastomas, meningiomas, schwannomas, hemangioblastomas, hemangiomas, hemangiopericytomas, neuromas, ganglioneuromas, neuroblastomas, retinoblastomas, neurinomas (e.g., acoustic), spinal axis tumors, neurogenic tumors (including neurofibroma, neurilemoma, malignant schwannoma, neurosarcoma, ganglioneuroblasto
  • B-cell non-Hodgkin lymphomas (including small lymphocytic lymphoma (SLL), lymphoplasmacytoid lymphoma (LPL), mantle cell lymphoma (MCL), follicular lymphoma (FL), diffuse large cell lymphoma (DLCL), Burkitt's lymphoma (BL)), Burkitt leukemia, T-cell non-Hodgkin lymphomas (including anaplastic large cell lymphoma (ALCL), adult T-cell leukemia/lymphoma (ATLL), cutaneous T-cell lymphoma (CTCL), peripheral T-cell lymphoma (PTCL)), lymphoblastic T-cell lymphoma (T-LBL), adult T-cell lymphoma, lymphoblastic B-cell lymphoma (B-LBL), immunocytoma, chronic B-cell lymphocytic leukemia (B-CLL), chronic T-cell
  • NHL small lymphocytic lymphoma
  • LPL lymph
  • All cancers/tumors/carcinomas mentioned above which are characterized by their specific location/origin in the body are meant to include both the primary tumors and the metastatic tumors derived therefrom.
  • epithelial cancers e.g., squamous cell carcinoma (SCC) (carcinoma in situ, superficially invasive, verrucous carcinoma, pseudosarcoma, anaplastic, transitional cell, lymphoepithelial), adenocarcinoma (AC) (well-differentiated, mucinous, papillary, pleomorphic giant cell, ductal, small cell, signet-ring cell, spindle cell, clear cell, oat cell, colloid, adenosquamous, mucoepidermoid, adenoid cystic), mucinous cystadenocarcinoma, acinar cell carcinoma, large cell carcinoma, small cell carcinoma, neuroendocrine tumors (small cell carcinoma, paraganglioma, carcinoid); oncocytic carcinoma; and
  • nonepithilial and mesenchymal cancers e.g., sarcomas (fibrosarcoma, chondrosarcoma, rhabdomyosarcoma, leiomyosarcoma, hemangiosarcoma, giant cell sarcoma, lymphosarcoma, fibrous histiocytoma, liposarcoma, angiosarcoma, lymphangiosarcoma, neurofibrosarcoma), lymphoma, melanoma, germ cell tumors, hematological neoplasms, mixed and undifferentiated carcinomas.
  • sarcomas fibrosarcoma, chondrosarcoma, rhabdomyosarcoma, leiomyosarcoma, hemangiosarcoma, giant cell sarcoma, lymphosarcoma, fibrous histiocytoma, liposarcoma, angiosarcoma, lymphangios
  • the compounds of the present disclosure may be used in therapeutic regimens in the context of first line, second line, or any further line treatments.
  • the compounds of the invention may be used for the prevention, short-term or long-term treatment of the above-mentioned diseases, optionally also in combination with radiotherapy and/or surgery and/or other compounds.
  • the above also includes the use of the compounds of the present disclosure in various methods of treating the above diseases by administering a therapeutically effective dose to a patient in need thereof, as well as the use of these compounds for the manufacture of medicaments for the treatment of such diseases, as well as pharmaceutical compositions including such compounds of the invention, as well as the preparation and/or manufacture of medicaments including such compounds of the invention, and the like. Additional Methods of Using the Disclosed Compounds
  • One aspect of the present disclosure relates to a method of inhibiting SOS1 in a subject in need thereof, comprising administering to the subject a SOS1 inhibitor of the present invention, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or isomer thereof.
  • Another aspect of the present disclosure relates to a method of treating or preventing a disease that is effected or characterized by modification of the interaction of SOS1 and a RAS-family protein and/or RAC1 in a subject in need thereof.
  • the method involves administering to a patient in need of treatment for diseases or disorders associated with SOS1 modulation an effective amount of a compound of any formula disclosed herein, or a pharmaceutically acceptable salt, solvate, isomer, prodrug, or tautomer thereof.
  • a method is provided of inhibiting the interaction of SOS1 and a RAS-family protein in a cell or inhibiting the interaction of SOS1 and RAC1 in a cell, comprising administering to the cell a compound of any formula disclosed herein, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or isomer thereof, and a pharmaceutically acceptable carrier.
  • a method is provided of treating or preventing cancer in a subject in need thereof, comprising administering to the subject an effective amount of a compound of any formula disclosed herein, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or isomer thereof.
  • the disease can be, but is not limited to, cancer.
  • the disease or cancer is selected from the group consisting of pancreatic cancer, lung cancer, colorectal cancer, cholangiocarcinoma, multiple myeloma, melanoma, uterine cancer, endometrial cancer, thyroid cancer, acute myeloid leukemia, JM11L (juvenile rnyelomonocytic leukemia), acute lymphoblastic leukemia/lymphoma, lymphomas, tumors of the central and peripheral nervous system, epithelial and nonepithelial tumors and mesenchymal tumor, bladder cancer, urothelial cancer, gastric cancer, cervical cancer, head and neck squamous cell carcinoma, diffuse large B cell lymphoma, esophageal cancer, chronic lymphocytic leukemia, hepatocellular cancer, breast cancer, ovarian cancer, prostate cancer, glioblastoma, renal cancer and sarcomas
  • the disease can be, but is not limited to, cancer.
  • the disease or cancer is selected from the group consisting of pancreatic cancer, lung cancer, colorectal cancer, cholangiocarcinoma, multiple myeloma, melanoma, uterine cancer, endometrial cancer, thyroid cancer, acute myeloid leukemia, ladder cancer, urothelial cancer, gastric cancer, cervical cancer, head and neck squamous cell carcinoma, diffuse large B cell lymphoma, esophageal cancer, chronic lymphocytic leukemia, hepatocellular cancer, breast cancer, ovarian cancer, prostate cancer, glioblastoma, renal cancer and sarcomas.
  • the disease can be, but is not limited to, a RASopathy.
  • the RASopathy is selected from the group consisting of Neurofibromatosis type 1 (NF1), Noonan Syndrome (NS), Noonan Syndrome with Multiple Lentigines (NSML), Capillary Malformation-Arteriovenous Malformation Syndrome (CM-AVM), Costello Syndrome (CS), Cardio-Facio-Cutaneous Syndrome (CFC), Legius Syndrome, and Hereditary gingival fibromatosis.
  • NF1 Neurofibromatosis type 1
  • NS Noonan Syndrome
  • NSML Noonan Syndrome with Multiple Lentigines
  • CM-AVM Capillary Malformation-Arteriovenous Malformation Syndrome
  • CS Costello Syndrome
  • CFC Cardio-Facio-Cutaneous Syndrome
  • Legius Syndrome and Hereditary gingival fibromatosis.
  • Another aspect of the present disclosure is directed to a method of inhibiting SOS1.
  • the method involves administering to a patient in need thereof an effective amount of a compound of any formula disclosed herein, or a pharmaceutically acceptable salt, solvate, isomer, prodrug, or tautomer thereof.
  • the present disclosure relates to compositions capable of modulating the activity of (e.g., inhibiting) SOS1.
  • the present disclosure also relates to the therapeutic use of such compounds.
  • the disclosed compound can be administered in effective amounts to treat or prevent a disorder and/or prevent the development thereof in subjects.
  • Another aspect of the present disclosure relates to a compound of any formula disclosed herein, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in treating or preventing a disease that is affected by modification of the interaction of SOS1 and a RAS-family protein and/or RAC1.
  • Another aspect of the present disclosure relates to a compound of any formula disclosed herein, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in treating or preventing a disease that is characterized by inhibition of the interaction of SOS1 with a RAS-family protein or the interaction of SOS1 with RAC1.
  • Another aspect of the present disclosure relates to a compound of any formula disclosed herein, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in treating or preventing a disease, wherein the treating or preventing is effected or characterized by inhibition of the interaction of SOS1 and a RAS-family protein or by inhibition of the interaction of SOS1 and RA.
  • Another aspect of the present disclosure relates to a compound of any formula disclosed herein, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use inhibiting the binding of hSOS1 to H- or N- or K-RAS including their clinically known mutations and which inhibits the nucleotide exchange reaction catalyzed by hSOS1 in the presence of a concentration of 20 M or lower, but which are substantially inactive against EGFR-kinase at concentrations of 2 ⁇ M or lower for the preparation of a medicament for the treatment or prophylaxis of a hyperproliferative disorder.
  • Another aspect of the present disclosure relates to a compound of any formula disclosed herein, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for the manufacture of a medicament for use inhibiting the binding of hSOS1 specifically to K-RAS G12C protein or another Ras mutant, as described herein, and which inhibits the nucleotide exchange reaction catalyzed by hSOS1 in the presence of a concentration of 2 ⁇ M or lower, but which are substantially inactive against EGFR-kinase at concentrations of 2 ⁇ M or lower for the preparation of a medicament for the treatment or prophylaxis of a hyperproliferative disorder.
  • the present disclosure relates to the use of a compound of any formula disclosed herein, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for treating or preventing a disease.
  • Administration of the disclosed compounds can be accomplished via any mode of administration for therapeutic agents. These modes include systemic or local administration such as oral, nasal, parenteral, intravenous, transdermal, subcutaneous, vaginal, buccal, rectal or topical administration modes. Likewise, they can also be administered in intravenous (both bolus and infusion), intraperitoneal, subcutaneous or intramuscular form, and all using forms well known to those skilled in the pharmaceutical arts.
  • the disclosed compounds or pharmaceutical compositions can be in solid, semi-solid or liquid dosage form, such as, for example, injectables, tablets, suppositories, pills, time-release capsules, elixirs, tinctures, emulsions, syrups, powders, liquids, suspensions, or the like, sometimes in unit dosages and consistent with conventional pharmaceutical practices.
  • Illustrative pharmaceutical compositions are tablets and gelatin capsules comprising a compound of the disclosure and a pharmaceutically acceptable carrier, such as a) a diluent, e.g., purified water, triglyceride oils, such as hydrogenated or partially hydrogenated vegetable oil, or mixtures thereof, corn oil, olive oil, sunflower oil, safflower oil, fish oils, such as EPA or DHA, or their esters or triglycerides or mixtures thereof, omega-3 fatty acids or derivatives thereof, lactose, dextrose, sucrose, mannitol, sorbitol, cellulose, sodium, saccharin, glucose and/or glycine; b) a lubricant, e.g., silica, talcum, stearic acid, its magnesium or calcium salt, sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and/or polyethylene glycol; for tablets also; c
  • Liquid, particularly injectable, compositions can, for example, be prepared by dissolution, dispersion, etc.
  • the disclosed compound is dissolved in or mixed with a pharmaceutically acceptable solvent such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form an injectable isotonic solution or suspension.
  • a pharmaceutically acceptable solvent such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like.
  • Proteins such as albumin, chylomicron particles, or serum proteins can be used to solubilize the disclosed compounds.
  • the disclosed compounds can be also formulated as a suppository that can be prepared from fatty emulsions or suspensions; using polyalkylene glycols such as propylene glycol, as the carrier.
  • the disclosed compounds can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles.
  • Liposomes can be formed from a variety of phospholipids, containing cholesterol, stearylamine or phosphatidylcholines.
  • a film of lipid components is hydrated with an aqueous solution of drug to a form lipid layer encapsulating the drug, as described for instance in U.S. Pat. No. 5,262,564, the contents of which are hereby incorporated by reference.
  • Disclosed compounds can also be delivered by the use of monoclonal antibodies as individual carriers to which the disclosed compounds are coupled.
  • the disclosed compounds can also be coupled with soluble polymers as targetable drug carriers.
  • Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspanamidephenol, or polyethyleneoxidepolylysine substituted with palmitoyl residues.
  • the disclosed compounds can be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels.
  • a polymer e.g., a polycarboxylic acid polymer, or a polyacrylate.
  • Parental injectable administration is generally used for subcutaneous, intramuscular or intravenous injections and infusions.
  • Injectables can be prepared in conventional forms, either as liquid solutions or suspensions or solid forms suitable for dissolving in liquid prior to injection.
  • compositions comprising a compound of the present disclosure and a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier can further include an excipient, diluent, or surfactant.
  • compositions can be prepared according to conventional mixing, granulating or coating methods, respectively, and the present pharmaceutical compositions can contain from about 0.1% to about 99%, from about 5% to about 90%, or from about 1% to about 20% of the disclosed compound by weight or volume.
  • the dosage regimen utilizing the disclosed compound is selected in accordance with a variety of factors including type, species, age, weight, sex, and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal or hepatic function of the patient; and the particular disclosed compound employed.
  • a physician or veterinarian of ordinary skill in the art can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition.
  • Effective dosage amounts of the disclosed compounds when used for the indicated effects, range from about 0.5 mg to about 5000 mg of the disclosed compound as needed to treat the condition.
  • Compositions for in vivo or in vitro use can contain about 0.5, 5, 20, 50, 75, 100, 150, 250, 500, 750, 1000, 1250, 2500, 3500, or 5000 mg of the disclosed compound, or, in a range of from one amount to another amount in the list of doses.
  • the compositions are in the form of a tablet that can be scored.
  • the methods of the invention may include a compound of the invention used alone or in combination with one or more additional therapies (e.g., non-drug treatments or therapeutic agents).
  • Combination therapy may, for example, combine two therapies or may combine three therapies (e.g., a triple therapy of three therapeutic agents), or more.
  • the dosages of one or more of the additional therapies may be reduced from standard dosages when administered alone. For example, doses may be determined empirically from drug combinations and permutations or may be deduced by isobolographic analysis (e.g., Black et al., Neurology 65:S3-S6 (2005)).
  • a compound of the present invention may be administered before, after, or concurrently with one or more of such additional therapies.
  • dosages of a compound of the invention and dosages of the one or more additional therapies e.g., non-drug treatment or therapeutic agent
  • a therapeutic effect e.g., synergistic or additive therapeutic effect
  • a compound of the present invention and an additional therapy such as an anti-cancer agent, may be administered together, such as in a unitary pharmaceutical composition, or separately and, when administered separately, this may occur simultaneously or sequentially. Such sequential administration may be close or remote in time.
  • the additional therapy is the administration of side-effect limiting agents (e.g., agents intended to lessen the occurrence or severity of side effects of treatment.
  • side-effect limiting agents e.g., agents intended to lessen the occurrence or severity of side effects of treatment.
  • the compounds of the present invention can also be used in combination with a therapeutic agent that treats nausea.
  • agents that can be used to treat nausea include: dronabinol, granisetron, metoclopramide, ondansetron, and prochlorperazine, or pharmaceutically acceptable salts thereof.
  • the one or more additional therapies includes a non-drug treatment (e.g., surgery or radiation therapy).
  • the one or more additional therapies includes a therapeutic agent (e.g., a compound or biologic that is an anti-angiogenic agent, signal transduction inhibitor, antiproliferative agent, glycolysis inhibitor, or autophagy inhibitor).
  • the one or more additional therapies includes a non-drug treatment (e.g., surgery or radiation therapy) and a therapeutic agent (e.g., a compound or biologic that is an anti-angiogenic agent, signal transduction inhibitor, antiproliferative agent, glycolysis inhibitor, or autophagy inhibitor).
  • the one or more additional therapies includes two therapeutic agents.
  • the one or more additional therapies includes three therapeutic agents.
  • the one or more additional therapies includes four or more therapeutic agents.
  • non-drug treatments include, but are not limited to, radiation therapy, cryotherapy, hyperthermia, surgery (e.g., surgical excision of tumor tissue), and T cell adoptive transfer (ACT) therapy.
  • radiation therapy e.g., radiation therapy, cryotherapy, hyperthermia
  • surgery e.g., surgical excision of tumor tissue
  • T cell adoptive transfer (ACT) therapy e.g., T cell adoptive transfer
  • the compounds of the invention may be used as an adjuvant therapy after surgery. In some embodiments, the compounds of the invention may be used as a neo-adjuvant therapy prior to surgery.
  • Radiation therapy may be used for inhibiting abnormal cell growth or treating a hyperproliferative disorder, such as cancer, in a subject (e.g., mammal (e.g., human)).
  • a subject e.g., mammal (e.g., human)
  • Techniques for administering radiation therapy are known in the art. Radiation therapy can be administered through one of several methods, or a combination of methods, including, without limitation, external-beam therapy, internal radiation therapy, implant radiation, stereotactic radiosurgery, systemic radiation therapy, radiotherapy and permanent or temporary interstitial brachy therapy.
  • brachy therapy refers to radiation therapy delivered by a spatially confined radioactive material inserted into the body at or near a tumor or other proliferative tissue disease site.
  • Suitable radiation sources for use as a cell conditioner of the present invention include both solids and liquids.
  • the radiation source can be a radionuclide, such as I-125, I-131, Yb-169, Ir-192 as a solid source, I-125 as a solid source, or other radionuclides that emit photons, beta particles, gamma radiation, or other therapeutic rays.
  • the radioactive material can also be a fluid made from any solution of radionuclide(s), e.g., a solution of I-125 or I-131, or a radioactive fluid can be produced using a slurry of a suitable fluid containing small particles of solid radionuclides, such as Au-198, or Y-90.
  • the radionuclide(s) can be embodied in a gel or radioactive micro spheres.
  • the compounds of the present invention can render abnormal cells more sensitive to treatment with radiation for purposes of killing or inhibiting the growth of such cells. Accordingly, this invention further relates to a method for sensitizing abnormal cells in a mammal to treatment with radiation which comprises administering to the mammal an amount of a compound of the present invention, which amount is effective to sensitize abnormal cells to treatment with radiation. The amount of the compound in this method can be determined according to the means for ascertaining effective amounts of such compounds described herein. In some embodiments, the compounds of the present invention may be used as an adjuvant therapy after radiation therapy or as a neo-adjuvant therapy prior to radiation therapy.
  • the non-drug treatment is a T cell adoptive transfer (ACT) therapy.
  • the T cell is an activated T cell.
  • the T cell may be modified to express a chimeric antigen receptor (CAR).
  • CAR modified T (CAR-T) cells can be generated by any method known in the art.
  • the CAR-T cells can be generated by introducing a suitable expression vector encoding the CAR to a T cell. Prior to expansion and genetic modification of the T cells, a source of T cells is obtained from a subject.
  • T cells can be obtained from a number of sources, including peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors. In certain embodiments of the present invention, any number of T cell lines available in the art may be used. In some embodiments, the T cell is an autologous T cell. Whether prior to or after genetic modification of the T cells to express a desirable protein (e.g., a CAR), the T cells can be activated and expanded generally using methods as described, for example, in U.S. Pat. Nos.
  • a desirable protein e.g., a CAR
  • a therapeutic agent may be a compound used in the treatment of cancer or symptoms associated therewith.
  • a therapeutic agent may be a steroid.
  • the one or more additional therapies includes a steroid.
  • Suitable steroids may include, but are not limited to, 21-acetoxypregnenolone, alclometasone, algestone, amcinonide, beclomethasone, betamethasone, budesonide, chloroprednisone, clobetasol, clocortolone, cloprednol, corticosterone, cortisone, cortivazol, deflazacort, desonide, desoximetasone, dexamethasone, diflorasone, diflucortolone, difuprednate, enoxolone, fluazacort, fiucloronide, flumethasone, flunisolide, fluocinolone acetonide, fluocinonide, fluocortin butyl, fluocortolone, fluorometholone
  • a therapeutic agent may be a biologic (e.g., cytokine (e.g., interferon or an interleukin such as IL-2)) used in treatment of cancer or symptoms associated therewith.
  • the biologic is an immunoglobulin-based biologic, e.g., a monoclonal antibody (e.g., a humanized antibody, a fully human antibody, an Fc fusion protein, or a functional fragment thereof) that agonizes a target to stimulate an anti-cancer response or antagonizes an antigen important for cancer.
  • antibody-drug conjugates are also included.
  • a therapeutic agent may be a checkpoint inhibitor.
  • the checkpoint inhibitor is an inhibitory antibody (e.g., a monospecific antibody such as a monoclonal antibody).
  • the antibody may be, e.g., humanized or fully human.
  • the checkpoint inhibitor is a fusion protein, e.g., an Fc-receptor fusion protein.
  • the checkpoint inhibitor is an agent, such as an antibody, that interacts with a checkpoint protein.
  • the checkpoint inhibitor is an agent, such as an antibody, that interacts with the ligand of a checkpoint protein.
  • the checkpoint inhibitor is an inhibitor (e.g., an inhibitory antibody or small molecule inhibitor) of CTLA-4 (e.g., an anti-CTLA-4 antibody or fusion a protein).
  • the checkpoint inhibitor is an inhibitor or antagonist (e.g., an inhibitory antibody or small molecule inhibitor) of PD-1.
  • the checkpoint inhibitor is an inhibitor or antagonist (e.g., an inhibitory antibody or small molecule inhibitor) of PDL-1.
  • the checkpoint inhibitor is an inhibitor or antagonist (e.g., an inhibitory antibody or Fc fusion or small molecule inhibitor) of PDL-2 (e.g., a PDL-2/Ig fusion protein).
  • the checkpoint inhibitor is an inhibitor or antagonist (e.g., an inhibitory antibody or small molecule inhibitor) of B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands, or a combination thereof.
  • an inhibitor or antagonist e.g., an inhibitory antibody or small molecule inhibitor of B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands, or a combination thereof.
  • the checkpoint inhibitor is pembrolizumab, nivolumab, PDR001 (NVS), REGN2810 (Sanofi/Regeneron), a PD-L1 antibody such as, e.g., avelumab, durvalumab, atezolizumab, pidilizumab, JNJ-63723283 (JNJ), BGB-A317 (BeiGene & Celgene) or a checkpoint inhibitor disclosed in Preusser, M. et al. (2015) Nat. Rev.
  • a PD-L1 antibody such as, e.g., avelumab, durvalumab, atezolizumab, pidilizumab, JNJ-63723283 (JNJ), BGB-A317 (BeiGene & Celgene) or a checkpoint inhibitor disclosed in Preusser, M. et al. (2015) Nat. Rev.
  • Neurol. including, without limitation, ipilimumab, tremelimumab, nivolumab, pembrolizumab, AMP224, AMP514/MEDIO680, BMS936559, MED14736, MPDL3280 ⁇ , MSB0010718C, BMS986016, IMP321, lirilumab, IPH2101, 1-7F9, and KW-6002.
  • a therapeutic agent may be an agent that treats cancer or symptoms associated therewith (e.g., a cytotoxic agent, non-peptide small molecules, or other compound useful in the treatment of cancer or symptoms associated therewith, collectively, an “anti-cancer agent”).
  • Anti-cancer agents can be, e.g., chemotherapeutics or targeted therapy agents.
  • Anti-cancer agents include mitotic inhibitors, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, alkylating agents, antimetabolites, folic acid analogs, pyrimidine analogs, purine analogs and related inhibitors, vinca alkaloids, epipodopyyllotoxins, antibiotics, L-Asparaginase, topoisomerase inhibitors, interferons, platinum coordination complexes, anthracenedione substituted urea, methyl hydrazine derivatives, adrenocortical suppressant, adrenocorticosteroides, progestins, estrogens, antiestrogen, androgens, antiandrogen, and gonadotropin-releasing hormone analog.
  • anti-cancer agents include leucovorin (LV), irenotecan, oxaliplatin, capecitabine, paclitaxel, and doxetaxel.
  • the one or more additional therapies includes two or more anti-cancer agents.
  • the two or more anti-cancer agents can be used in a cocktail to be administered in combination or administered separately. Suitable dosing regimens of combination anti-cancer agents are known in the art and described in, for example, Saltz et al., Proc. Am. Soc. Cin. Oncol. 18:233a (1999), and Douillard et al., Lancet 355(9209):1041-1047 (2000).
  • anti-cancer agents include Gleevec® (Imatinib Mesylate); Kyprolis® (carfilzomib); Velcade® (bortezomib); Casodex (bicalutamide); Iressa® (gefitinib); alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; call
  • dynemicin such as dynemicin A; bisphosphonates such as clodronate; an esperamicin; neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores, aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, calicheamicin, carabicin, caminomycin, carminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, adriamycin (doxorubicin), morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin, deoxydoxorubicin,
  • doxorubicin morpholino-doxorubi
  • anti-cancer agents include trastuzumab (Herceptin®), bevacizumab (Avastin®), cetuximab (Erbitux®), rituximab (Rituxan®), Taxol®, Arimidex®, ABVD, avicine, abagovomab, acridine carboxamide, adecatumumab, 17-N-allylamino-17-demethoxygeldanamycin, alpharadin, alvocidib, 3-aminopyridine-2-carboxaldehyde thiosemicarbazone, amonafide, anthracenedione, anti-CD22 immunotoxins, antineoplastics (e.g., cell-cycle nonspecific antineoplastic agents, and other antineoplastics described herein), antitumorigenic herbs, apaziquone, atiprimod, azathioprine, belotecan, bendamustine, BIBW 2992,
  • anti-cancer agents include natural products such as vinca alkaloids (e.g., vinblastine, vincristine, and vinorelbine), epidipodophyllotoxins (e.g., etoposide and teniposide), antibiotics (e.g., dactinomycin (actinomycin D), daunorubicin, and idarubicin), anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin), mitomycin, enzymes (e.g., L-asparaginase which systemically metabolizes L-asparagine and deprives cells which do not have the capacity to synthesize their own asparagine), antiplatelet agents, antiproliferative/antimitotic alkylating agents such as nitrogen mustards (e.g., mechlorethamine, cyclophosphamide and analogs, melphalan, and chlorambucil),
  • nitrogen mustards
  • an anti-cancer agent is selected from mechlorethamine, camptothecin, ifosfamide, tamoxifen, raloxifene, gemcitabine, Navelbine®, sorafenib, or any analog or derivative variant of the foregoing.
  • an anti-cancer agent is an ALK inhibitor.
  • ALK inhibitors include ceritinib, TAE-684 (NVP-TAE694), PF02341066 (crizotinib or 1066), alectinib; brigatinib; entrectinib; ensartinib (X-396); lorlatinib; ASP3026; CEP-37440; 4SC-203; TL-398; PLB1003; TSR-011; CT-707; TPX-0005, and AP26113. Additional examples of ALK kinase inhibitors are described in examples 3-39 of WO05016894.
  • an anti-cancer agent is an inhibitor of a member downstream of a Receptor Tyrosine Kinase (RTK)/Growth Factor Receptor (e.g., a SHP2 inhibitor (e.g., SHP099, TNO155, RMC-4550, RMC-4630, JAB-3068, RLY-1971), another SOS1 inhibitor (e.g., BI-1701963), a Raf inhibitor, a MEK inhibitor, an ERK inhibitor, a PI3K inhibitor, a PTEN inhibitor, an AKT inhibitor, or an mTOR inhibitor (e.g., mTORCl inhibitor or mTORC2 inhibitor).
  • RTK Receptor Tyrosine Kinase
  • Growth Factor Receptor e.g., a SHP2 inhibitor (e.g., SHP099, TNO155, RMC-4550, RMC-4630, JAB-3068, RLY-1971), another SOS1 inhibitor (e.g., BI-1701
  • an anti-cancer agent is a Ras inhibitor (e.g., AMG 510, MRTX1257, LY349946, MRTX849, ARS-3248 (JNJ-74699157), MRTX1133 or ARS-1620), or a Ras vaccine, or another therapeutic modality designed to directly or indirectly decrease the oncogenic activity of Ras.
  • a Ras inhibitor e.g., AMG 510, MRTX1257, LY349946, MRTX849, ARS-3248 (JNJ-74699157), MRTX1133 or ARS-1620
  • Ras vaccine e.g., another therapeutic modality designed to directly or indirectly decrease the oncogenic activity of Ras.
  • the Ras protein is wild-type. Accordingly, in some embodiments, a compound of the present invention is employed in a method of treating a patient having a cancer comprising a Ras WT (e.g., K-Ras WT , H-Ras WT or N-Ras WT ). In some embodiments, the Ras protein is Ras amplification (e.g., K-Ras amp ). Accordingly, in some embodiments, a compound of the present invention is employed in a method of treating a patient having a cancer comprising a Ras amp (K-Ras amp , H-Ras amp or N-Ras amp ). In some embodiments, the cancer comprises a Ras mutation (Ras MUT ). In some embodiments, a mutation is selected from:
  • a cancer comprises an NF1 LOF mutation.
  • the cancer comprises a Ras MUT and a compound of the present invention is administered to, e.g., a patient in need thereof, in combination with an additional therapeutic agent, e.g., a EK inhibitor, such as a MEK inhibitor described herein.
  • the cancer is colorectal cancer and a compound of the present invention is administered to, e.g., a patient in need thereof, in combination with an additional therapeutic agent, such as a topoisomerase I inhibitor (e.g., irinotecan).
  • a topoisomerase I inhibitor e.g., irinotecan
  • the cancer is non-small cell lung cancer and a compound of the present invention is administered to, e.g., a patient in need thereof, in combination with an additional therapeutic agent, e.g., a MEK inhibitor, such as a MEK inhibitor described herein (e.g., trametinib).
  • a MEK inhibitor such as a MEK inhibitor described herein (e.g., trametinib).
  • the cancer is non-small cell lung cancer or colorectal cancer, and a compound of the present invention is administered to, e.g., a patient in need thereof, in combination with a Ras inhibitor, such as a Ras inhibitor described herein (e.g., MRTX849, MRTX1133 or AMG-510).
  • a therapeutic agent that may be combined with a compound of the present invention is an inhibitor of the MAP kinase (MAPK) pathway (or “MAPK inhibitor”).
  • MAPK inhibitors include, but are not limited to, one or more MAPK inhibitor described in Cancers (Basel) 2015 September; 7(3): 1758-1784.
  • the MAPK inhibitor may be selected from one or more of trametinib, binimetinib, selumetinib, cobimetinib, LErafAON (NeoPharm), ISIS 5132; vemurafenib, pimasertib, TAK733, R04987655 (CH4987655); CI-1040; PD-0325901; CH5126766; MAP855; AZD6244; refametinib (RDEA 119/BAY 86-9766); GDC-0973/XL581; AZD8330 (ARRY-424704/ARRY-704); RO5126766 (Roche, described in PLoS One. 2014 Nov. 25; 9(11)); and GSK1120212 (or JTP-74057, described in Clin Cancer Res. 2011 Mar. 1; 17(5):989-1000).
  • an anti-cancer agent is a disrupter or inhibitor of the RAS-RAF-ERK or PI3K-AKT-TOR or PI3K-AKT signaling pathways.
  • the PI3K/AKT inhibitor may include, but is not limited to, one or more PI3K/AKT inhibitor described in Cancers (Basel) 2015 September; 7(3): 1758-1784.
  • the PI3K/AKT inhibitor may be selected from one or more of NVP-BEZ235; BGT226; XL765/SAR245409; SF1126; GDC-0980; PI-103; PF-04691502; PKI-587; GSK2126458.
  • an anti-cancer agent is a PD-1 or PD-L1 antagonist.
  • additional therapeutic agents include EGFR inhibitors, IGF-1R inhibitors, MEK inhibitors, PI3K inhibitors, AKT inhibitors, TOR inhibitors, MCL-1 inhibitors, BCL-2 inhibitors, SHP2 inhibitors, proteasome inhibitors, and immune therapies.
  • IGF-1R inhibitors include linsitinib, or a pharmaceutically acceptable salt thereof.
  • EGFR inhibitors include, but are not limited to, small molecule antagonists, antibody inhibitors, or specific antisense nucleotide or siRNA.
  • Useful antibody inhibitors of EGFR include cetuximab (Erbitux®), panitumumab (Vectibix®), zalutumumab, nimotuzumab, and matuzumab.
  • Further antibody-based EGFR inhibitors include any anti-EGFR antibody or antibody fragment that can partially or completely block EGFR activation by its natural ligand.
  • Non-limiting examples of antibody-based EGFR inhibitors include those described in Modjtahedi et al., Br. J.
  • the EGFR inhibitor can be monoclonal antibody Mab E7.6.3 (Yang, 1999 supra), or Mab C225 (ATCC Accession No. HB-8508), or an antibody or antibody fragment having the binding specificity thereof.
  • Small molecule antagonists of EGFR include gefitinib (Iressa®), erlotinib (Tarceva®), and lapatinib (TykerB®). See, e.g., Yan et al., Pharmacogenetics and Pharmacogenomics In Oncology Therapeutic Antibody Development, BioTechniques 2005, 39(4):565-8; and Paez et al., EGFR Mutations In Lung Cancer Correlation With Clinical Response To Gefitinib Therapy, Science 2004, 304(5676):1497-500.
  • small molecule EGFR inhibitors include any of the EGFR inhibitors described in the following patent publications, and all pharmaceutically acceptable salts of such EGFR inhibitors: EP 0520722; EP 0566226; WO96/33980; U.S. Pat. No.
  • EGFR inhibitors include any of the EGFR inhibitors described in Traxler et al., Exp. Opin. Ther. Patents 1998, 8(12):1599-1625.
  • an EGFR inhibitor is osimertinib.
  • MEK inhibitors include, but are not limited to, pimasertib, selumetinib, cobimetinib (Cotellic®), trametinib (Mekinist®), and binimetinib (Mektovi®).
  • a MEK inhibitor targets a MEK mutation that is a Class I MEK1 mutation selected from D67N; P124L; P124S; and L177V.
  • the MEK mutation is a Class II MEK1 mutation selected from ⁇ E51-Q58; ⁇ F53-Q58; E203K; L177M; C121S; F53L; K57E; Q56P; and K57N.
  • PI3K inhibitors include, but are not limited to, wortmannin; 17-hydroxywortmannin analogs described in WO06/044453; 4-[2-(1H-Indazol-4-yl)-6-[[4-(methylsulfonyl)piperazin-1-yl]methyl]thieno[3,2-d]pyrimidin-4-yl]morpholine (also known as pictilisib or GDC-0941 and described in WO09/036082 and WO09/055730); 2-methyl-2-[4-[3-methyl-2-oxo-8-(quinolin-3-yl)-2,3-dihydroimidazo[4,5-c]quinolin-1-yl]phenyl]propionitrile (also known as BEZ 235 or NVP-BEZ 235, and described in WO6/122806); (S)-1-(4-((2-(2-aminopyrimidin-5-yl)-7-methyl-4-morpholinothieno[3,
  • PI3K inhibitors include demethoxyviridin, perifosine, CAL101, PX-866, BEZ235, SF1126, INK1117, IPI-145, BKM120, XL147, XL765, Palomid 529, GSK1059615, ZSTK474, PWT33597, IC87114, TGI 00-115, CAL263, PI-103, GNE-477, CUDC-907, and AEZS-136.
  • AKT inhibitors include, but are not limited to, Akt-1-1 (inhibits Aktl) (Barnett et al., Biochem. J. 2005, 385(Pt. 2): 399-408); Akt-1-1,2 (inhibits Akl and 2) (Barnett et al., Biochem. J. 2005, 385(Pt. 2): 399-408); API-59CJ-Ome (e.g., Jin et al., Br. J. Cancer 2004, 91:1808-12); 1-H-imidazo[4,5-c]pyridinyl compounds (e.g., WO 05/011700); indole-3-carbinol and derivatives thereof (e.g., U.S. Pat. No.
  • mTOR inhibitors include, but are not limited to, ATP-competitive mTORC1/mTORC2 inhibitors, e.g., PI-103, PP242, PP30; Torin 1; FKBP12 enhancers; 4H-1-benzopyran-4-one derivatives; and rapamycin (also known as sirolimus) and derivatives thereof, including: temsirolimus (Torisel®); everolimus (Afinitor®; WO94/09010); ridaforolimus (also known as deforolimus or AP23573); rapalogs, e.g., as disclosed in WO98/02441 and WO01/14387, e.g., AP23464 and AP23841; 40-(2-hydroxyethyl)rapamycin; 40-[3-hydroxy(hydroxymethyl)methylpropanoate]-rapamycin (also known as CC1779); 40-epi-(tetrazolyt)-rap
  • the mTOR inhibitor is a bisteric inhibitor (see, e.g., WO2018204416, WO2019212990 and WO2019212991), such as RMC-5552.
  • BRAF inhibitors that may be used in combination with compounds of the invention include, for example, vemurafenib, dabrafenib, and encorafenib.
  • a BRAF may comprise a Class 3 BRAF mutation.
  • the Class 3 BRAF mutation is selected from one or more of the following amino acid substitutions in human BRAF: D287H; P367R; V459L; G466V; G466E; G466A; S467L; G469E; N581S; N5811; D594N; D594G; D594A; D594H; F595L; G596D; G596R and A762E.
  • MCL-1 inhibitors include, but are not limited to, AMG-176, MIK665, and S63845.
  • the myeloid cell leukemia-1 (MCL-1) protein is one of the key anti-apoptotic members of the B-cell lymphoma-2 (BCL-2) protein family.
  • BCL-1 B-cell lymphoma-2
  • Over-expression of MCL-1 has been closely related to tumor progression as well as to resistance, not only to traditional chemotherapies but also to targeted therapeutics including BCL-2 inhibitors such as ABT-263.
  • the additional therapeutic agent is a SHP2 inhibitor.
  • SHP2 is a non-receptor protein tyrosine phosphatase encoded by the PTPN11 gene that contributes to multiple cellular functions including proliferation, differentiation, cell cycle maintenance and migration.
  • SHP2 has two N-terminal Src homology 2 domains (N—SH2 and C—SH2), a catalytic domain (PTP), and a C-terminal tail.
  • the two SH2 domains control the subcellular localization and functional regulation of SHP2.
  • the molecule exists in an inactive, self-inhibited conformation stabilized by a binding network involving residues from both the N—SH2 and PTP domains. Stimulation by, for example, cytokines or growth factors acting through receptor tyrosine kinases (RTKs) leads to exposure of the catalytic site resulting in enzymatic activation of SHP2.
  • RTKs receptor tyrosine kinases
  • SHP2 is involved in signaling through the RAS-mitogen-activated protein kinase (MAPK), the JAK-STAT or the phosphoinositol 3-kinase-AKT pathways.
  • MAPK RAS-mitogen-activated protein kinase
  • JAK-STAT the JAK-STAT
  • phosphoinositol 3-kinase-AKT the phosphoinositol 3-kinase-AKT pathways.
  • Mutations in the PTPN11 gene and subsequently in SHP2 have been identified in several human developmental diseases, such as Noonan Syndrome and Leopard Syndrome, as well as human cancers, such as juvenile myelomonocytic leukemia, neuroblastoma, melanoma, acute myeloid leukemia and cancers of the breast, lung and colon. Some of these mutations destabilize the auto-inhibited conformation of SHP2 and promote autoactivation or enhanced growth factor driven activation of SHP2.
  • SHP2 therefore, represents a highly attractive target for the development of novel therapies for the treatment of various diseases including cancer.
  • a SHP2 inhibitor e.g., RMC-4550 or SHP099
  • a RAS pathway inhibitor e.g., a MEK inhibitor
  • combination therapy involving a SHP2 inhibitor with a RAS pathway inhibitor could be a general strategy for preventing tumor resistance in a wide range of malignancies, and may form the basis of a triple combination inhibitor with a SOS1 inhibitor.
  • Non-limiting examples of such SHP2 inhibitors include: Chen et al. Mol Pharmacol. 2006, 70, 562; Sarver et al., J. Med. Chem. 2017, 62, 1793; Xie et al., J. Med. Chem.
  • a SHP2 inhibitor binds in the active site.
  • a SHP2 inhibitor is a mixed-type irreversible inhibitor.
  • a SHP2 inhibitor binds an allosteric site e.g., a non-covalent allosteric inhibitor.
  • a SHP2 inhibitor is a covalent SHP2 inhibitor, such as an inhibitor that targets the cysteine residue (C333) that lies outside the phosphatase's active site.
  • a SHP2 inhibitor is a reversible inhibitor.
  • a SHP2 inhibitor is an irreversible inhibitor.
  • the SHP2 inhibitor is SHP099.
  • the SHP2 inhibitor is TNO155.
  • the SHP2 inhibitor is RMC-4550.
  • the SHP2 inhibitor is RCM-4630.
  • the SHP2 inhibitor is JAB-3068.
  • Proteasome inhibitors include, but are not limited to, carfilzomib (Kyprolis®), bortezomib (Velcade®), and oprozomib.
  • Immune therapies include, but are not limited to, monoclonal antibodies, immunomodulatory imides (IMiDs), GITR agonists, genetically engineered T-cells (e.g., CAR-T cells), bispecific antibodies (e.g., BiTEs), and anti-PD-1, anti-PDL-1, anti-CTLA4, anti-LAGI, and anti-OX40 agents).
  • IMDs immunomodulatory imides
  • GITR agonists e.g., CAR-T cells
  • bispecific antibodies e.g., BiTEs
  • anti-PD-1, anti-PDL-1, anti-CTLA4, anti-LAGI, and anti-OX40 agents include, but are not limited to, monoclonal antibodies, immunomodulatory imides (IMiDs), GITR agonists, genetically engineered T-cells (e.g., CAR-T cells), bispecific antibodies (e.g., BiTEs), and anti-PD-1, anti-PDL-1, anti-CTLA4, anti
  • Immunomodulatory agents are a class of immunomodulatory drugs (drugs that adjust immune responses) containing an imide group.
  • the IMiD class includes thalidomide and its analogues (lenalidomide, pomalidomide, and apremilast).
  • anti-PD-1 antibodies and methods for their use are described by Goldberg et al., Blood 2007, 110(1):186-192; Thompson et al., Clin. Cancer Res. 2007, 13(6):1757-1761; and WO06/121168 A1), as well as described elsewhere herein.
  • GITR agonists include, but are not limited to, GITR fusion proteins and anti-GITR antibodies (e.g., bivalent anti-GITR antibodies), such as, a GITR fusion protein described in U.S. Pat. Nos. 6,111,090, 8,586,023, WO2010/003118 and WO2011/090754; or an anti-GITR antibody described, e.g., in U.S. Pat. No. 7,025,962, EP 1947183, U.S. Pat. Nos.
  • anti-GITR antibodies e.g., bivalent anti-GITR antibodies
  • Anti-angiogenic agents are inclusive of, but not limited to, in vitro synthetically prepared chemical compositions, antibodies, antigen binding regions, radionuclides, and combinations and conjugates thereof.
  • An anti-angiogenic agent can be an agonist, antagonist, allosteric modulator, toxin or, more generally, may act to inhibit or stimulate its target (e.g., receptor or enzyme activation or inhibition), and thereby promote cell death or arrest cell growth.
  • the one or more additional therapies include an anti-angiogenic agent.
  • Anti-angiogenic agents can be MMP-2 (matrix-metalloproteinase 2) inhibitors, MMP-9 (matrix-metalloprotienase 9) inhibitors, and COX-II (cyclooxygenase 11) inhibitors.
  • Non-limiting examples of anti-angiogenic agents include rapamycin, temsirolimus (CCI-779), everolimus (RAD001), sorafenib, sunitinib, and bevacizumab.
  • Examples of useful COX-II inhibitors include alecoxib, valdecoxib, and rofecoxib.
  • WO96/33172 examples include WO96/27583, WO98/07697, WO98/03516, WO98/34918, WO98/34915, WO98/33768, WO98/30566, WO90/05719, WO99/52910, WO99/52889, WO99/29667, WO99007675, EP0606046, EP0780386, EP1786785, EP1181017, EP0818442, EP1004578, and US20090012085, and U.S. Pat. Nos. 5,863,949 and 5,861,510.
  • MMP-2 and MMP-9 inhibitors are those that have little or no activity inhibiting MMP-1. More preferred, are those that selectively inhibit MMP-2 or AMP-9 relative to the other matrix-metalloproteinases (i.e., MAP-1, MMP-3, MMP-4, MMP-5, MMP-6, MMP-7, MMP-8, MMP-10, MMP-11, MMP-12, and MMP-13).
  • MMP inhibitors are AG-3340, RO 32-3555, and RS 13-0830.
  • anti-angiogenic agents include KDR (kinase domain receptor) inhibitory agents (e.g., antibodies and antigen binding regions that specifically bind to the kinase domain receptor), anti-VEGF agents (e.g., antibodies or antigen binding regions that specifically bind VEGF, or soluble VEGF receptors or a ligand binding region thereof) such as VEGF-TRAPTM, and anti-VEGF receptor agents (e.g., antibodies or antigen binding regions that specifically bind thereto), EGFR inhibitory agents (e.g., antibodies or antigen binding regions that specifically bind thereto) such as Vectibix® (panitumumab), erlotinib (Tarceva®), anti-Angl and anti-Ang2 agents (e.g., antibodies or antigen binding regions specifically binding thereto or to their receptors, e.g., Tie2/Tek), and anti-Tie2 kinase inhibitory agents (e.g., antibodies or antigen binding regions that specifically bind to the
  • anti-angiogenic agents include Campath, IL-8, B-FGF, Tek antagonists (US2003/0162712; U.S. Pat. No. 6,413,932), anti-TWEAK agents (e.g., specifically binding antibodies or antigen binding regions, or soluble TWEAK receptor antagonists; see U.S. Pat. No. 6,727,225), ADAM distintegrin domain to antagonize the binding of integrin to its ligands (US 2002/0042368), specifically binding anti-eph receptor or anti-ephrin antibodies or antigen binding regions (U.S. Pat. Nos.
  • anti-PDGF-BB antagonists e.g., specifically binding antibodies or antigen binding regions
  • antibodies or antigen binding regions specifically binding to PDGF-BB ligands
  • PDGFR kinase inhibitory agents e.g., antibodies or antigen binding regions that specifically bind thereto
  • Additional anti-angiogenic agents include: SD-7784 (Pfizer, USA); cilengitide (Merck KGaA, Germany, EPO 0770622); pegaptanib octasodium, (Gilead Sciences, USA); Alphastatin, (BioActa, UK); M-PGA, (Celgene, USA, U.S. Pat. No. 5,712,291); ilomastat, (Arriva, USA, U.S. Pat. No. 5,892,112); emaxanib, (Pfizer, USA, U.S. Pat. No.
  • vatalanib (Novartis, Switzerland); 2-methoxyestradiol (EntreMed, USA); TLC ELL-12 (Elan, Ireland); anecortave acetate (Alcon, USA); alpha-D148 Mab (Amgen, USA); CEP-7055 (Cephalon, USA); anti-Vn Mab (Crucell, Netherlands), DACantiangiogenic (ConjuChem, Canada); Angiocidin (InKine Pharmaceutical, USA); KM-2550 (Kyowa Hakko, Japan); SU-0879 (Pfizer, USA); CGP-79787 (Novartis, Switzerland, EP 0970070); ARGENT technology (Ariad, USA); YIGSR-Stealth (Johnson & Johnson, USA); fibrinogen-E fragment (BioActa, UK); angiogenic inhibitor (Trigen, UK); TBC-1635 (Encysive Pharmaceuticals, USA); SC-236 (Pfizer, USA); ABT-567 (Abbott,
  • therapeutic agents that may be used in combination with compounds of the invention include agents (e.g., antibodies, antigen binding regions, or soluble receptors) that specifically bind and inhibit the activity of growth factors, such as antagonists of hepatocyte growth factor (HGF, also known as Scatter Factor), and antibodies or antigen binding regions that specifically bind its receptor, c-Met.
  • agents e.g., antibodies, antigen binding regions, or soluble receptors
  • HGF hepatocyte growth factor
  • Scatter Factor also known as Scatter Factor
  • Autophagy inhibitors include, but are not limited to chloroquine, 3-methyladenine, hydroxychloroquine (PlaquenilTM), bafilomycin A1, 5-amino-4-imidazole carboxamide riboside (AICAR), okadaic acid, autophagy-suppressive algal toxins which inhibit protein phosphatases of type 2A or type 1, analogues of cAMP, and drugs which elevate cAMP levels such as adenosine, LY204002, N6-mercaptopurine riboside, and vinblastine.
  • antisense or siRNA that inhibits expression of proteins including but not limited to ATG5 (which are implicated in autophagy), may also be used.
  • the one or more additional therapies include an autophagy inhibitor.
  • anti-neoplastic agent Another example of a therapeutic agent that may be used in combination with compounds of the invention is an anti-neoplastic agent.
  • the one or more additional therapies include an anti-neoplastic agent.
  • anti-neoplastic agents include acemannan, aclarubicin, aldesleukin, alemtuzumab, alitretinoin, altretamine, amifostine, aminolevulinic acid, amrubicin, amsacrine, anagrelide, anastrozole, ancer, ancestim, arglabin, arsenic trioxide, BAM-002 (Novelos), bexarotene, bicalutamide, broxuridine, capecitabine, celmoleukin, cetrorelix, cladribine, clotrimazole, cytarabine ocfosfate, DA 3030 (Dong-A), daclizumab, denileukin dif
  • therapeutic agents include ipilimumab (Yervoy@); tremelimumab; galiximab; nivolumab, also known as BMS-936558 (Opdivo®); pembrolizumab (Keytruda®); avelumab (Bavencio®); AMP224; BMS-936559; MPDL3280A, also known as RG7446; MEDI-570; AMG557; MGA271; IMP321; BMS-663513; PF-05082566; CDX-1127; anti-OX40 (Providence Health Services); huMAbOX40L; atacicept; CP-870893; lucatumumab; dacetuzumab; muromonab-CD3; ipilumumab; MEDI4736 (Imfinzi®); MSB0010718C; AMP 224; ad
  • an additional compound used in combination therapy with a compound of the present invention is selected from the group consisting of a CDK4/6 inhibitor (e.g., abemaciclib, palbociclib, or ribociclib), a KRAS:GDP G12C inhibitor (e.g., AMG 510, MRTX 1257) or other mutant Ras:GDP inhibitor, a KRAS:GTP G12C inhibitor or other mutant Ras:GTP inhibitor, a MEK inhibitor (e.g., refametinib, selumetinib, trametinib, or cobimetinib), a SHP2 inhibitor (e.g., TNO155, RMC-4630), an ERK inhibitor, and an RTK inhibitor (e.g., an EGFR inhibitor).
  • a CDK4/6 inhibitor e.g., abemaciclib, palbociclib, or ribociclib
  • KRAS:GDP G12C inhibitor e.g., AMG 510,
  • a SOS1 inhibitor may be used in combination with a Ras inhibitor, a SHP2 inhibitor, or a MK inhibitor.
  • a combination therapy includes a SOS1 inhibitor, a RAS inhibitor and a EK inhibitor.
  • an additional compound used in combination therapy with a compound of the present invention is selected from the group consisting of ABT-737, AT-7519, carfilzomib, cobimetinib, danusertib, dasatinib, doxorubicin, GSK-343, JQ1, MLN-7243, NVP-ADW742, paclitaxel, palbociclib and volasertib.
  • an additional compound used in combination therapy with a compound of the present invention is selected from the group consisting of neratinib, acetinib and reversine.
  • the compounds described herein can be used in combination with the agents disclosed herein or other suitable agents, depending on the condition being treated. Hence, in some embodiments the one or more compounds of the disclosure will be co-administered with other therapies as described herein.
  • the compounds described herein may be administered with the second agent simultaneously or separately.
  • This administration in combination can include simultaneous administration of the two agents in the same dosage form, simultaneous administration in separate dosage forms, and separate administration. That is, a compound described herein and any of the agents described herein can be formulated together in the same dosage form and administered simultaneously. Alternatively, a compound of the invention and any of the therapies described herein can be simultaneously administered, wherein both the agents are present in separate formulations.
  • a compound of the present disclosure can be administered and followed by any of the therapies described herein, or vice versa.
  • a compound of the invention and any of the therapies described herein are administered a few minutes apart, or a few hours apart, or a few days apart.
  • a combination therapeutic regimen employs two therapeutic agents, one compound of the present invention and a second selected from the therapeutic agents described herein. In some embodiments, a combination therapeutic regimen employs three therapeutic agents, one compound of the present invention and two selected from the therapeutic agents described herein. In some embodiments, a combination therapeutic regimen employs four or more therapeutic agents, one compound of the present invention and three selected from the therapeutic agents described herein.
  • the first therapy e.g., a compound of the invention
  • one or more additional therapies are administered simultaneously or sequentially, in either order.
  • the first therapeutic agent may be administered immediately, up to 1 hour, up to 2 hours, up to 3 hours, up to 4 hours, up to 5 hours, up to 6 hours, up to 7 hours, up to, 8 hours, up to 9 hours, up to 10 hours, up to 11 hours, up to 12 hours, up to 13 hours, 14 hours, up to hours 16, up to 17 hours, up 18 hours, up to 19 hours up to 20 hours, up to 21 hours, up to 22 hours, up to 23 hours, up to 24 hours, or up to 1-7, 1-14, 1-21 or 1-30 days before or after the one or more additional therapies.
  • kits including (a) a pharmaceutical composition including an agent (e.g., a compound of the invention) described herein, and (b) a package insert with instructions to perform any of the methods described herein.
  • the kit includes (a) a pharmaceutical composition including an agent (e.g., a compound of the invention) described herein, (b) one or more additional therapies (e.g., non-drug treatment or therapeutic agent), and (c) a package insert with instructions to perform any of the methods described herein.
  • kits may comprise two separate pharmaceutical compositions: a compound of the present invention, and one or more additional therapies.
  • the kit may comprise a container for containing the separate compositions such as a divided bottle or a divided foil packet. Additional examples of containers include syringes, boxes, and bags.
  • the kit may comprise directions for the use of the separate components.
  • the kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing health care professional.
  • Triethylamine (3.02 ml, 21.72 mmol), 2-[(Z)-[(4-methoxyphenyl)methylidene]amino]acetate (3.0 g, 14.48 mmol) and 4-chloro-6-(methylamino)pyrimidine-5-carbaldehyde (2.5 g, 14.50 mmol) were dissolved in MeOH (60 ml). The mixture was stirred for 48 h at room temperature. Acetic acid (4.14 ml, 72.40 mmol) was added and the mixture was stirred for 30 min at 50° C. After cooling to room temperature, the solvent was removed under reduced pressure. The residue was diluted with EtOAc, washed with water and dried with Na 2 SO 4 .
  • Example 22 Synthesis of 4-(((R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethyl)amino)-6-(1-imino-1-oxido-1,2,3,6-tetrahydro-1 ⁇ 6 -thiopyran-4-yl)-8-methylpyrido[2,3-d]pyrimidin-7(8H)-one
  • Example 23 Synthesis of 4-(((R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethyl)amino)-6-(1-imino-1-oxido-1,2,3,6-tetrahydro-1 ⁇ 6 -thiopyran-4-yl)-8-methylpyrido[2,3-d]pyrimidin-7(8H)-one
  • Example 23-1 shown in Table 7 was synthesized in the manner similar to Example 23.
  • Example 24 Synthesis of 4-(((R)-1-(3-(difluoromethyl)-2-fluorophenyl)ethyl)amino)-8-methyl-6-(1-(methylimino)-1-oxido-1,2,3,6-tetrahydro-1 ⁇ 6 -thiopyran-4-yl)pyrido[2,3-d]pyrimidin-7(8H)-one
  • Triethylsilane (114 ⁇ L, 712 ⁇ mol) was added and the mixture was stirred at 25° C. for 4 h. The mixture was concentrated under reduced pressure, partitioned between water and DCM, and neutralized by the addition of NaHCO 3 . The aqueous layer was extracted with DCM and the combined organic layers were dried over MgSO 4 , filtered, and concentrated under reduced pressure.
  • TEA 900 ⁇ L, 4.73 mmol
  • 4-hydroxy-2,8-dimethyl-6-(morpholine-4-carbonyl)-7H,8H-pyrido[2,3-d]pyrimidin-7-one 320 mg, 1.05 mmol
  • DMF 11.2 mL
  • 2,4,6-Triisopropylbenzenesulfonyl chloride 637 mg, 2.1 mmol
  • DMAP 13 mg, 0.11 mmol
  • Example 28 Synthesis of 4-(4- ⁇ [(1R)-1-[3-(difluoromethyl)-2-fluorophenyl]-ethyl]amino ⁇ -8-methyl-7-oxo-7H,8H-pyrido[2,3-d]pyrimidin-6-yl)-4-hydroxy-1 ⁇ 6 -thiane-1,1-dione
  • Example 29 Synthesis of 4-(4- ⁇ [(1R)-1-[3-(difluoromethyl)-2-fluorophenyl]ethyl]amino ⁇ -8-methyl-7-oxo-7H,8H-pyrido[2,3-d]-pyrimidin-6-yl)-4-fluoro-1 ⁇ 6 -thiane-1,1-dione
  • Example 32 Synthesis of 4-(4- ⁇ [(1R)-1-[3-(difluoro-methyl)-2-fluorophenyl]ethyl]amino ⁇ -8-methyl-7-oxo-7H,8H-pyrido[2,3-d]pyrimidin-6-yl)-4-methoxy-1 ⁇ 6 -thiane-1,1-dione
  • Example 37 Synthesis of 4-(4- ⁇ [(1R)-1-[3-(difluoromethyl)-2-fluorophenyl]ethyl]amino ⁇ -8-methyl-7-oxo-7H,8H-pyrido[2,3-d]pyrimidin-6-yl)-4-methyl-1 ⁇ 6 -thiane-1,1-dione
  • Example 40 Synthesis of 2- ⁇ 3-[(1R)-1- ⁇ [6-(1,1-dioxo-3,6-dihydro-2H-1 ⁇ 6 -thiopyran-4-yl)-8-methyl-7-oxo-7H,8H-pyrido[2,3-d]pyrimidin-4-yl]amino ⁇ ethyl]phenyl ⁇ -2,2-difluoroacetonitrile
  • Example 41 Synthesis of 2- ⁇ 3-[(1R)-1- ⁇ [6-(1,1-dioxo-3,6-dihydro-2H-1 ⁇ 6 -thiopyran-4-yl)-8-methyl-7-oxo-7H,8H-pyrido[2,3-d]pyrimidin-4-yl]amino ⁇ ethyl]-2-fluorophenyl ⁇ -2,2-difluoroacetonitrile
  • Example 41-1 shown in Table 15 was synthesized in the manner similar to Example 41.
  • Example 42 Synthesis of 4-(4- ⁇ [(1R)-1-[3-(2-amino-1,1-difluoroethyl)-2-fluorophenyl]ethyl]amino ⁇ -8-methyl-7-oxo-7H,8H-pyrido[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-1 ⁇ 6 -thiopyran-1,1-dione

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pyridine Compounds (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
US17/090,342 2019-11-08 2020-11-05 Bicyclic heteroaryl compounds and uses thereof Abandoned US20210139517A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US17/090,342 US20210139517A1 (en) 2019-11-08 2020-11-05 Bicyclic heteroaryl compounds and uses thereof
US17/133,340 US11168102B1 (en) 2019-11-08 2020-12-23 Bicyclic heteroaryl compounds and uses thereof
US18/045,647 US20230365605A1 (en) 2019-11-08 2022-10-11 Bicyclic heteroaryl compounds and uses thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201962933141P 2019-11-08 2019-11-08
US202063031318P 2020-05-28 2020-05-28
US202063070593P 2020-08-26 2020-08-26
US17/090,342 US20210139517A1 (en) 2019-11-08 2020-11-05 Bicyclic heteroaryl compounds and uses thereof

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US17/133,340 Continuation US11168102B1 (en) 2019-11-08 2020-12-23 Bicyclic heteroaryl compounds and uses thereof
US18/045,647 Continuation US20230365605A1 (en) 2019-11-08 2022-10-11 Bicyclic heteroaryl compounds and uses thereof

Publications (1)

Publication Number Publication Date
US20210139517A1 true US20210139517A1 (en) 2021-05-13

Family

ID=73646489

Family Applications (3)

Application Number Title Priority Date Filing Date
US17/090,342 Abandoned US20210139517A1 (en) 2019-11-08 2020-11-05 Bicyclic heteroaryl compounds and uses thereof
US17/133,340 Active US11168102B1 (en) 2019-11-08 2020-12-23 Bicyclic heteroaryl compounds and uses thereof
US18/045,647 Pending US20230365605A1 (en) 2019-11-08 2022-10-11 Bicyclic heteroaryl compounds and uses thereof

Family Applications After (2)

Application Number Title Priority Date Filing Date
US17/133,340 Active US11168102B1 (en) 2019-11-08 2020-12-23 Bicyclic heteroaryl compounds and uses thereof
US18/045,647 Pending US20230365605A1 (en) 2019-11-08 2022-10-11 Bicyclic heteroaryl compounds and uses thereof

Country Status (16)

Country Link
US (3) US20210139517A1 (es)
EP (1) EP4055017A1 (es)
JP (1) JP2023500328A (es)
KR (1) KR20220100903A (es)
CN (2) CN114901662A (es)
AU (1) AU2020380315A1 (es)
BR (1) BR112022008858A2 (es)
CA (1) CA3156359A1 (es)
CL (1) CL2023000416A1 (es)
CO (1) CO2022005968A2 (es)
CR (1) CR20220200A (es)
IL (1) IL292679A (es)
MX (1) MX2022005525A (es)
PE (1) PE20230249A1 (es)
TW (1) TW202128688A (es)
WO (1) WO2021092115A1 (es)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022212546A1 (en) * 2021-03-31 2022-10-06 Acerand Therapeutics (Usa) Limited Pyridopyrimidinone compounds
WO2022240947A1 (en) * 2021-05-12 2022-11-17 Revolution Medicines, Inc. Use of sos1 inhibitors with mtor inhibitors to treat cancers
WO2023059597A1 (en) * 2021-10-05 2023-04-13 Mirati Therapeutics, Inc. Combination therapies of kras g12d inhibitors with sos1 inhibitors
US20230339952A1 (en) * 2022-04-20 2023-10-26 Kumquat Biosciences Inc. Macrocyclic heterocycles and uses thereof
WO2023215256A1 (en) * 2022-05-03 2023-11-09 Revolution Medicines, Inc. Sos1 inhibitors and uses thereof
WO2023215257A3 (en) * 2022-05-03 2024-04-04 Revolution Medicines, Inc. Sos1 inhibitors and uses thereof

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114901662A (zh) * 2019-11-08 2022-08-12 锐新医药公司 双环杂芳基化合物及其用途
CN115605479B (zh) * 2020-06-11 2024-05-14 江苏恒瑞医药股份有限公司 吡啶酮并嘧啶类衍生物、其制备方法及其在医药上的应用
CN113801114B (zh) * 2020-06-11 2022-11-18 江苏恒瑞医药股份有限公司 稠合二环杂芳基类衍生物、其制备方法及其在医药上的应用
EP4208261A1 (en) 2020-09-03 2023-07-12 Revolution Medicines, Inc. Use of sos1 inhibitors to treat malignancies with shp2 mutations
EP4214209A1 (en) 2020-09-15 2023-07-26 Revolution Medicines, Inc. Indole derivatives as ras inhibitors in the treatment of cancer
WO2022135590A1 (zh) * 2020-12-27 2022-06-30 上海凌达生物医药有限公司 一类嘧啶并杂环类化合物、制备方法和用途
CN116669738A (zh) * 2021-02-09 2023-08-29 苏州阿尔脉生物科技有限公司 一种作为sos1抑制剂的嘧啶并吡啶酮类衍生物、其制备方法及用途
WO2022171018A1 (zh) * 2021-02-09 2022-08-18 苏州泽璟生物制药股份有限公司 取代苯并或吡啶并嘧啶胺类抑制剂及其制备方法和应用
US11648254B2 (en) 2021-03-02 2023-05-16 Kumquat Biosciences Inc. Substituted pyrido[2,3-d]pyrimidines as inhibitors of Ras pathway signaling
JP2024514127A (ja) 2021-04-09 2024-03-28 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング 抗がん剤療法
CN117986265A (zh) * 2021-09-14 2024-05-07 北京福元医药股份有限公司 苄氨基取代的嘧啶并吡喃酮衍生物及其组合物、制剂和用途
WO2023051628A1 (zh) 2021-09-29 2023-04-06 上海海和药物研究开发股份有限公司 一类具有吡啶并六元环结构的sos1抑制剂
AR127308A1 (es) 2021-10-08 2024-01-10 Revolution Medicines Inc Inhibidores ras
WO2023114954A1 (en) 2021-12-17 2023-06-22 Genzyme Corporation Pyrazolopyrazine compounds as shp2 inhibitors
WO2023118250A1 (en) * 2021-12-23 2023-06-29 Boehringer Ingelheim International Gmbh 8-aza quinazolines as brain-penetrant sos1-inhibitors
WO2023135260A1 (en) 2022-01-14 2023-07-20 Jazz Pharmaceuticals Ireland Limited Novel amine-substituted phthalazines and derivatives as sos1 inhibitors
EP4227307A1 (en) 2022-02-11 2023-08-16 Genzyme Corporation Pyrazolopyrazine compounds as shp2 inhibitors
GB202203976D0 (en) 2022-03-22 2022-05-04 Jazz Pharmaceuticals Ireland Ltd Tricyclic phthalazines and derivatives as sos1 inhibitors
WO2023240263A1 (en) 2022-06-10 2023-12-14 Revolution Medicines, Inc. Macrocyclic ras inhibitors
WO2024040131A1 (en) 2022-08-17 2024-02-22 Treeline Biosciences, Inc. Pyridopyrimidine kras inhibitors

Family Cites Families (236)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6534055B1 (en) 1988-11-23 2003-03-18 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US6352694B1 (en) 1994-06-03 2002-03-05 Genetics Institute, Inc. Methods for inducing a population of T cells to proliferate using agents which recognize TCR/CD3 and ligands which stimulate an accessory molecule on the surface of the T cells
US6905680B2 (en) 1988-11-23 2005-06-14 Genetics Institute, Inc. Methods of treating HIV infected subjects
US5858358A (en) 1992-04-07 1999-01-12 The United States Of America As Represented By The Secretary Of The Navy Methods for selectively stimulating proliferation of T cells
GB8827305D0 (en) 1988-11-23 1988-12-29 British Bio Technology Compounds
JP2762522B2 (ja) 1989-03-06 1998-06-04 藤沢薬品工業株式会社 血管新生阻害剤
PT98990A (pt) 1990-09-19 1992-08-31 American Home Prod Processo para a preparacao de esteres de acidos carboxilicos de rapamicina
US5892112A (en) 1990-11-21 1999-04-06 Glycomed Incorporated Process for preparing synthetic matrix metalloprotease inhibitors
US5120842A (en) 1991-04-01 1992-06-09 American Home Products Corporation Silyl ethers of rapamycin
US5100883A (en) 1991-04-08 1992-03-31 American Home Products Corporation Fluorinated esters of rapamycin
US5118678A (en) 1991-04-17 1992-06-02 American Home Products Corporation Carbamates of rapamycin
WO1992020642A1 (en) 1991-05-10 1992-11-26 Rhone-Poulenc Rorer International (Holdings) Inc. Bis mono-and bicyclic aryl and heteroaryl compounds which inhibit egf and/or pdgf receptor tyrosine kinase
US5118677A (en) 1991-05-20 1992-06-02 American Home Products Corporation Amide esters of rapamycin
NZ243082A (en) 1991-06-28 1995-02-24 Ici Plc 4-anilino-quinazoline derivatives; pharmaceutical compositions, preparatory processes, and use thereof
US5151413A (en) 1991-11-06 1992-09-29 American Home Products Corporation Rapamycin acetals as immunosuppressant and antifungal agents
GB9125660D0 (en) 1991-12-03 1992-01-29 Smithkline Beecham Plc Novel compound
AU661533B2 (en) 1992-01-20 1995-07-27 Astrazeneca Ab Quinazoline derivatives
US5521184A (en) 1992-04-03 1996-05-28 Ciba-Geigy Corporation Pyrimidine derivatives and processes for the preparation thereof
DE4221054A1 (de) 1992-06-30 1994-01-05 Herbst Bremer Goldschlaegerei Präparat zur prophylaktischen und therapeutischen Behandlung von Karies sowie Verfahren zum Herstellen desselben
ZA935111B (en) 1992-07-17 1994-02-04 Smithkline Beecham Corp Rapamycin derivatives
ZA935112B (en) 1992-07-17 1994-02-08 Smithkline Beecham Corp Rapamycin derivatives
US5256790A (en) 1992-08-13 1993-10-26 American Home Products Corporation 27-hydroxyrapamycin and derivatives thereof
GB9221220D0 (en) 1992-10-09 1992-11-25 Sandoz Ag Organic componds
US5262564A (en) 1992-10-30 1993-11-16 Octamer, Inc. Sulfinic acid adducts of organo nitroso compounds useful as retroviral inactivating agents anti-retroviral agents and anti-tumor agents
US5258389A (en) 1992-11-09 1993-11-02 Merck & Co., Inc. O-aryl, O-alkyl, O-alkenyl and O-alkynylrapamycin derivatives
NZ258697A (en) 1992-11-13 1996-03-26 Immunex Corp Elk-l polypeptide, coding sequence and vector therefor
US5455258A (en) 1993-01-06 1995-10-03 Ciba-Geigy Corporation Arylsulfonamido-substituted hydroxamic acids
US5629327A (en) 1993-03-01 1997-05-13 Childrens Hospital Medical Center Corp. Methods and compositions for inhibition of angiogenesis
US5516658A (en) 1993-08-20 1996-05-14 Immunex Corporation DNA encoding cytokines that bind the cell surface receptor hek
US5612340A (en) 1993-10-01 1997-03-18 Ciba-Geigy Corporation Pyrimidineamine derivatives and processes for the preparation thereof
US5656643A (en) 1993-11-08 1997-08-12 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono-and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
CA2175215C (en) 1993-11-19 2008-06-03 Yat Sun Or Semisynthetic analogs of rapamycin (macrolides) being immunomodulators
SG64372A1 (en) 1993-12-17 1999-04-27 Novartis Ag Rapamycin derivatives
US5700823A (en) 1994-01-07 1997-12-23 Sugen, Inc. Treatment of platelet derived growth factor related disorders such as cancers
IL112248A0 (en) 1994-01-25 1995-03-30 Warner Lambert Co Tricyclic heteroaromatic compounds and pharmaceutical compositions containing them
IL112249A (en) 1994-01-25 2001-11-25 Warner Lambert Co Pharmaceutical compositions containing di and tricyclic pyrimidine derivatives for inhibiting tyrosine kinases of the epidermal growth factor receptor family and some new such compounds
WO1995024190A2 (en) 1994-03-07 1995-09-14 Sugen, Inc. Receptor tyrosine kinase inhibitors for inhibiting cell proliferative disorders and compositions thereof
EP1464706A3 (en) 1994-04-15 2004-11-03 Amgen Inc., HEK5, HEK7, HEK8, HEK11, EPH-like receptor protein tyrosine kinases
ATE159257T1 (de) 1994-05-03 1997-11-15 Ciba Geigy Ag Pyrrolopyrimidinderivate mit antiproliferativer wirkung
US7175843B2 (en) 1994-06-03 2007-02-13 Genetics Institute, Llc Methods for selectively stimulating proliferation of T cells
US6303769B1 (en) 1994-07-08 2001-10-16 Immunex Corporation Lerk-5 dna
US5919905A (en) 1994-10-05 1999-07-06 Immunex Corporation Cytokine designated LERK-6
US6057124A (en) 1995-01-27 2000-05-02 Amgen Inc. Nucleic acids encoding ligands for HEK4 receptors
US5863949A (en) 1995-03-08 1999-01-26 Pfizer Inc Arylsulfonylamino hydroxamic acid derivatives
EP1110953B1 (en) 1995-03-30 2009-10-28 Pfizer Products Inc. Quinazoline derivatives
US5981533A (en) 1995-04-03 1999-11-09 Novartis Ag Pyrazole derivatives and processes for the preparation thereof
EP0821671B1 (en) 1995-04-20 2000-12-27 Pfizer Inc. Arylsulfonyl hydroxamic acid derivatives as mmp and tnf inhibitors
GB9508538D0 (en) 1995-04-27 1995-06-14 Zeneca Ltd Quinazoline derivatives
US6692964B1 (en) 1995-05-04 2004-02-17 The United States Of America As Represented By The Secretary Of The Navy Methods for transfecting T cells
US7067318B2 (en) 1995-06-07 2006-06-27 The Regents Of The University Of Michigan Methods for transfecting T cells
US5747498A (en) 1996-05-28 1998-05-05 Pfizer Inc. Alkynyl and azido-substituted 4-anilinoquinazolines
US5650415A (en) 1995-06-07 1997-07-22 Sugen, Inc. Quinoline compounds
US5880141A (en) 1995-06-07 1999-03-09 Sugen, Inc. Benzylidene-Z-indoline compounds for the treatment of disease
HU228234B1 (en) 1995-06-09 2013-02-28 Novartis Ag Rapamycin derivatives, pharmaceutical compositions containing them and their use
DE69619114T2 (de) 1995-07-06 2002-10-02 Novartis Ag Pyrolopyrimidine und verfahren zu ihrer herstellung
AR004010A1 (es) 1995-10-11 1998-09-30 Glaxo Group Ltd Compuestos heterociclicos
GB9523675D0 (en) 1995-11-20 1996-01-24 Celltech Therapeutics Ltd Chemical compounds
DK0780386T3 (da) 1995-12-20 2003-02-03 Hoffmann La Roche Matrixmetalloproteaseinhibitorer
PT888349E (pt) 1996-01-23 2002-10-31 Novartis Ag Pirrolopirimidinas e processos para a sua preparacao
JP3406763B2 (ja) 1996-01-30 2003-05-12 東レ・ダウコーニング・シリコーン株式会社 シリコーンゴム組成物
GB9603095D0 (en) 1996-02-14 1996-04-10 Zeneca Ltd Quinazoline derivatives
GB9603097D0 (en) 1996-02-14 1996-04-10 Zeneca Ltd Quinazoline compounds
DE19629652A1 (de) 1996-03-06 1998-01-29 Thomae Gmbh Dr K 4-Amino-pyrimidin-Derivate, diese Verbindungen enthaltende Arzneimittel, deren Verwendung und Verfahren zu ihrer Herstellung
DE19608588A1 (de) 1996-03-06 1997-09-11 Thomae Gmbh Dr K Pyrimido [5,4-d]pyrimidine, diese Verbindungen enthaltende Arzneimittel, deren Verwendung und Verfahren zu ihrer Herstellung
KR20000064601A (ko) 1996-03-15 2000-11-06 한스 루돌프 하우스, 헨리테 브룬너, 베아트리체 귄터 신규의n-7-헤테로시클릴피롤로[2,3-d]피리딘및�그의용도
EP0892789B2 (en) 1996-04-12 2009-11-18 Warner-Lambert Company LLC Irreversible inhibitors of tyrosine kinases
GB9607729D0 (en) 1996-04-13 1996-06-19 Zeneca Ltd Quinazoline derivatives
EP0907642B1 (en) 1996-06-24 2005-11-02 Pfizer Inc. Phenylamino-substituted tricyclic derivatives for treatment of hyperproliferative diseases
WO1998002441A2 (en) 1996-07-12 1998-01-22 Ariad Pharmaceuticals, Inc. Non immunosuppressive antifungal rapalogs
EP0818442A3 (en) 1996-07-12 1998-12-30 Pfizer Inc. Cyclic sulphone derivatives as inhibitors of metalloproteinases and of the production of tumour necrosis factor
HRP970371A2 (en) 1996-07-13 1998-08-31 Kathryn Jane Smith Heterocyclic compounds
DE69718472T2 (de) 1996-07-13 2003-11-06 Glaxo Group Ltd Bicyclische heteroaromatische verbindungen als protein tyrosine kinase inhibitoren
AR007857A1 (es) 1996-07-13 1999-11-24 Glaxo Group Ltd Compuestos heterociclicos fusionados como inhibidores de proteina tirosina quinasa, sus metodos de preparacion, intermediarios uso en medicina ycomposiciones farmaceuticas que los contienen.
EP0923585B1 (en) 1996-07-18 2002-05-08 Pfizer Inc. Phosphinate based inhibitors of matrix metalloproteases
US6111090A (en) 1996-08-16 2000-08-29 Schering Corporation Mammalian cell surface antigens; related reagents
JP4440344B2 (ja) 1996-08-16 2010-03-24 シェーリング コーポレイション 哺乳動物細胞表面抗原;関連試薬
JP2000501423A (ja) 1996-08-23 2000-02-08 ファイザー インク. アリールスルホニルアミノヒドロキサム酸誘導体
AU720429B2 (en) 1996-08-23 2000-06-01 Novartis Ag Substituted pyrrolopyrimidines and processes for their preparation
ID18494A (id) 1996-10-02 1998-04-16 Novartis Ag Turunan pirazola leburan dan proses pembuatannya
JP4205168B2 (ja) 1996-10-02 2009-01-07 ノバルティス アクチエンゲゼルシヤフト ピリミジン誘導体およびその製造法
WO1998014449A1 (en) 1996-10-02 1998-04-09 Novartis Ag Fused pyrazole derivatives and processes for their preparation
EP0837063A1 (en) 1996-10-17 1998-04-22 Pfizer Inc. 4-Aminoquinazoline derivatives
GB9621757D0 (en) 1996-10-18 1996-12-11 Ciba Geigy Ag Phenyl-substituted bicyclic heterocyclyl derivatives and their use
PT950059E (pt) 1997-01-06 2004-10-29 Pfizer Derivados de sulfona ciclicos
ID22799A (id) 1997-02-03 1999-12-09 Pfizer Prod Inc Turunan-turunan asam arilsulfonilamino hidroksamat
EP0964864B1 (en) 1997-02-05 2008-04-09 Warner-Lambert Company LLC Pyrido 2,3-d pyrimidines and 4-aminopyrimidines as inhibitors of cellular proliferation
JP2000507975A (ja) 1997-02-07 2000-06-27 ファイザー・インク N−ヒドロキシ−β−スルホニルプロピオンアミド誘導体類及びそれらのマトリックスメタロプロテイナーゼ阻害薬としての使用
IL131123A0 (en) 1997-02-11 2001-01-28 Pfizer Arylsulfonyl hydroxamic acid derivatives
CO4950519A1 (es) 1997-02-13 2000-09-01 Novartis Ag Ftalazinas, preparaciones farmaceuticas que las comprenden y proceso para su preparacion
US6150395A (en) 1997-05-30 2000-11-21 The Regents Of The University Of California Indole-3-carbinol (I3C) derivatives and methods
WO1999007701A1 (en) 1997-08-05 1999-02-18 Sugen, Inc. Tricyclic quinoxaline derivatives as protein tyrosine kinase inhibitors
US6214872B1 (en) 1997-08-08 2001-04-10 Pfizer Inc. Aryloxyarylsulfonylamino hydroxamic acid derivatives
WO2000012089A1 (en) 1998-08-31 2000-03-09 Merck & Co., Inc. Novel angiogenesis inhibitors
AU1102399A (en) 1997-10-21 1999-05-10 Human Genome Sciences, Inc. Human tumor necrosis factor receptor-like proteins tr11, tr11sv1, and tr11sv2
GB9725782D0 (en) 1997-12-05 1998-02-04 Pfizer Ltd Therapeutic agents
GB9800569D0 (en) 1998-01-12 1998-03-11 Glaxo Group Ltd Heterocyclic compounds
GB9800575D0 (en) 1998-01-12 1998-03-11 Glaxo Group Ltd Heterocyclic compounds
WO1999040196A1 (en) 1998-02-09 1999-08-12 Genentech, Inc. Novel tumor necrosis factor receptor homolog and nucleic acids encoding the same
WO1999045009A1 (en) 1998-03-04 1999-09-10 Bristol-Myers Squibb Company Heterocyclo-substituted imidazopyrazine protein tyrosine kinase inhibitors
PA8469501A1 (es) 1998-04-10 2000-09-29 Pfizer Prod Inc Hidroxamidas del acido (4-arilsulfonilamino)-tetrahidropiran-4-carboxilico
PA8469401A1 (es) 1998-04-10 2000-05-24 Pfizer Prod Inc Derivados biciclicos del acido hidroxamico
DK2020408T3 (da) 1998-05-29 2013-09-30 Sugen Inc Pyrrol-substitueret 2-indolinon som proteinkinaseinhibitor
UA60365C2 (uk) 1998-06-04 2003-10-15 Пфайзер Продактс Інк. Похідні ізотіазолу, спосіб їх одержання, фармацевтична композиція та спосіб лікування гіперпроліферативного захворювання у ссавця
JP2002520324A (ja) 1998-07-10 2002-07-09 メルク エンド カムパニー インコーポレーテッド 新規な血管形成インヒビター
US6114361A (en) 1998-11-05 2000-09-05 Pfizer Inc. 5-oxo-pyrrolidine-2-carboxylic acid hydroxamide derivatives
DE60028740T2 (de) 1999-03-30 2007-05-24 Novartis Ag Phthalazinderivate zur behandlung von entzündlichen erkrankungen
GB9912961D0 (en) 1999-06-03 1999-08-04 Pfizer Ltd Metalloprotease inhibitors
US6521424B2 (en) 1999-06-07 2003-02-18 Immunex Corporation Recombinant expression of Tek antagonists
JP4587626B2 (ja) 1999-06-07 2010-11-24 イミュネックス・コーポレーション Tekアンタゴニスト
JP2003504343A (ja) 1999-07-12 2003-02-04 ジェネンテック・インコーポレーテッド 腫瘍壊死因子リガンド/レセプター相同体による血管形成及び心血管形成の促進と抑制
EP1212331B1 (en) 1999-08-24 2004-04-21 Ariad Gene Therapeutics, Inc. 28-epirapalogs
UA72946C2 (uk) 1999-11-05 2005-05-16 Астразенека Аб Похідні хіназоліну як інгібітори васкулярного ендотеліального фактора росту (vegf)
WO2001037820A2 (en) 1999-11-24 2001-05-31 Sugen, Inc. Ionizable indolinone derivatives and their use as ptk ligands
US6515004B1 (en) 1999-12-15 2003-02-04 Bristol-Myers Squibb Company N-[5-[[[5-alkyl-2-oxazolyl]methyl]thio]-2-thiazolyl]-carboxamide inhibitors of cyclin dependent kinases
US6727225B2 (en) 1999-12-20 2004-04-27 Immunex Corporation TWEAK receptor
US6867041B2 (en) 2000-02-24 2005-03-15 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US7572631B2 (en) 2000-02-24 2009-08-11 Invitrogen Corporation Activation and expansion of T cells
AU2001243288B2 (en) 2000-02-24 2005-11-24 Life Technologies Corporation Simultaneous stimulation and concentration of cells
US6797514B2 (en) 2000-02-24 2004-09-28 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
JP2003523768A (ja) 2000-02-25 2003-08-12 イミュネックス・コーポレーション インテグリンアンタゴニスト
US6630500B2 (en) 2000-08-25 2003-10-07 Cephalon, Inc. Selected fused pyrrolocarbazoles
SI1343782T1 (sl) 2000-12-21 2009-10-31 Smithkline Beecham Corp Pirimidinamini kot modulatorji angiogeneze
US7102009B2 (en) 2001-01-12 2006-09-05 Amgen Inc. Substituted amine derivatives and methods of use
US7105682B2 (en) 2001-01-12 2006-09-12 Amgen Inc. Substituted amine derivatives and methods of use
US20020147198A1 (en) 2001-01-12 2002-10-10 Guoqing Chen Substituted arylamine derivatives and methods of use
US6878714B2 (en) 2001-01-12 2005-04-12 Amgen Inc. Substituted alkylamine derivatives and methods of use
US6995162B2 (en) 2001-01-12 2006-02-07 Amgen Inc. Substituted alkylamine derivatives and methods of use
US7307088B2 (en) 2002-07-09 2007-12-11 Amgen Inc. Substituted anthranilic amide derivatives and methods of use
TWI329112B (en) 2002-07-19 2010-08-21 Bristol Myers Squibb Co Novel inhibitors of kinases
WO2004107618A2 (en) 2003-05-23 2004-12-09 Wyeth Gitr ligand and gitr ligand-related molecules and antibodies and uses thereof
EP1646634B1 (en) 2003-07-08 2008-11-12 Novartis AG Use of rapamycin and rapamycin derivatives for the treatment of bone loss
EP1660126A1 (en) 2003-07-11 2006-05-31 Schering Corporation Agonists or antagonists of the clucocorticoid-induced tumour necrosis factor receptor (gitr) or its ligand for the treatment of immune disorders, infections and cancer
WO2005016252A2 (en) 2003-07-11 2005-02-24 Ariad Gene Therapeutics, Inc. Phosphorus-containing macrocycles
TW200523262A (en) 2003-07-29 2005-07-16 Smithkline Beecham Corp Inhibitors of AKT activity
PT2287156E (pt) 2003-08-15 2013-08-26 Novartis Ag 2,4-di(fenilamino)-pirimidinas úteis no tratamento de doenças neoplásicas, desordens inflamatórias e do sistema imunitário
WO2005055808A2 (en) 2003-12-02 2005-06-23 Genzyme Corporation Compositions and methods to diagnose and treat lung cancer
US20080058431A1 (en) 2004-03-26 2008-03-06 Gen-Sheng Feng Modulators of Shp2 Tyrosine Phosphatase and Their Use in the Treatment of Body Weight Disorders
GB0409799D0 (en) 2004-04-30 2004-06-09 Isis Innovation Method of generating improved immune response
EP1765402A2 (en) 2004-06-04 2007-03-28 Duke University Methods and compositions for enhancement of immunity by in vivo depletion of immunosuppressive cell activity
WO2006021884A2 (en) 2004-08-26 2006-03-02 Pfizer Inc. Enantiomerically pure aminoheteroaryl compounds as protein kinase inhibitors
PE20060608A1 (es) 2004-10-13 2006-08-22 Wyeth Corp Analogos de 17-hidroxiwortmanina como inhibidores de pi3k
ES2657443T3 (es) 2005-03-25 2018-03-05 Gitr, Inc. Anticuerpos anti-GITR y usos de los mismos
US7745451B2 (en) 2005-05-04 2010-06-29 Renovis, Inc. Tetrahydronaphthyridine and tetrahydropyrido[4,3-d]pyrimidine compounds and compositions thereof useful in the treatment of conditions associated with neurological and inflammatory disorders and dysfunctions
SI2439273T1 (sl) 2005-05-09 2019-05-31 Ono Pharmaceutical Co., Ltd. Človeška monoklonska protitelesa za programirano smrt 1 (PD-1) in postopki za zdravljenje raka z uporabo protiteles proti PD-1 samostojno ali v kombinaciji z ostalimi imunoterapevtiki
GB0510390D0 (en) 2005-05-20 2005-06-29 Novartis Ag Organic compounds
CA2622870A1 (en) 2005-09-20 2007-03-29 Pfizer Products Inc. Dosage forms and methods of treatment using a tyrosine kinase inhibitor
EP1981969A4 (en) 2006-01-19 2009-06-03 Genzyme Corp ANTI-GITRANT ANTIBODIES FOR THE TREATMENT OF CANCER
US8987474B2 (en) 2006-04-07 2015-03-24 University Of South Florida Inhibition of Shp2/PTPN11 protein tyrosine phosphatase by NSC-87877, NSC-117199 and their analogs
JP5284977B2 (ja) 2006-12-07 2013-09-11 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト ホスホイノシチド3−キナーゼ阻害剤化合物及び使用方法
WO2008124815A1 (en) 2007-04-10 2008-10-16 University Of South Florida Method of activating nk cells
EP3124046B1 (en) 2007-07-12 2019-12-25 GITR, Inc. Combination therapies employing gitr binding molecules
BRPI0816769A2 (pt) 2007-09-12 2016-11-29 Hoffmann La Roche combinações de compostos inibidores de fosfoinositida 3-cinase e agentes quimioterapêuticos, e métodos de uso
WO2009049098A2 (en) 2007-10-09 2009-04-16 Indiana University Research & Technology Corporation Materials and methods for regulating the activity of phosphatases
WO2009055730A1 (en) 2007-10-25 2009-04-30 Genentech, Inc. Process for making thienopyrimidine compounds
WO2009135000A2 (en) 2008-04-30 2009-11-05 H. Lee Moffitt Cancer Center And Research Institute, Inc. Inhibition of shp2/ptpn11 protein tyrosine phosphatase by nsc-117199 and analogs
AU2009266873A1 (en) 2008-07-02 2010-01-07 Emergent Product Development Seattle, Llc TGF-beta antagonist multi-target binding proteins
US9174969B2 (en) 2008-07-21 2015-11-03 University Of South Florida Indoline scaffold SHP-2 inhibitors and cancer treatment method
WO2010030002A1 (ja) 2008-09-12 2010-03-18 国立大学法人三重大学 外来性gitrリガンド発現細胞
WO2010121212A2 (en) 2009-04-17 2010-10-21 H. Lee Moffit Cancer Center And Research Institute, Inc. Indoline scaffold shp-2 inhibitors and method of treating cancer
AU2010284255B2 (en) 2009-08-17 2016-11-17 Memorial Sloan-Kettering Cancer Center Heat shock protein binding compounds, compositions, and methods for making and using same
EP2473531A4 (en) 2009-09-03 2013-05-01 Merck Sharp & Dohme ANTI-GITRANT ANTIBODIES
GB0919054D0 (en) 2009-10-30 2009-12-16 Isis Innovation Treatment of obesity
EP3112382A1 (en) 2009-12-29 2017-01-04 Emergent Product Development Seattle, LLC Heterodimer binding proteins and uses thereof
US8637684B2 (en) 2010-05-12 2014-01-28 Wisconsin Alumni Research Foundation Tautomycetin and tautomycetin analog biosynthesis
WO2012041524A1 (en) 2010-10-01 2012-04-05 Max-Delbrück-Centrum Für Molekulare Medizin (Mdc) Hydrazonopyrazolones as protein tyrosine phosphatase inhibitors
EP2655374B1 (en) 2010-12-20 2019-10-23 Incyte Holdings Corporation N-(1-(substituted-phenyl)ethyl)-9h-purin-6-amines as pi3k inhibitors
PT2688887E (pt) 2011-03-23 2015-07-06 Amgen Inc Inibidores duais tricíclicos fusionados de cdk 4/6 e flt3
WO2013039954A1 (en) 2011-09-14 2013-03-21 Sanofi Anti-gitr antibodies
ES2667726T3 (es) 2012-07-20 2018-05-14 Cleave Biosciences, Inc. Pirimidinas condensadas como inhibidores del complejo p97
WO2014113584A1 (en) 2013-01-16 2014-07-24 Rhode Island Hospital Compositions and methods for the prevention and treatment of osteolysis and osteoporosis
US9498543B2 (en) 2013-03-15 2016-11-22 Novartis Ag Antibody drug conjugates
AU2014256984B2 (en) 2013-04-26 2019-02-14 Indiana University Research & Technology Corporation Hydroxyindole carboxylic acid based inhibitors for oncogenic Src homology-2 domain containing protein tyrosine phosphatase-2 (SHP2)
US9844535B2 (en) 2013-07-03 2017-12-19 Indiana University Research And Technology Corporation SHP2 inhibitors and methods of treating autoimmune and/or glomerulonephritis-associated diseases using SHP2 inhibitors
US9815813B2 (en) 2014-01-17 2017-11-14 Novartis Ag 1-(triazin-3-yl/pyridazin-3-yl)-piper(-azine)idine derivatives and compositions therefor for inhibiting the activity of SHP2
JO3517B1 (ar) 2014-01-17 2020-07-05 Novartis Ag ان-ازاسبيرو الكان حلقي كبديل مركبات اريل-ان مغايرة وتركيبات لتثبيط نشاط shp2
ES2699351T3 (es) 2014-01-17 2019-02-08 Novartis Ag Derivados de 1-piridazin/triazin-3-il-piper(-azina)/idina/pirolidina y composiciones de las mismas para inhibir la actividad de SHP2
JO3556B1 (ar) 2014-09-18 2020-07-05 Araxes Pharma Llc علاجات مدمجة لمعالجة السرطان
CN105418632B (zh) 2014-09-19 2020-02-21 上海创诺医药集团有限公司 噻吩并嘧啶类衍生物、其制备方法及其在医药上的应用
WO2016077793A1 (en) 2014-11-14 2016-05-19 Children's Hospital Medical Center Sos1 inhibitors for cancer treatment
EA037663B1 (ru) 2015-01-16 2021-04-28 Те Дженерал Хоспитал Корпорейшн СОЕДИНЕНИЯ ДЛЯ УЛУЧШЕНИЯ СПЛАЙСИНГА мРНК
WO2016191328A1 (en) 2015-05-22 2016-12-01 Allosta Pharmaceuticals Methods to prepare and employ binding site models for modulation of phosphatase activity and selectivity determination
US10532977B2 (en) 2015-06-01 2020-01-14 Indiana University Research And Technology Corporation Small molecule inhibitors of protein tyrosine phosphatases and uses thereof
EP3310771B1 (en) 2015-06-19 2020-07-22 Novartis AG Compounds and compositions for inhibiting the activity of shp2
ES2741746T3 (es) 2015-06-19 2020-02-12 Novartis Ag Compuestos y composiciones para inhibir la actividad de SHP2
CN112625028A (zh) 2015-06-19 2021-04-09 诺华股份有限公司 用于抑制shp2活性的化合物和组合物
EP3325490B1 (en) 2015-07-23 2019-12-18 Takeda Pharmaceutical Company Limited 1-substituted 1,2,3,4-tetrahydro-1,7-naphthyridin-8-amine derivatives and their use as ep4 receptor antagonists
WO2017078499A2 (ko) 2015-11-06 2017-05-11 경북대학교 산학협력단 단백질 타이로신 탈인산화효소 억제제를 포함하는 신경염증성 질환의 예방 또는 치료용 조성물
US11008372B2 (en) 2015-11-07 2021-05-18 Board Of Regents, The University Of Texas System Targeting proteins for degradation
US9932288B2 (en) 2015-12-09 2018-04-03 West Virginia University Chemical compound for inhibition of SHP2 function and for use as an anti-cancer agent
WO2017156397A1 (en) 2016-03-11 2017-09-14 Board Of Regents, The University Of Texas Sysytem Heterocyclic inhibitors of ptpn11
WO2017210134A1 (en) 2016-05-31 2017-12-07 Board Of Regents, University Of Texas System Heterocyclic inhibitors of ptpn11
CA3026784A1 (en) 2016-06-07 2017-12-14 Jacobio Pharmaceuticals Co., Ltd. Heterocyclic pyrazine derivatives useful as shp2 inhibitors
SG11201900157RA (en) 2016-07-12 2019-02-27 Revolution Medicines Inc 2,5-disubstituted 3-methyl pyrazines and 2,5,6-trisubstituted 3-methyl pyrazines as allosteric shp2 inhibitors
EP3515916B1 (en) 2016-09-22 2023-06-07 Relay Therapeutics, Inc. Shp2 phosphatase inhibitors and methods of use thereof
TW201819386A (zh) 2016-10-24 2018-06-01 美商傳達治療有限公司 Shp2磷酸酶抑制劑及其使用方法
EP3558979B1 (en) 2016-12-22 2021-02-17 Boehringer Ingelheim International GmbH Novel benzylamino substituted quinazolines and derivatives as sos1 inhibitors
WO2018129402A1 (en) 2017-01-06 2018-07-12 Oregon Health & Science University Compositions and methods used in diagnosing and treating colorectal cancer
WO2018130928A1 (en) 2017-01-10 2018-07-19 Novartis Ag Pharmaceutical combination comprising an alk inhibitor and a shp2 inhibitor
JP7240319B2 (ja) 2017-01-23 2023-03-15 レヴォリューション・メディスンズ,インコーポレイテッド アロステリックshp2阻害剤としての二環式化合物
EP3571189B1 (en) 2017-01-23 2023-03-29 Revolution Medicines, Inc. Pyridine compounds as allosteric shp2 inhibitors
EP3589647A1 (en) 2017-02-28 2020-01-08 Novartis AG Shp inhibitor compositions and uses for chimeric antigen receptor therapy
US20220235013A1 (en) 2017-03-21 2022-07-28 Bayer Pharma Aktiengesellschaft 2-methyl-quinazolines
EP3601239A4 (en) 2017-03-23 2020-05-13 Jacobio Pharmaceuticals Co., Ltd. INNOVATIVE HETEROCYCLIC DERIVATIVES USEFUL AS SHP2 INHIBITORS
CN110770243A (zh) 2017-05-02 2020-02-07 锐新医药公司 作为mtor抑制剂的雷帕霉素类似物
EP3630770A1 (en) 2017-05-26 2020-04-08 Relay Therapeutics, Inc. Pyrazolo[3,4-b]pyrazine derivatives as shp2 phosphatase inhibitors
WO2019051084A1 (en) 2017-09-07 2019-03-14 Revolution Medicines, Inc. SHP2 INHIBITOR COMPOSITIONS AND METHODS OF TREATING CANCER
US10435389B2 (en) 2017-09-11 2019-10-08 Krouzon Pharmaccuticals, Inc. Octahydrocyclopenta[c]pyrrole allosteric inhibitors of SHP2
US11701354B2 (en) 2017-09-29 2023-07-18 D. E. Shaw Research, Llc Pyrazolo[3,4-b]pyrazine derivatives as SHP2 phosphatase inhibitors
EA202091491A1 (ru) * 2017-12-21 2020-11-13 Бёрингер Ингельхайм Интернациональ Гмбх Новые бензиламинозамещенные пиридопиримидиноны и производные в качестве ингибиторов sos1
US11426422B2 (en) 2018-01-30 2022-08-30 Research Development Foundation SHP2 inhibitors and methods of use thereof
AU2019222026B2 (en) 2018-02-13 2022-05-12 Shanghai Blueray Biopharma Co., Ltd. Pyrimidine-fused cyclic compound, preparation method therefor and application thereof
WO2019165073A1 (en) 2018-02-21 2019-08-29 Relay Therapeutics, Inc. Shp2 phosphatase inhibitors and methods of use thereof
SG11202006778TA (en) 2018-03-02 2020-08-28 Otsuka Pharma Co Ltd Pharmaceutical compounds
TW202003471A (zh) 2018-03-21 2020-01-16 美商傳達治療有限公司 Shp2磷酸酶抑制劑及其使用方法
US20210069188A1 (en) 2018-03-21 2021-03-11 Relay Therapeutics, Inc. Pyrazolo[3,4-b]pyrazine shp2 phosphatase inhibitors and methods of use thereof
US10561655B2 (en) 2018-03-21 2020-02-18 Synblia Therapeutics, Inc. SHP2 inhibitors and uses thereof
CN112203689A (zh) 2018-04-10 2021-01-08 锐新医药公司 治疗癌症的shp2抑制剂组合物和方法
CA3097231A1 (en) 2018-04-18 2019-10-24 Bayer Pharma Aktiengesellschaft 2-methyl-aza-quinazolines
MX2020011564A (es) 2018-05-01 2021-01-29 Revolution Medicines Inc Analogos de rapamicina unidos a c26 como inhibidores de mtor.
CA3098692A1 (en) 2018-05-01 2019-11-07 Revolution Medicines, Inc. C40-, c28-, and c-32-linked rapamycin analogs as mtor inhibitors
MX2020011528A (es) 2018-05-02 2021-02-09 Navire Pharma Inc Inhibidores heterociclicos sustituidos de ptpn11.
EP3801613A1 (en) 2018-06-04 2021-04-14 Bayer Aktiengesellschaft Inhibitors of shp2
EA202190342A1 (ru) 2018-07-24 2021-07-22 Тайхо Фармасьютикал Ко., Лтд. Гетеробициклические соединения для ингибирования активности shp2
WO2020033286A1 (en) 2018-08-06 2020-02-13 Purdue Research Foundation Novel sesquiterpenoid analogs
EP4356973A2 (en) 2018-08-10 2024-04-24 Navire Pharma, Inc. 6-(4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-3-(2,3-dichlorophenyl)-2-methylpyrimidin-4(3h)-one derivatives and related compounds as ptpn11 (shp2) inhibitors for treating cancer
CA3113233A1 (en) 2018-09-18 2020-03-26 Nikang Therapeutics, Inc. Fused tricyclic ring derivatives as src homology-2 phosphatase inhibitors
WO2020063760A1 (en) 2018-09-26 2020-04-02 Jacobio Pharmaceuticals Co., Ltd. Novel heterocyclic derivatives useful as shp2 inhibitors
WO2020072656A1 (en) 2018-10-03 2020-04-09 Gilead Sciences, Inc. Imidozopyrimidine derivatives
JP2022508651A (ja) 2018-10-08 2022-01-19 レヴォリューション・メディスンズ,インコーポレイテッド 癌を処置するためのshp2阻害剤組成物および方法
TW202028183A (zh) 2018-10-10 2020-08-01 大陸商江蘇豪森藥業集團有限公司 含氮雜芳類衍生物調節劑、其製備方法和應用
CN111295384B (zh) 2018-10-10 2022-08-12 江苏豪森药业集团有限公司 双环类衍生物抑制剂、其制备方法和应用
BR112021005733A2 (pt) 2018-10-17 2021-07-27 Array Biopharma Inc. inibidores de proteína tirosina fosfatase
CN113767100A (zh) 2019-03-01 2021-12-07 锐新医药公司 双环杂芳基化合物及其用途
WO2020180770A1 (en) 2019-03-01 2020-09-10 Revolution Medicines, Inc. Bicyclic heterocyclyl compounds and uses thereof
US20230029385A1 (en) 2019-10-15 2023-01-26 Bayer Aktiengesellschaft 2-methyl-aza-quinazolines
CN114901662A (zh) * 2019-11-08 2022-08-12 锐新医药公司 双环杂芳基化合物及其用途

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022212546A1 (en) * 2021-03-31 2022-10-06 Acerand Therapeutics (Usa) Limited Pyridopyrimidinone compounds
WO2022240947A1 (en) * 2021-05-12 2022-11-17 Revolution Medicines, Inc. Use of sos1 inhibitors with mtor inhibitors to treat cancers
WO2023059597A1 (en) * 2021-10-05 2023-04-13 Mirati Therapeutics, Inc. Combination therapies of kras g12d inhibitors with sos1 inhibitors
US20230339952A1 (en) * 2022-04-20 2023-10-26 Kumquat Biosciences Inc. Macrocyclic heterocycles and uses thereof
US11912708B2 (en) * 2022-04-20 2024-02-27 Kumquat Biosciences Inc. Macrocyclic heterocycles and uses thereof
WO2023215256A1 (en) * 2022-05-03 2023-11-09 Revolution Medicines, Inc. Sos1 inhibitors and uses thereof
WO2023215257A3 (en) * 2022-05-03 2024-04-04 Revolution Medicines, Inc. Sos1 inhibitors and uses thereof

Also Published As

Publication number Publication date
JP2023500328A (ja) 2023-01-05
US11168102B1 (en) 2021-11-09
PE20230249A1 (es) 2023-02-07
CN116425742A (zh) 2023-07-14
IL292679A (en) 2022-07-01
CA3156359A1 (en) 2021-05-14
KR20220100903A (ko) 2022-07-18
CN114901662A (zh) 2022-08-12
CL2023000416A1 (es) 2023-07-28
BR112022008858A2 (pt) 2022-09-06
CR20220200A (es) 2022-07-28
EP4055017A1 (en) 2022-09-14
MX2022005525A (es) 2022-06-08
CO2022005968A2 (es) 2022-07-29
WO2021092115A1 (en) 2021-05-14
AU2020380315A1 (en) 2022-05-26
US20230365605A1 (en) 2023-11-16
TW202128688A (zh) 2021-08-01

Similar Documents

Publication Publication Date Title
US11168102B1 (en) Bicyclic heteroaryl compounds and uses thereof
US20230096028A1 (en) Bicyclic heterocyclyl compounds and uses thereof
US11739074B2 (en) Ras inhibitors
US11053226B2 (en) KRAS G12C inhibitors and methods of using the same
US10640504B2 (en) Inhibitors of KRAS G12C and methods of using the same
US20230148450A9 (en) Bicyclic heteroaryl compounds and uses thereof
AU2022268962A1 (en) Ras inhibitors for the treatment of cancer
US20230303591A1 (en) Ras inhibitors
US20240051956A1 (en) Sos1 inhibitors and uses thereof
WO2022146698A1 (en) Sos1 inhibitors and uses thereof
WO2023215256A1 (en) Sos1 inhibitors and uses thereof
WO2023215257A2 (en) Sos1 inhibitors and uses thereof
JP2024517024A (ja) がん治療におけるsos1阻害剤とmtor阻害剤の併用

Legal Events

Date Code Title Description
AS Assignment

Owner name: REVOLUTION MEDICINES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GILL, ADRIAN L.;BUCKL, ANDREAS;KOLTUN, ELENA S.;AND OTHERS;SIGNING DATES FROM 20200827 TO 20201030;REEL/FRAME:054300/0001

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

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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