WO2023173083A1 - Tetrahydroindole derivatives as egfr and/or her2 inhibtors useful for the treatment of cancer - Google Patents

Tetrahydroindole derivatives as egfr and/or her2 inhibtors useful for the treatment of cancer Download PDF

Info

Publication number
WO2023173083A1
WO2023173083A1 PCT/US2023/064133 US2023064133W WO2023173083A1 WO 2023173083 A1 WO2023173083 A1 WO 2023173083A1 US 2023064133 W US2023064133 W US 2023064133W WO 2023173083 A1 WO2023173083 A1 WO 2023173083A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
ring
group
independently selected
optionally substituted
Prior art date
Application number
PCT/US2023/064133
Other languages
French (fr)
Inventor
Benjamin C. MILGRAM
Ryan D. WHITE
Sejal Patel
Angel Guzman-Perez
Original Assignee
Scorpion Therapeutics, 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 Scorpion Therapeutics, Inc. filed Critical Scorpion Therapeutics, Inc.
Publication of WO2023173083A1 publication Critical patent/WO2023173083A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • 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
    • 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
    • C07D491/04Ortho-condensed systems

Definitions

  • This disclosure provides chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit epidermal growth factor receptor (EGFR, ERBB1) and/or Human epidermal growth factor receptor 2 (HER2, ERBB2).
  • EGFR epidermal growth factor receptor
  • HER2 ERBB2 Human epidermal growth factor receptor 2
  • These chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) EGFR and/or HER2 activation contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human).
  • This disclosure also provides compositions containing the same as well as methods of using and making the same.
  • Epidermal growth factor receptor (EGFR, ERBB1) and Human epidermal growth factor receptor 2 (HER2, ERBB2) are members of a family of proteins which regulate cellular processes implicated in tumor growth, including proliferation and differentiation.
  • EGFR Epidermal growth factor receptor
  • HER2 ERBB2 Human epidermal growth factor receptor 2
  • Several investigators have demonstrated the role of EGFR and HER2 in development and cancer (Reviewed in Salomon, et al., Crit. Rev. Oncol. Hematol. (1995) 19:183-232, Klapper, etal., Adv. Cancer Res. (2000) 77, 25-79 and Hynes and Stern, Biochim. Biophys. Acta (1994) 1198:165-184).
  • EGFR overexpression is present in at least 70% of human cancers, such as non-small cell lung carcinoma (NSCLC), breast cancer, glioma, and prostate cancer.
  • HER2 overexpression occurs in approximately 30% of all breast cancer. It has also been implicated in other human cancers including colon, ovary, bladder, stomach, esophagus, lung, uterus and prostate.
  • HER2 overexpression has also been correlated with poor prognosis in human cancer, including metastasis, and early relapse.
  • EGFR and HER2 are, therefore, widely recognized as targets for the design and development of therapies that can specifically bind and inhibit tyrosine kinase activity and its signal transduction pathway in cancer cells, and thus can serve as diagnostic or therapeutic agents.
  • EGFR tyrosine kinase inhibitors are effective clinical therapies for EGFR mutant advanced non-small cell lung cancer (NSCLC) patients.
  • NSCLC non-small cell lung cancer
  • TKIs EGFR tyrosine kinase inhibitors
  • NSCLC tumors can have EGFR exon 20 insertion mutations that are intrinsically resistant to current EGFR TKIs.
  • BUB1 Budding uninhibited by benzimidazole, BUB1
  • BUB1 Budding uninhibited by benzimidazole, BUB1
  • This protein is an essential part of the complex network of proteins that form the mitotic checkpoint.
  • the major function of an unsatisfied mitotic checkpoint is to keep the anaphase-promoting complex/cyclosome (APC/C) in an inactive state.
  • APC/C anaphase-promoting complex/cyclosome
  • mitotic checkpoint inhibition through inhibition of BUB1 kinase represents an approach for the treatment of proliferative disorders, including solid tumors such as carcinomas, sarcomas, leukemias and lymphoid malignancies or other disorders, associated with uncontrolled cellular proliferation.
  • This disclosure provides chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit epidermal growth factor receptor (EGFR, ERBB1) and/or Human epidermal growth factor receptor 2 (HER2, ERBB2).
  • EGFR epidermal growth factor receptor
  • HER2 ERBB2 Human epidermal growth factor receptor 2
  • These chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) EGFR and/or HER2 activation contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human).
  • This disclosure also provides compositions containing the same as well as methods of using and making the same.
  • this disclosure features compounds of of Formula (I):
  • Ring C is selected from the group consisting of:
  • bicyclic heteroaryl including 7-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with X 1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c ;
  • bicyclic C5-10 cycloalkyl or C5-10 cycloalkenyl each of which is optionally substituted with X 1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c ;
  • heterocyclyl or heterocycloalkenyl including from 5-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with X 1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c ; and
  • X 1 is selected from the group consisting of (a) -(X 2 )m-L 1 -R 5 ; and (b) ; wherein: m is 0 or 1 ;
  • X 2 is selected from the group consisting of:
  • L 1 and L 2 is selected from the group consisting of: a bond and C1-10 alkylene optionally substituted with from 1-6 R a ;
  • R 5 is selected from the group consisting of:
  • R 6 is selected from the group consisting of:
  • R 5 is selected from the group consisting of: H, -R g , -R g2 -
  • X 1 is other than H, -OH, or NH2;
  • L 5 is selected from the group consisting of: -O-, -S(0)o-2, -NH-, and -N(R d )-;
  • R Y is selected from the group consisting of: -R g and -(L g ) g -R g ; each of R la , R lb , R 2a , R 2b , R 3a , and R 3b is independently selected from the group consisting of:
  • Ring A is R g ;
  • heterocyclyl or heterocycloalkenyl including from 3-8 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o- 2 , and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c ;
  • heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o- 2 , and wherein the heteroaryl is optionally substituted with from 1-4 R c ;
  • each occurrence of L g is independently selected from the group consisting of: -O-, -NH-, -NR d , -S(0)o- 2 , C(O), and C1-3 alkylene optionally substituted with from 1-3 R a ; each g is independently 1, 2, or 3; each R g2 is a divalent R g group; each occurrence of R’ and R” is independently selected from the group consisting of: H; -OH; andC1-4 alkyl;
  • composition comprising a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a pharmaceutically acceptable carrier e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a method for treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a pharmaceutically acceptable salt thereof e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • Also provided herein is a method for treating cancer in a subject in need thereof, the method comprising (a) determining that the cancer is associated with a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same; and (b) administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a method of treating an EGFR-associated disease or disorder in a subject comprising administering to a subject identified or diagnosed as having an EGFR-associated disease or disorder a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a pharmaceutically acceptable salt thereof e.g., a pharmaceutically acceptable salt thereof
  • This disclosure also provides a method of treating an EGFR-associated disease or disorder in a subject, the method comprising: determining that the cancer in the subject is an EGFR-associated disease or disorder; and administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a method of treating an EGFR-associated cancer in a subject comprising administering to a subject identified or diagnosed as having an EGFR-associated cancer a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a pharmaceutically acceptable salt thereof e.g., a pharmaceutically acceptable salt thereof
  • This disclosure also provides a method of treating an EGFR-associated cancer in a subject, the method comprising: determining that the cancer in the subject is an EGFR- associated cancer; and administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • Also provided herein is a method of treating a subject having a cancer, wherein the method comprises:
  • step (b) after (a), determining whether a cancer cell in a sample obtained from the subject has at least one EGFR inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor of step (a);
  • step (c) administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has been determined to have a cancer cell that has at least one EGFR inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor of step (a); or
  • step (d) administering additional doses of the first EGFR inhibitor of step (a) to the subject if the subject has not been determined to have a cancer cell that has at least one EGFR inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor of step (a).
  • a method of treating a subject having a cancer comprises: (a) determining whether a cancer cell in a sample obtained from a subject having a cancer and previously administered one or more doses of a first EGFR inhibitor has one or more EGFR inhibitor resistance mutations that confer increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor that was previously administered to the subject; and
  • Also provided herein is a method of treating a subject having a cancer, wherein the method comprises:
  • This disclosure also provides a method for inhibiting EGFR in a mammalian cell, the method comprising contacting the mammalian cell with an effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a pharmaceutically acceptable salt thereof e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • Also provided herein is a method for treating cancer in a subject in need thereof, the method comprising (a) determining that the cancer is associated with a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same; and (b) administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a method of treating a HER2-associated cancer in a subject comprising administering to a subject identified or diagnosed as having a HER2-associated cancer a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • This disclosure also provides a method of treating a HER2-associated cancer in a subject, the method comprising: determining that the cancer in the subject is a HER2- associated cancer; and administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a method of treating a subject having a cancer comprising administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein, to a subject having a clinical record that indicates that the subject has a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a pharmaceutically acceptable salt thereof e.g., a pharmaceutical composition as provided herein
  • Also provided herein is a method of treating a subject having a cancer, wherein the method comprises:
  • step (c) administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has been determined to have a cancer cell that has at least one HER2 inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first HER2 inhibitor of step (a); or
  • step (d) administering additional doses of the first HER2 inhibitor of step (a) to the subject if the subject has not been determined to have a cancer cell that has at least one HER2 inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first HER2 inhibitor of step (a).
  • Also provided herein is a method of treating a subject having a cancer, wherein the method comprises:
  • This disclosure also provides a method for inhibiting HER2 in a mammalian cell, the method comprising contacting the mammalian cell with an effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a pharmaceutically acceptable salt thereof e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • Also provided herein is a method for treating cancer in a subject in need thereof, the method comprising (a) determining that the cancer is associated with a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same and that the cancer is associated with a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same; and (b) administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a method of treating an EGFR-associated and HER2- associated cancer in a subject comprising administering to a subject identified or diagnosed as having an EGFR-associated and a HER2-associated cancer a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a pharmaceutically acceptable salt thereof e.g., a pharmaceutically acceptable salt thereof
  • This disclosure also provides a method of treating a an EGFR-associated and HER2-associated cancer in a subject, the method comprising: determining that the cancer in the subject is an EGFR-associated and a HER2-associated cancer; and administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • This disclosure also provides a method for inhibiting EGFR and HER2 in a mammalian cell, the method comprising contacting the mammalian cell with an effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a pharmaceutically acceptable salt thereof e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a method for inhibiting a BUB (budding uninhibited by benzimidazole, BUB1-3) kinase in addition to the above, provided herein is a method for inhibiting a BUB (budding uninhibited by benzimidazole, BUB1-3) kinase.
  • the methods provided herein include methods for inhibiting BUB11.
  • a method for inhibiting BUB1 in a mammalian cell the method comprising contacting the mammalian cell with an effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I- C), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof.
  • API refers to an active pharmaceutical ingredient.
  • an “effective amount” or “therapeutically effective amount,” as used herein, refer to a sufficient amount of a chemical entity being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result includes reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system.
  • an “effective amount” for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms.
  • An appropriate “effective” amount in any individual case is determined using any suitable technique, such as a dose escalation study.
  • excipient or “pharmaceutically acceptable excipient” means a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, carrier, solvent, or encapsulating material.
  • each component is “pharmaceutically acceptable” in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salt refers to a formulation of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound.
  • pharmaceutically acceptable salts are obtained by reacting a compound described herein, with acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
  • pharmaceutically acceptable salts are obtained by reacting a compound having acidic group described herein with a base to form a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, A-methyl-D-glucamine, tris(hydroxymethyl)methylamine, and salts with amino acids such as arginine, lysine, and the like, or by other methods previously determined.
  • a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, A-methyl-D-glucamine, tris(hydroxymethyl)methylamine, and salts with amino acids such as arginine, lysine, and the like, or by other methods previously determined.
  • Examples of a salt that the compounds described hereinform with a base include the following: salts thereof with inorganic bases such as sodium, potassium, magnesium, calcium, and aluminum; salts thereof with organic bases such as methylamine, ethylamine and ethanolamine; salts thereof with basic amino acids such as lysine and ornithine; and ammonium salt.
  • the salts may be acid addition salts, which are specifically exemplified by acid addition salts with the following: mineral acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, nitric acid, and phosphoric acid: organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, methanesulfonic acid, and ethanesulfonic acid; acidic amino acids such as aspartic acid and glutamic acid.
  • mineral acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, nitric acid, and phosphoric acid
  • organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric
  • composition refers to a mixture of a compound described herein with other chemical components (referred to collectively herein as “excipients”), such as carriers, stabilizers, diluents, dispersing agents, suspending agents, and/or thickening agents.
  • excipients such as carriers, stabilizers, diluents, dispersing agents, suspending agents, and/or thickening agents.
  • the pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but not limited to: rectal, oral, intravenous, aerosol, parenteral, ophthalmic, pulmonary, and topical administration.
  • subject refers to an animal, including, but not limited to, a primate (e.g., human), monkey, cow, pig, sheep, goat, horse, dog, cat, rabbit, rat, or mouse.
  • primate e.g., human
  • monkey cow, pig, sheep, goat
  • horse dog, cat, rabbit, rat
  • patient are used interchangeably herein in reference, for example, to a mammalian subject, such as a human.
  • alkyl refers to a saturated acyclic hydrocarbon radical that may be a straight chain or branched chain, containing the indicated number of carbon atoms.
  • Ci-io indicates that the group may have from 1 to 10 (inclusive) carbon atoms in it.
  • Alkyl groups can either be unsubstituted or substituted with one or more substituents. Non-limiting examples include methyl, ethyl, Ao-propyl, tert-butyl, /7-hexyl.
  • saturated as used in this context means only single bonds present between constituent carbon atoms and other available valences occupied by hydrogen and/or other substituents as defined herein.
  • haloalkyl refers to an alkyl, in which one or more hydrogen atoms is/are replaced with an independently selected halo.
  • alkoxy refers to an -O-alkyl radical (e.g., -OCH3).
  • alkenyl refers to an acyclic hydrocarbon chain that may be a straight chain or branched chain having one or more carbon-carbon double bonds.
  • the alkenyl moiety contains the indicated number of carbon atoms. For example, C2-6 indicates that the group may have from 2 to 6 (inclusive) carbon atoms in it.
  • Alkenyl groups can either be unsubstituted or substituted with one or more substituents.
  • alkynyl refers to an acyclic hydrocarbon chain that may be a straight chain or branched chain having one or more carbon-carbon triple bonds.
  • the alkynyl moiety contains the indicated number of carbon atoms. For example, C2-6 indicates that the group may have from 2 to 6 (inclusive) carbon atoms in it.
  • Alkynyl groups can either be unsubstituted or substituted with one or more substituents.
  • aryl refers to a 6-20 carbon mono-, bi-, tri- or polycyclic group wherein at least one ring in the system is aromatic (e.g., 6-carbon monocyclic, 10-carbon bicyclic, or 14-carbon tricyclic aromatic ring system); and wherein 0, 1, 2, 3, or 4 atoms of each ring may be substituted by a substituent.
  • aryl groups include phenyl, naphthyl, tetrahydronaphthyl, and the like.
  • cycloalkyl refers to cyclic saturated hydrocarbon groups having, e.g., 3 to 20 ring carbons, preferably 3 to 16 ring carbons, and more preferably 3 to 12 ring carbons or 3-10 ring carbons or 3-6 ring carbons, wherein the cycloalkyl group may be optionally substituted.
  • cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • Cycloalkyl may include multiple fused and/or bridged rings.
  • Non-limiting examples of fused/bridged cycloalkyl includes: bicyclo[1.1.0]butane, bicyclo[2.1.0]pentane, bicyclo [1.1.1] pentane, bicyclo[3.1.0]hexane, bicyclo[2.1.1]hexane, bicyclo [3.2.0] heptane, bicyclo[4.1.0]heptane, bicyclo[2.2.1]heptane, bicyclo[3.1.1]heptane, bicyclo[4.2.0]octane, bicyclo[3.2.1]octane, bicyclo[2.2.2]octane, and the like.
  • Cycloalkyl also includes spirocyclic rings (e.g., spirocyclic bicycle wherein two rings are connected through just one atom).
  • spirocyclic cycloalkyls include spiro[2.2]pentane, spiro[2.5] octane, spiro[3.5]nonane, spiro[3.5]nonane, spiro[3.5]nonane, spiro [4.4] nonane, spiro[2.6]nonane, spiro [4.5] decane, spiro[3.6]decane, spiro[5.5]undecane, and the like.
  • saturated as used in this context means only single bonds present between constituent carbon atoms.
  • cycloalkenyl as used herein means partially unsaturated cyclic hydrocarbon groups having 3 to 20 ring carbons, preferably 3 to 16 ring carbons, and more preferably 3 to 12 ring carbons or 3-10 ring carbons or 3-6 ring carbons, wherein the cycloalkenyl group may be optionally substituted.
  • Examples of cycloalkenyl groups include, without limitation, cyclopentenyl, cyclohexenyl, cycloheptenyl, and cyclooctenyl.
  • cycloalkenyl groups may have any degree of unsaturation provided that one or more double bonds is present in the ring, none of the rings in the ring system are aromatic, and the cycloalkenyl group is not fully saturated overall.
  • Cycloalkenyl may include multiple fused and/or bridged and/or spirocyclic rings.
  • heteroaryl means a mono-, bi-, tri- or polycyclic group having 5 to 20 ring atoms, alternatively 5, 6, 9, 10, or 14 ring atoms; wherein at least one ring in the system contains one or more heteroatoms independently selected from the group consisting of N, O, and S and at least one ring in the system is aromatic (but does not have to be a ring which contains a heteroatom, e.g. tetrahydroisoquinolinyl, e.g., tetrahydroquinolinyl). Heteroaryl groups can either be unsubstituted or substituted with one or more substituents.
  • heteroaryl examples include thienyl, pyridinyl, furyl, oxazolyl, oxadiazolyl, pyrrolyl, imidazolyl, triazolyl, thiodiazolyl, pyrazolyl, isoxazolyl, thiadiazolyl, pyranyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, thiazolyl benzothienyl, benzoxadiazolyl, benzofuranyl, benzimidazolyl, benzotriazolyl, cinnolinyl, indazolyl, indolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, purinyl, thienopyridinyl, pyrido[2,3- t7]pyrimidinyl, pyrrolo[2,3-/>]pyridinyl, quinazol
  • the heteroaryl is selected from thienyl, pyridinyl, furyl, pyrazolyl, imidazolyl, isoindolinyl, pyranyl, pyrazinyl, and pyrimidinyl.
  • heterocyclyl refers to a mono-, bi-, tri-, or polycyclic saturated ring system with 3-16 ring atoms (e.g., 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system) having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic or polycyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2 or 3 atoms of each ring may be substituted by a substituent.
  • ring atoms e.g., 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system
  • heteroatoms selected from O, N, or S (e.g.
  • heterocyclyl groups include piperazinyl, pyrrolidinyl, dioxanyl, morpholinyl, tetrahydrofuranyl, and the like.
  • Heterocyclyl may include multiple fused and bridged rings.
  • fused/bridged heteorocyclyl includes: 2-azabicyclo[ 1.1.0] butane, 2-azabicyclo[2.1.0]pentane, 2- azabicyclo[l. l.
  • Heterocyclyl also includes spirocyclic rings (e.g., spirocyclic bicycle wherein two rings are connected through just one atom).
  • Non-limiting examples of spirocyclic heterocyclyls include 2- azaspiro[2.2]pentane, 4-azaspiro[2.5 ] octane, 1 -azaspiro [3.5] nonane, 2- azaspiro [3.5] nonane, 7-azaspiro[3.5]nonane, 2-azaspiro[4.4] nonane, 6- azaspiro[2.6]nonane, 1 , 7-diazaspir o [4.5] decane, 7-azaspiro[4.5] decane 2,5- diazaspiro [ 3.6] decane, 3 -azaspiro [5.5] undecane, 2-oxaspiro[2.2]pentane, 4- oxaspiro[2.5] octane, 1 -oxaspiro [3.5] nonane, 2-oxaspiro[3.5]nonane, 7- oxaspiro [3.5] nonane
  • heterocycloalkenyl as used herein means partially unsaturated cyclic ring system with 3-16 ring atoms (e.g., 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system) having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic or polycyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2 or 3 atoms of each ring may be substituted by a substituent.
  • ring atoms e.g., 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system
  • heteroatoms selected from O, N, or S (e.g., carbon atom
  • heterocycloalkenyl groups include, without limitation, tetrahydropyridyl, dihydropyrazinyl, dihydropyridyl, dihydropyrrolyl, dihydrofuranyl, dihydrothiophenyl.
  • partially unsaturated cyclic groups heterocycloalkenyl groups may have any degree of unsaturation provided that one or more double bonds is present in the ring, none of the rings in the ring system are aromatic, and the heterocycloalkenyl group is not fully saturated overall.
  • Heterocycloalkenyl may include multiple fused and/or bridged and/or spirocyclic rings.
  • aromatic rings include: benzene, pyridine, pyrimidine, pyrazine, pyridazine, pyridone, pyrrole, pyrazole, oxazole, thioazole, isoxazole, isothiazole, and the like.
  • a ring when a ring is described as being “partially unsaturated”, it means said ring has one or more additional degrees of unsaturation (in addition to the degree of unsaturation attributed to the ring itself; e.g., one or more double or tirple bonds between constituent ring atoms), provided that the ring is not aromatic.
  • additional degrees of unsaturation in addition to the degree of unsaturation attributed to the ring itself; e.g., one or more double or tirple bonds between constituent ring atoms
  • examples of such rings include: cyclopentene, cyclohexene, cycloheptene, dihydropyridine, tetrahydropyridine, dihydropyrrole, dihydrofuran, dihydrothiophene, and the like.
  • rings and cyclic groups e.g., aryl, heteroaryl, heterocyclyl, heterocycloalkenyl, cycloalkenyl, cycloalkyl, and the like described herein
  • rings and cyclic groups encompass those having fused rings, including those in which the points of fusion are located (i) on adjacent ring atoms (e.g., [x.x.O] ring systems, in of ring atoms (bridged ring systems having all bridge lengths > 0) (e.g.,
  • atoms making up the compounds of the present embodiments are intended to include all isotopic forms of such atoms.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include tritium and deuterium
  • isotopes of carbon include 13 C and 14 C.
  • a compound containing the encompasses the tautomeric form containing the moiety: . y, a pyridinyl or pyrimidinyl moiety that is described to be optionally substituted with hydroxyl encompasses pyridone or pyrimidone tautomeric forms.
  • the compounds provided herein may encompass various stereochemical forms.
  • the compounds also encompass diastereomers as well as optical isomers, e.g., mixtures of enantiomers including racemic mixtures, as well as individual enantiomers and diastereomers, which arise as a consequence of structural asymmetry in certain compounds.
  • optical isomers e.g., mixtures of enantiomers including racemic mixtures, as well as individual enantiomers and diastereomers, which arise as a consequence of structural asymmetry in certain compounds.
  • a disclosed compound is named or depicted by a structure without specifying the stereochemistry and has one or more chiral centers, it is understood to represent all possible stereoisomers of the compound.
  • the chemical entities provided herein can inhibit an EGFR kinase and/or a HER2 kinase that has an exon 20 mutation (e.g., any of the exon 20 mutations described herein).
  • Exon 20 mutations can confer intrinsic resistance to EGFR and/or HER2 inhibitors, and there are currently only limited targeted therapies that have been approved for subjects with these mutations.
  • This disclosure also provides compositions containing the chemical entities provided herein as well as methods of using and making the same.
  • this disclosure features compounds of of Formula (I):
  • Ring C is selected from the group consisting of:
  • bicyclic heteroaryl including 7-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with X 1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c ;
  • bicyclic C5-10 cycloalkyl or C5-10 cycloalkenyl each of which is optionally substituted with X 1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c ;
  • heterocyclyl or heterocycloalkenyl including from 5-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with X 1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c ; and
  • X 1 is selected from the group consisting of (a) -(X 2 )m-L 1 -R 5 ; and (b) ; wherein: m is 0 or 1 ;
  • X 2 is selected from the group consisting of:
  • L 1 and L 2 is selected from the group consisting of: a bond and C1-10 alkylene optionally substituted with from 1-6 R a ;
  • R 5 is selected from the group consisting of:
  • R 6 is selected from the group consisting of:
  • R 5 is selected from the group consisting of: H, -R g , -R g2 -
  • X 1 is other than H, -OH, or NH2;
  • L 5 is selected from the group consisting of: -O-, -S(0)o-2, -NH-, and -N(R d )-;
  • R Y is selected from the group consisting of: -R g and -(L g ) g -R g ; each of R la , R lb , R 2a , R 2b , R 3a , and R 3b is independently selected from the group consisting of:
  • Ring A is R g ;
  • heterocyclyl or heterocycloalkenyl including from 3-8 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o- 2 , and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c ;
  • heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o- 2 , and wherein the heteroaryl is optionally substituted with from 1-4 R c ;
  • each occurrence of L g is independently selected from the group consisting of: -O-, -NH-, -NR d , -S(0)o- 2 , C(O), and C1-3 alkylene optionally substituted with from 1-3 R a ; each g is independently 1, 2, or 3; each R g2 is a divalent R g group; each occurrence of R’ and R” is independently selected from the group consisting of: H; -OH; andC1-4 alkyl; In some embodiments, it is provided that when Ring A is unsubstituted phenyl; each of R 2a , R 2b , R 3a , and R 3b is H, and -O-L-R 5 is -O-(unsubstituted C1-4 alkyl); OCH2CHF2; OCH2CF3; OCH 2 -cyclopropyl; OCH2CH2OCH3; or OCH
  • Ring C is X1 ( R7 ) n .
  • Ring C is bicyclic heteroaryl including 7-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with X 1 and further optionally substituted with from 1-4 R cA , wherein each R cA is an independently selected R c .
  • Ring C is bicyclic heteroaryl including 9-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with X 1 and further optionally substituted with from 1-4 R cA , wherein each R cA is an independently selected R c .
  • Ring C is bicyclic heteroaryl including 9-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 R cA , wherein each R cA is an independently selected R c In certain of these embodiments, Ring C is connected to
  • Ring Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA ; n is 0, 1, or 2; and each R cA is an independently selected R c .
  • Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • Ring C can be selected from the group consisting wherein each R cA is an independently selected R c .
  • Ring C can be selected from the group consisting wherein each R cA is an independently selected R c .
  • Ring C can be selected from the group consisting wherein each R cA is an independently selected R c .
  • Ring C can be selected from the group consisting wherein each R cA is an independently selected R c .
  • Ring C is selected from optionally substituted with R cA , wherein each R cA is an independently selected R c .
  • Ring wherein R cA is an independently selected R c .
  • R CA is an independently selected R c .
  • R cA is independently selected from the group consisting of:C1-4 alkoxyC; 1-4 haloalkoxy; C1-3 alkyl; and C1-3 alkyl substituted with from 1-3 independently selected halo.
  • Ring Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • Ring C can be selected each further optionally substituted with R cA , wherein each
  • R CA is an independently selected R c .
  • Ring C can be any independently selected R c .
  • Ring C can be any independently selected R c .
  • Ring C can be any independently selected R c .
  • Ring Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA ; n2 is 0 or 1; and each R CA is an independently selected R c .
  • Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • Ring C can be selected
  • R CA is an independently selected R c .
  • Ring C can be any independently selected R c .
  • Ring C can be any independently selected R c .
  • Ring C can be any independently selected R c .
  • Ring Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • Ring C can be selected from the group consisting of: each further optionally substituted with R cA , wherein each
  • R CA is an independently selected R c .
  • Ring C is selected from the group consisting of: each further optionally substituted with R cA , wherein each R cA is an independently selected R c .
  • Ring C is bicyclic heteroaryl including 9-10 ring atoms, wherein from 1 -4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 R cA , wherein each R cA is an independently selected
  • Ring C is connected t via a 5-membered ring.
  • Ring C can be selected from the group consisting of:
  • Ring C is bicyclic heteroaryl including 9-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is substituted with X 1 and further optionally substituted with from 1-4 R cA , wherein each R cA is an independently selected R c .
  • Ring Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA ; n is 0, 1, or 2; and each R cA is an independently selected R c .
  • Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • Ring C can be selected from the group consisting of: each further optionally substituted with R cA , wherein each R cA is an independently selected
  • Ring C can
  • Ring each of which is further optionally substituted with from 1-2 R cA , wherein each R cA is an independently selected R c .
  • Ring In certain of these embodiments, Ring wherein R cA is an independently selected R c .
  • Ring Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • Ring C can be selected from the group consisting of: and each further optionally substituted with R cA , wherein each R cA is an independently selected R c .
  • Ring C can
  • Ring Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA ; n2 is 0 or 1; and each R CA is an independently selected R c .
  • Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • Ring C can be selected optionally substituted with R cA , wherein each R cA is an independently selected R c .
  • Ring unsaturated or aromatic ring including 5 ring atoms wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • Ring C is heterocyclyl or heterocycloalkenyl including from 5-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with X 1 and further optionally substituted with from 1 -4 substituents independently selected from the group consisting of oxo and R cA , wherein each R cA is an independently selected R c .
  • Ring C is heterocyclyl including from 4-8, such as 5-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with X 1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R cA , wherein each R CA is an independently selected R c .
  • Ring C can be
  • n is 1. In some embodiments, m is 0.
  • R 6 is -R g2 -R w .
  • -R 6 is , wherein Ring
  • D is heterocyclylene including from 3-10 ring atoms, wherein from 0-2 ring atoms (in addition to the ring nitrogen atom bonded to R w ) are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclylene is optionally substituted with from 1-3 substituents each independently selected from the group consisting of: oxo and -R c .
  • -R 6 is , wherein Ring
  • D is heterocyclylene including from 3-10 ring atoms, wherein from 0-2 ring atoms (in addition to the ring nitrogen atom bonded to R w ) are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclylene is optionally substituted with from 1-3 substituents each independently selected from the group consisting of: oxo and -R c ; optionally wherein -R 6 is a monocyclic heterocyclylene ring including from 3-10 ring atoms as defined above with a nitrogen atom bonded t optionally wherein -R 6 is a bicyclic heterocyclylene ring including from 3-10 ring atoms as defined above with a nitrogen atom bonded to R w (e.g. wherein xl and x2 are each independently 0, 1 , or 2.
  • R 6 is selected from the group consisting of:
  • W is C2-6 alkenyl optionally substituted with from 1-3 R a and further optionally substituted with R g , wherein W is attached to L w via an sp 2 hybridized carbon atom.
  • W is C2-6 alkenyl or C2-6 optionally substituted with from 1 -3 R a and further optionally substituted with R g , wherein W is attached to L w via an sp 2 or sp hybridized carbon atom.
  • W is C2-4 alkenyl optionally substituted with from 1-3 R a and further optionally substituted with R g , wherein W is attached to L w via an sp 2 hybridized carbon atom.
  • X 2 is selected from the group consisting of: -O-, -N(R N )-, and -S(0)o- 2 .
  • X 2 is -N(R N )-.
  • X 2 can be - N(H)-.
  • X 2 can be -O-.
  • X 2 is C2-6 alkenyl ene optionally substituted with from 1-3
  • R a For example, X 2 can In certain embodiments, L 1 is a bond.
  • L 1 is C1-10 alkylene optionally substituted with from 1-6 R a
  • L 1 is C1-3 alkylene optionally substituted with from 1-6 R a . In certain of the foregoing embodiments, L 1 is unsubstituted C1-3 alkylene. As non-limiting examples of the foregoing embodiments, L 1 can be -CH2-, -CH2CH2-, - CH2CF2-, or -CH(Me)-. For example, L 1 can be -CH2-, -CH2CH2-, or -CH(Me)-.
  • L 1 is branched C3-6 alkylene optionally substituted with from 1-6 R a .
  • L 1 can be w h e rein aa is the point of attachment to R 5 .
  • R 5 is -C1-6 alkoxy or -S(0)o-2(C1-6 alkyl), each optionally substituted with from 1-6 R a .
  • R 5 is -C1-6 alkoxy optionally substituted with from 1-6 R a .
  • R 5 can be -C1-3 alkoxy.
  • R 5 can be methoxy.
  • R 5 is H or halo.
  • R 5 can be H or -F.
  • R 5 can be H.
  • R 5 is -OH or -NR e R f .
  • R 5 can be -OH.
  • R 5 is -R g .
  • R 5 is selected from the group consisting of:
  • heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 R c ;
  • R 5 is Ce-io aryl optionally substituted with from 1-4 R c . In certain of these embodiments, R 5 is phenyl optionally substituted with from 1-4 R c . As non-limiting examples of the foregoing embodiments, R 5 can be phenyl optionally substituted with from 1-2 independently selected halo, such as -F.
  • R 5 is heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 R c .
  • R 5 is heteroaryl including from 5-6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 R c .
  • R 5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(R d ), and wherein the heteroaryl is optionally substituted with
  • R 5 is heteroaryl including 5 ring atoms, wherein from 1 -4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 R c .
  • R 5 can be
  • R 5 is selected from the group consisting of: • C3-10 cycloalkyl or C3-10 cycloalkenyl, each of which is optionally substituted with from 1 -4 substituents independently selected from the group consisting of oxo and R c ; and
  • heterocyclyl or heterocycloalkenyl including from 3-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c .
  • R 5 is C3-10 cycloalkyl or C3-10 cycloalkenyl, each of which is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c .
  • R 5 is heterocyclyl or heterocycloalkenyl including from 3- 10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c .
  • the -R g2 group present in R 5 is Ce-io arylene optionally substituted with from 1-4 R c .
  • the -R g2 group present in R 5 is phenylene optionally substituted with from 1-4 R c .
  • the -R g2 group present in R 5 is 1,3- phenylene or 1 ,4-phenylene, each optionally substituted with from 1-4 R c .
  • -R g2 can be wherein bb is the point of attachment to R Y .
  • the R Y group present in R 5 is -R g .
  • the R Y group present in R 5 is heterocyclyl or heterocycloalkenyl including from 3-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c .
  • the R Y group present in R 5 is heterocyclyl including from 4-8, such as 4-6, ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c , such as wherein R Y is
  • R 5 is -L 5 -R g .
  • R 5 is -O-R g . In certain embodiments, R 5 is -0-(Ce-io aryl) wherein the Ce-io aryl is optionally substituted with from 1-4 R c .
  • R 5 can be -O-phenyl wherein the phenyl is optionally substituted with from 1-2 R c .
  • R 5 can be
  • X 1 is -(X 2 ) m -L 1 -R 5 , wherein:
  • X 2 is -N(R N )- or -O-;
  • L 1 is a bond or C1-6 alkylene optionally substituted with from 1-3 R a ;
  • R 5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(R d ), and wherein the heteroaryl is optionally substituted with from 1 -4 R c , such as wherein
  • R 5 is heteroaryl including 5 ring atoms, wherein from 1-4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 R c , such as wherein
  • R 5 is C3-10 cycloalkyl, such as C3-6 cycloalkyl, optionally substituted with from 1-4 R c , such as wherein R 5 is cyclopropyl.
  • R 5 is heterocyclyl including from 4-8, such as 4-
  • R 5 can
  • m is 1.
  • X 2 is -N(R N )- (e.g., N(H)).
  • X 2 is -O-.
  • L 1 is a bond
  • L 1 is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or -
  • L 1 is branched C3-6 alkylene.
  • L 1 can [BB]:
  • X 1 is -X -L -R 5 , wherein:
  • L 1 is a bond or C1-6 alkylene optionally substituted with from 1-3 R a ;
  • R 5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(R d ), and wherein the heteroaryl is optionally substituted with
  • R 5 is heteroaryl including 5 ring atoms, wherein from 1-4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 R c , such as wherein
  • R 5 is heterocyclyl including from 4-8, such as 4- 6, ring atoms, wherein from 1 -3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c .
  • R 5 can benzocyclyl including from 4-8, such as 4- 6, ring atoms, wherein from 1 -3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c .
  • R 5 can benzocyclyl including from 4-8, such as 4- 6, ring atoms, wherein from 1 -3 ring atoms are heteroatom
  • X 2 is -N(R N )S(O)2-, such as -N(H)S(O)2-*.
  • L 1 is a bond
  • L 1 is Ci-3 alkylene (e g., -CH 2 -, -CH2CH2-, or -
  • L 1 is branched C3-6 alkylene.
  • L 1 can wherein aa is the point of attachment to R 5 .
  • X 1 is -X 2 -L 1 -R 5 , wherein:
  • L 1 is a bond or C1-6 alkylene optionally substituted with from 1-3 R a ;
  • R 5 is - R" In certain embodiments of [CC], R 5 is phenyl optionally substituted with from 1-4
  • R c such as wherein R 5 is phenyl optionally substituted with from 1-2 independently selected halo, such as -F.
  • R 5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(R d ), and wherein the heteroaryl is optionally substituted with
  • R 5 is heteroaryl including 5 ring atoms, wherein from 1-4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 R c , such as wherein
  • R 5 is C3-10 cycloalkyl, such as C3-6 cycloalkyl, optionally substituted with from 1-4 R c , such as wherein R 5 is cyclopropyl.
  • R 5 is heterocyclyl including from 4-8, such as 4- 6, ring atoms, wherein from 1 -3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c .
  • R 5 can such as
  • L 1 is a bond
  • L 1 is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or -
  • L 1 is branched C3-6 alkylene.
  • L 1 can wherein aa is the point of attachment to R 5 .
  • X 1 is -(X 2 )m-L 1 -R 5 , wherein:
  • X 2 is -N(R N )- or -O-;
  • L 1 is a bond or C1-6 alkylene optionally substituted with from 1-3 R a ;
  • R 5 is R" 2 -R v
  • the -R g2 group present in R 5 is 1,3-phenylene or 1,4-phenylene, each optionally substituted with from 1-4 R c , such as wherein -R g2 is wherein bb is the point of attachment to R Y .
  • the R Y group present in R 5 is -R g .
  • the R Y group present in R 5 is heterocyclyl including from 4-8, such as 4-6, ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c .
  • R Y can be
  • X 2 is -N(R N )- (e.g., N(H)).
  • X 2 is -O-.
  • L 1 is a bond
  • L 1 is Ci-3 alkylene (e g., -CH 2 -, -CH2CH2-, or -
  • L 1 is branched C3-6 alkylene.
  • L 1 can , wherein aa is the point of attachment to R 5 .
  • X 1 is -X 2 -L 1 -R 5 , wherein:
  • L 1 is C1-6 alkylene optionally substituted with from 1-3 R a ;
  • R 5 is H, halo, C1-6 alkoxy optionally substituted with from 1-3 R a , or -OH. In certain embodiments of [EE], R 5 is H.
  • R 5 is halo (e.g., -F).
  • R 5 is C1-6 alkoxy optionally substituted with from 1-3 R a , such as wherein R 5 is C1-3 alkoxy such as methoxy.
  • R 5 is -OH.
  • X 2 is -N(R N )- (e.g., N(H)).
  • X 2 is -O-.
  • X 2 is -N(R N )S(O)2-, such as -N(H)S(O)2-*.
  • L 1 is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or - CH(Me)-).
  • L 1 is branched C3-6 alkylene.
  • L 1 can , wherein aa is the point of attachment to R 5 .
  • X 1 is -L 1 -R 5 , wherein L 1 is C1-6 alkylene optionally substituted with from 1-3 R a ; and R 5 is -L 5 -R g .
  • R 5 is O-R
  • R 5 is -O-(phenyl), wherein the phenyl is optionally substituted with from 1-2 R c .
  • L 1 is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or - CH(Me)-).
  • R cA is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or - CH(Me)-).
  • one occurrence of R cA is -NR e R f .
  • one occurrence of R cA is -NH2.
  • one occurrence of R cA is -NH(CI-6 alkyl), wherein the C1-6 alkyl is optionally substituted with from 1-3 substituents each independently selected from the group consisting of NR’R”, -OH, C1-6 alkoxy, C1-6 haloalkoxy, and halo.
  • R cA can be -NHMe, -NHCH2CF3, -NHCH2CH2OH, or -NHiPr.
  • R cA is N(CI-3 alkyl)2 such as NMe2.
  • R cA is C a1l-4koxy optionally substituted withC1-4 alkoxy or C1-4 haloalkoxy.
  • R cA can be OMe or OCH2CH2OMe.
  • R cA is C1-4 haloalkoxy (e.g., - OCH2CF3).
  • one occurrence of R cA is C1 t-h4ioalkoxy (e.g., -SCH3).
  • one occurrence of R cA is C1-4 alkyl, such as methyl; or wherein one occurrence of R cA is C1-4 alkyl substituted with from 1-6 independently selected halo.
  • one occurrence of R cA can be -CF3.
  • R CA can
  • one occurrence of R cA is halo (e.g., -F). In certain embodiments, one occurrence of R cA is -OH.
  • R la and R lb are both H.
  • R 2a and R 2b are both H.
  • R 2a and R 2b are independently selected substituents that is other than H.
  • one of R 2a and R 2b is a substituent that is other than H.
  • one of R 2a and R 2b is R b .
  • one of R 2a and R 2b is C1-6 alkyl which is optionally substituted with from 1-6 R a .
  • one of R 2a and R 2b is Ci -3 alkyl, such as methyl or ethyl.
  • the other of R 2a and R 2b is H.
  • R 3a and R 3b are both H.
  • R 3a and R 3b are independently selected substituents that is other than H.
  • one of R 3a and R 3b is a substituent that is other than H.
  • one of R 3a and R 3b e.g., R 3a
  • R b is R b .
  • one of R 3a and R 3b is C1-6 alkyl which is optionally substituted with from 1-6 R a .
  • one of R 3a and R 3b can be C1-3 alkyl, such as methyl or ethyl.
  • the other of R 3a and R 3b is H.
  • one of R 3a and R 3b is C1-3 alkyl optionally substituted with from 1-3 independently selected halo.
  • one of R 3a and R 3b , such as R 3a is -CH3, -CH2CH3, -CH2F, - CHF 2 , -CF 3 , -CH2CHF2, or -CH2CH2F.
  • one of R 3a and R 3b , such as R 3a is C1-3 alkyl substituted with C1-4 alkoxy, C1-4 haloalkoxy, or NR e R f .
  • one of R 3a and R 3b is -CH 2 OMe, -CH 2 CH 2 OMe, - CH(Me)CH 2 OMe, -CH 2 CH(Me)OMe, -CH 2 OEt, -CH 2 CH 2 OCHF 2 , -CH 2 NR e R f (e g., - CH 2 N(CF 3 )Me), or -CH 2 CH 2 NR e R f (e g., -CH 2 CH 2 NMe 2 ).
  • one of R 3a and R 3b is C1-3 alkyl substituted withC1-4 alkoxy.
  • one of R 3a and R 3b is -CH 2 OMe, -CH 2 CH 2 OMe, -CH(Me)CH 2 OMe, -CH 2 CH(Me)OMe, or -CH 2 OEt, such as -CH 2 OMe.
  • one of R 3a and R 3b is C1-3 alkyl substituted withC1-4 alkoxy.
  • one of R 3a and R 3b is -CH 2 OMe, -CH 2 CH 2 OMe, -CH(Me)CH 2 OMe, -CH 2 CH(Me)OMe, or -CH 2 OEt, such as -CH 2 OMe; such as -CH 2 CH 2 OMe; optionally the other one of R 3a and R 3b , such as R 3b is H.
  • one of R 3a and R 3b is C1-3 alkyl substituted withC1-4 alkoxy, C1-4 haloalkoxy, or NR e R f and further substituted with from 1-3 independently selected halo.
  • one of R 3a and R 3b is C1-3 alkyl substituted with C1-4 alkoxy and further substituted with from 1-3 independently ''O selected halo.
  • one of R 3a and R 3b can be V —1— F (e.g., V — I—
  • one of R 3a and R 3b is C3-6 alkyl substituted withC1-4 alkoxy, C1-4 haloalkoxy, or NR e R f .
  • one of R 3a and R 3b is branched C3-6 alkyl substituted with C a1-l4koxy, C h1-a4loalkoxy, or NR e R f .
  • one of R 3a and R 3b is branched C3-6 alkyl substituted with C1-4 alkoxy.
  • one of R 3a and R 3b such as R 3a
  • one of R 3a and R 3b , such as R 3a is R g or -(L g ) g -R g .
  • one of R 3a and R 3b is selected from the group consisting of: heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c ; and
  • one of R 3a and R 3b is selected from the group consisting of: cyclopropyl, cyclobutyl, oxetanyl, and azetidinyl, each of which is optionally substituted with from 1 -2 substituents independently selected from the group consisting of: C1-3 alkyl and halo, wherein the ring nitrogen of the azetidinyl is optionally substituted with R d .
  • one of R 3a and R 3b is -(C1-3 alkylene)-R g or -(C1-3 alkylene)-O-R g , and optionally the R g group of R 3a or R 3b is:
  • one of R 3a and R 3b is -CH2- R g , -CHoCHoR 8 , or -CH2-0-R g , wherein the R g group of R 3a or R 3b is selected from the group:
  • C3-6 cycloalkyl e.g., cyclopropyl, cyclobutyl
  • heterocyclyl including from 4-6 ring atoms (e.g., oxetanyl, azetidinyl), wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1 -4 substituents independently selected from the group consisting of oxo and R c (e.g., C1-3 alkyl, halo).
  • one of R 3a and R 3b is -CH2- R g , -CH2CH2R g , or -CH2-0-R g , wherein the R g group of R 3a or R 3b is selected from the group consisting of: cyclopropyl, cyclobutyl, oxetanyl, and azetidinyl, each of which is optionally substituted with from 1-2 substituents independently selected from the group consisting of: C1-3 alkyl and halo, wherein the ring nitrogen of the azetidinyl is optionally substituted with R d
  • one of R 3a and R 3b is -CH2-R g , -CHzCHzR 8 , or -CH2-0-R g , wherein the R g group of R 3a or R 3b is selected from the group consisting of: cyclopropyl, cyclobutyl, oxetanyl, 1 ,4-dioxanyl, and azetidinyl, each of which is optionally substituted with from 1-2 substituents independently selected from the group consisting of: C1-3 alkyl and halo, wherein the ring nitrogen of the azetidinyl is optionally substituted with R d .
  • one of R 3a and R 3b can be selected from the group consisting of: , such as As further non-limiting examples of the forgoing embodiments, one of R 3a and R 3b , such as R 3a , can be selected from the group consisting of:
  • one of R 3a and R 3b , such as R 3a is-(L g ) g -R w .
  • one of R 3a and R 3b is -(C1-3 alkylene)-R w ; optionally one of R 3a and R 3b , such as R 3a , is -CH2-R W , or -CH2CH2-R W .
  • one of R 3a and R 3b can be
  • one of R 3a and R 3b is -(L g ) g -R g2 -R w .
  • one of R 3a and R 3b is -(C1-3 alkyl ene)-R g2 -R w
  • one of R 3a and R 3b is -CH2-R g2 -R w
  • -CH2CH2-R g2 -R w is optionally one of R 3a and R 3b , such as R 3a , is -CH2-R g2 -R w , or -CH2CH2-R g2 -R w .
  • one of R 3a and R 3b such as R 3a , is -CH2-R g2 -R w , and wherein the R g2 group wherein the waveline represents the point of attachment to L g
  • one of R 3a and R 3b such as In some embodiments, the other of R 3a and R 3b is -H.
  • the other of R 3a and R 3b is C1-3 alkyl, such as methyl.
  • the other of R 3a and R 3b is halo, such as -F.
  • the other of R 3a and R 3b is selected from the group consisting of: -H; C1-3 alkyl (e.g., methyl); and -F.
  • R 3a and R 3b together with the Ring B ring atom to which each is attached, form a fused saturated or unsaturated ring of 3-12 ring atoms;
  • each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(R d ), O, and S(0)o-2;
  • R 3a and R 3b together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-8 ring atoms;
  • each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(R d ), O, and S(0)o-2;
  • R 3a and R 3b together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-6 ring atoms;
  • each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(R d ), O, and S(0)o-2; and wherein the fused saturated ring of 4-6 ring atoms is optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and
  • R 3a and R 3b together with the Ring B ring atom to which each is attached, form
  • R 3a and R 3b together with the Ring B ring atom to which each is attached, form: , which is optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and R c , wherein: pl and p2 are independently 0, 1, or 2;
  • R 3a and R 3b together with the Ring B ring atom to which each is attached, form a fused ring selected from the group consisting of:
  • R z is H.
  • R z is R d .
  • R z is C1-6 alkyl optionally substituted with from 1-3 independently selected R a .
  • W is C2-4 alkenyl.
  • R z can be
  • R 3a and R 3b together with the Ring B ring atom to which each is attached, form a fused C3-6 cycloalkyl, wherein the fused C3-6 cycloalkyl is optionally substituted with from 1-2 R c .
  • R 3a and R 3b together with the Ring B ring atom to which each is attached, form
  • R la , R lb , R 2a , and R 2b are each H;
  • R 3a and R 3b taken together with the Ring B ring carbon atom to which each is attached form a fused C3-6 (such as C3 or C4) cycloalkyl, wherein the fused cycloalkyl ring is optionally substituted with from 1-2 R c .
  • each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(R d ), O, and S(0)o-2;
  • R 2a and R 2b taken together with the Ring B ring atoms to which each is attached, form a fused saturated ring of 3-8 ring atoms;
  • each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(R d ), O, and S(0)o-2;
  • R 2a and R 2b such as R 2a
  • R 3a and R 3b taken together with the Ring B ring atoms to which each is attached, form a fused C3-6 cycloalkyl which is optionally substituted with from 1-2 R c .
  • R 2a and R 2b taken together with the Ring B ring atoms to which each is attached, form a fused cyclobutyl or cyclopropyl ring, e.g.,
  • the other one of R 3a and R 3b is R g or -(L g ) g -R g .
  • the other one of R 3a and R 3b is -(L g ) g -R g .
  • R 3a and R 3b are -(C1-3 alkylene)-R g or -(C1-3 alkylene)-O-R g , and optionally the R g group of R 3a or R 3b is:
  • the other one of R 3a and R 3b such as R 3a , is -CH2-R g , - CHiCHiR 8 , or -CH2-0-R g , wherein the R g group of R 3a or R 3b is:
  • the other one of R 3a and R 3b is -CH2-R g , - CH2CH2R g , or -CH2-0-R g , wherein the R g group of R 3a or R 3b is selected from the group consisting of: cyclopropyl, cyclobutyl, oxetanyl, 1 ,4-dioxanyl, and azetidinyl, each of which is optionally substituted with from 1-2 substituents independently selected from the group consisting of: C1-3 alkyl and halo, wherein the ring nitrogen of the azetidinyl is optionally substituted with R d .
  • the other one of R 3a and R 3b is selected from the
  • R la , R lb , R 2a , and R 2b are each H, and R 3a and R 3b are independently selected C1-3 alkyl.
  • R la , R lb , R 2a , and R 2b are each H; one of R 3a and R 3b , such as R 3a , is C1-3 alkyl optionally substituted with from 1-3 R a ; and the other of R 3a and R 3b is H, optionally each R a substituent present in R 3a or R 3b is independently selected from the group consisting of: halo, C1-4 alkoxy, and C1-4 haloalkoxy.
  • R la , R lb , R 2a , and R 2b are each H; one of R 3a and R 3b , such as R 3a , is C1-3 alkyl optionally substituted with from C1-4 alkoxy; optionally one of R 3a and R 3b , such as R 3a , is -CHiCHi-OMe; and the other of R 3a and R 3b is H.
  • R la , R lb , R 2a , and R 2b are each H; one of R 3a and R 3b , such as R 3a , is C1-3 alkyl optionally substituted with from 1-3 R a ; and the other of R 3a and R 3b is -F, optionally each R a substituent present in R 3a or R 3b is independently selected from the group consisting of: halo, C1-4 alkoxy, and C1-4 haloalkoxy.
  • R la , R lb , R 2a , and R 2b are each H; one of R 3a and R 3b , such as R 3a , is C1-3 alkyl optionally substituted with from 1-3 R a ; and the other of R 3a and R 3b is C1-3 alkyl (e.g., methyl), optionally each R a substituent present in R 3a or R 3b is independently selected from the group consisting of: halo, C1 a-4lkoxy, and C h1-a4loalkoxy.
  • R la , R lb , R 2a , and R 2b are each H; one of R 3a and R 3b , such as R 3a , is C3-6 (e.g., C4) alkyl optionally substituted with from 1-3 R a ; and the other of R 3a and R 3b is H, -F, or C1-3 alkyl (e.g., methyl), optionally each R a substituent present in R 3a or R 3b is independently selected from the group consisting of: halo, C1-4 alkoxy, and CM haloalkoxy.
  • R la , R lb , R 2a , and R 2b are each H, and one of R 3a and R 3b , such as R 3a , is -R g , -(C1-3 alkylene)-R g , or -(C1-3 alkylene)-O-R g , optionally wherein the R g group of R 3a or R 3b is:
  • R la , R lb , R 2a , and R 2b are each H; and R 3a and R 3b together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-6 ring atoms;
  • R la , R lb , R 2a , and R 2b are each H; and R 3a and R 3b taken together with the Ring B ring carbon atom to which each is attached form a fused C3-6 (such as C3 or C4) cycloalkyl, wherein the fused cycloalkyl ring is optionally substituted with from 1 -2 R c .
  • R la , R lb , R 2a , and R 2b are each H; and R 3a and R 3b are independently selected C1-3 alkyl.
  • R la , R lb is H, and one of R 2a and R 2b (such as R 2a ) and one of R 3a and R 3b (such as R 3a ) taken together with the Ring B ring atoms to which each is attached, form a fused C3-6 (such as C3 or C4) cycloalkyl which is optionally substituted with from 1 -2 R c ; and the other of R 2a and R 2b and the other of R 3a and R 3b are each H.
  • R 2a and R 2b and the other of R 3a and R 3b are each H.
  • the other of R 3a and R 3b is C1-3 alkyl.
  • the other of R 3a and R 3b is -CH3, -CH2CH3.
  • R la , R lb is H; one of R 2a and R 2b (such as R 2a ) and one of R 3a and R 3b (such as R 3a ) taken together with the Ring B ring atoms to which each is attached, form a fused C3-6 (such as C3 or C4) cycloalkyl which is optionally substituted with from 1 -2 R c ; and the other of R 2a and R 2b and the other of R 3a and R 3b are each H.
  • R la , R lb , R 2a , R 2b , R 3a , and R 3b are each H.
  • R 4 is hydrogen
  • R 4 is hydrogen; and R 4 is hydrogen.
  • Ring A is , wherein each R cB is an independently selected R c ; and ml is 0, 1, 2, 3, or 4.
  • ml is 1, 2, or 3.
  • ml can be 1 or 2 (e g., 2).
  • Ring is asymmetrical
  • each R cB is an independently selected R c .
  • Ring A can be any organic radical
  • Ring A is selected from the group consisting of:
  • each R cB is independently selected from the group consisting of: -halo, such as -Cl and -F; -CN; C1-4 alkoxy; C1-4 haloalkoxy; C1-3 alkyl; and C1-3 alkyl substituted with from 1-6 independently selected halo.
  • Ring A is , wherein R cB1 is R c ; and R cB2 is H or R c , optionally wherein R cB1 and R cB2 are each independently selected from the group consisting of: -halo, such as -Cl and -F; -CN; C1-4 alkoxy; C1-4 haloalkoxy; C1-3 alkyl; and C1-3 alkyl substituted with from 1-6 independently selected halo.
  • -halo such as -Cl and -F
  • -CN C1-4 alkoxy; C1-4 haloalkoxy; C1-3 alkyl; and C1-3 alkyl substituted with from 1-6 independently selected halo.
  • Ring halo such as -F or
  • R cB1 is C1-3 alkyl or C1-3 alkyl substituted with from 1-6 independently selected halo.
  • R cB1 can be methyl, -CHF2, or -CF3.
  • R cB2 is selected from the group consisting of: halo; -CN; CM alkoxy;C1-4 haloalkoxy; C1-3 alkyl; and C1-3 alkyl substituted with from 1-6 independently selected halo. In certain of these embodiments, R cB2 is C1-4 alkoxy or CM haloalkoxy.
  • R cB2 is selected from the group consisting of cyano; C1-3 alkyl; and C1-3 alkyl substituted with from 1-6 independently selected halo.
  • R cB2 can be cyano, methyl, ethyl, -CHF2, -CF3, or -CH2CHF2.
  • Ring A is heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1 -4 substituents independently selected from the group consisting of R c and oxo.
  • Ring A is bicyclic heteroaryl including from 9-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 substituents independently selected from the group consisting of R c and oxo.
  • Ring A is selected from the group consisting of: each of which is further optionally substituted with R c .
  • the compound is a compound of Formula (I-a): or a pharmaceutically acceptable salt thereof, wherein: each R cA is an independently selected R c ; n is 0, 1, or 2; and
  • Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA . each further optionally substituted with R cA , wherein each R cA is an independently selected R c .
  • each R cA is an independently selected R c .
  • R cA is an independently selected R c .
  • each R cA is an independently selected R c .
  • each occurrence of R cA is independently selected from the group consisting of: halo; NR e R f ; C1-4 alkoxy; C1- h4aloalkoxy; C1-3 alkyl; C1-3 alkyl substituted with from 1-3 independently selected halo; C1-3 alkyl substituted with C1-4 alkoxy; and C1-4 alkoxy substituted with C1-4 alkoxy; such as wherein each occurrence of R cA is independently selected from the group consisting of: C1-4 alkoxy; C1-4 haloalkoxy; C1-3 alkyl; and C1-3 alkyl substituted with from 1-3 independently selected halo.
  • Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • each further optionally substituted with R cA selected from the group consisting each further optionally substituted with R cA , wherein each R cA is an independently selected
  • R c In certain of these embodiments, selected from the group consisting each further optionally substituted with R cA , wherein each R cA is an independently selected R c .
  • the compound is a compound of Formula (I-b): Formula (I-b) or a pharmaceutically acceptable salt thereof, wherein: each R cA is an independently selected R c ; n is 0 or 1 ; and
  • Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • the compound is a compound of Formula (I-c): Formula (I-c) or a pharmaceutically acceptable salt thereof; wherein n is 0, 1, or 2; each R CA is an independently selected R c ; and
  • Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • each further optionally substituted with R cA selected from the group consisting each further optionally substituted with R cA , wherein each R cA is an independently selected R c .
  • each R cA is an independently selected R c .
  • Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA . In certain of these embodiments, selected from the group consisting optionally substituted with R cA , wherein each R cA is an independently selected R c .
  • X 1 Formula (I-d) or a pharmaceutically acceptable salt thereof wherein n is 0 or 1 ; each R CA is an independently selected R c ; and Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • R CA In certain embodiments of Formula selected from the group each further optionally substituted with R CA , wherein each R cA is an independently selected R c .
  • Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 R cA .
  • the compound of Formula (I) is a compound of Formula
  • Ring D is heterocyclylene including from 3-10 ring atoms, wherein from 0-2 ring atoms (in addition to the ring nitrogen atom bonded to R w ) are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclylene is optionally substituted with from 1-3 substituents each independently selected from the group consisting of: oxo and -R c .
  • W is C2-6 alkenyl optionally substituted with from 1-3 R a and further optionally substituted with R g , wherein W is attached to L w via an sp 2 hybridized carbon atom.
  • W is C2-6 alkenyl or C2-6 alkynyl optionally substituted with from 1-3 R a and further optionally substituted with R g , wherein W is attached to L w via an sp 2 or sp hybridized carbon atom.
  • xl is 0.
  • Ring D can be selected
  • Ring D can be any organic radical
  • heterocyclylene bonded to R w including from 3-10 ring atoms, wherein from 0-2 ring atoms (in addition to the ring nitrogen atom bonded to R w ) are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclylene is optionally substituted with from 1-3 substituents each independently selected from the group consisting of: oxo and -R c ; optionally wherein is a monocyclic heterocyclylene ring including from 3-10 ring atoms as defined above with a nitrogen atom bonded to R w (e.g., , such , such as heterocyclylene ring including from 3-10 ring atoms as defined above with a nitrogen atom
  • n 0.
  • n is 1 or 2, such as wherein n is 1.
  • R 7 is NR e R f , such as NH 2 , NH(CI- 3 alkyl), or N(CI-3 alkyl) 2 , such as wherein R 7 is NH 2 .
  • one occurrence ofR cA is -NH 2 .
  • one occurrence of R CA is -NH(CI-6 alkyl), wherein the C1-6 alkyl is optionally substituted with from 1-3 substituents each independently selected from the group consisting of NR’R”, -OH, C1-6 alkoxy, C1-6 haloalkoxy, and halo.
  • R cA can be -NHMe, - NHCH2CF3, -NHCH2CH2OH, or -NHiPr.
  • one occurrence of R CA is C1-4 alkoxy optionally substituted with C1-4 alkoxy or C1-4 haloalkoxy.
  • one occurrence of R cA can be OMe or OCFbCFFOMe.
  • R cA can be C1-4 haloalkoxy, such as -OCH2CF3.
  • R CA is C1-4 thioalkoxy (e.g., SCH3).
  • one occurrence of R CA is C1-6 alkyl, such as methyl; or wherein one occurrence of R cA is C1-6 alkyl substituted with from 1-6 independently selected halo (e.g., R cA can be -CF3).
  • R cA can be , or
  • R CA is halo (e.g., -F).
  • one occurrence ofR cA is -OH.
  • X 1 can be as defined anywhere herein. In certain embodiments, X 1 can be as defined in [AA1], [BB1], [CC1], [DD1], [EE1], or [FF1], infra.
  • X 1 is -(X 2 ) m - L 1 -R 5 , wherein:
  • X 2 is -N(R N )- or -O-;
  • L 1 is a bond or C1-6 alkylene optionally substituted with from 1-3 R a ;
  • R 5 is phenyl optionally substituted with from 1 - 4 R c , such as wherein R 5 is phenyl optionally substituted with from 1-2 independently selected halo, such as -F.
  • R 5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(R d ), and wherein the heteroaryl is optionally substituted with from 1 -4 R c , such as wherein
  • R 5 is heteroaryl including 5 ring atoms, wherein from 1-4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 R c , such as wherein
  • R 5 is C3-10 cycloalkyl, such as C3-6 cycloalkyl, optionally substituted with from 1-4 R c , such as wherein R 5 is cyclopropyl.
  • R 5 is heterocyclyl including from 4-8, such as 4-
  • R 5 can
  • X 2 is -N(R N )- (e.g., N(H)).
  • X 2 is -O-.
  • L 1 is a bond.
  • L 1 is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or - CH(Me)-).
  • L 1 is branched C3-6 alkylene.
  • L 1 can be wherein aa is the point of attachment to R 5 .
  • X 1 is -X 2 -L 1 - R 5 , wherein:
  • L 1 is a bond or C1-6 alkylene optionally substituted with from 1-3 R a ;
  • R 5 is phenyl optionally substituted with from 1 -
  • R c such as wherein R 5 is phenyl optionally substituted with from 1-2 independently selected halo, such as -F.
  • R 5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(R d ), and wherein the heteroaryl is optionally substituted with from 1 -4 R c , such as wherein
  • R 5 is heteroaryl including 5 ring atoms, wherein from 1-4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 R c , such as wherein
  • R 5 is C3-10 cycloalkyl, such as C3-6 cycloalkyl, optionally substituted with from 1-4 R c , such as wherein R 5 is cyclopropyl.
  • R 5 is heterocyclyl including from 4-8, such as 4-
  • R 5 can
  • X 2 is -N(R N )S(O)2-, such as -N(H)S(O)2-*.
  • L 1 is a bond
  • L 1 is Ci-3 alkylene (e g., -CH 2 -, -CH2CH2-, or -
  • L 1 is branched C3-6 alkylene.
  • L 1 can be wherein aa is the point of attachment to R 5 .
  • X 1 is -X 2 -L 1 -
  • L 1 is a bond or C1-6 alkylene optionally substituted with from 1-3 R a ;
  • R 5 is - R"
  • R 5 is phenyl optionally substituted with from 1 - 4 R c , such as wherein R 5 is phenyl optionally substituted with from 1-2 independently selected halo, such as -F.
  • R 5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(R d ), and wherein the heteroaryl is optionally substituted with
  • R 5 is heteroaryl including 5 ring atoms, wherein from 1-4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 R c , such as wherein
  • R 5 is C3-10 cycloalkyl, such as C3-6 cycloalkyl, optionally substituted with from 1-4 R c , such as wherein R 5 is cyclopropyl.
  • R 5 is heterocyclyl including from 4-8, such as 4-
  • R 5 can
  • L 1 is a bond
  • L 1 is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or - CH(Me)-).
  • L 1 is branched C3-6 alkylene.
  • L 1 can be wherein aa is the point of attachment to R 5 .
  • X 1 is -(X 2 )m- L'-R 5 , wherein:
  • X 2 is -N(R N )- or -O-;
  • L 1 is a bond or C1-6 alkylene optionally substituted with from 1-3 R a ;
  • R 5 is R" 2 -R v
  • the -R g2 group present in R 5 is 1,3 -phenylene or
  • 1,4-phenylene each optionally substituted with from 1-4 R c , such as wherein -R g2 is wherein bb is the point of attachment to R Y .
  • the R Y group present in R 5 is -R g .
  • the R Y group present in R 5 is heterocyclyl including from 4-8, such as 4-6, ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c .
  • R Y can be
  • X 2 is -N(R N )- (e.g., N(H)).
  • X 2 is -O-.
  • L 1 is a bond
  • L 1 is Ci-3 alkylene (e g., -CH 2 -, -CH2CH2-, or -
  • L 1 is branched C3-6 alkylene.
  • L 1 can be , wherein aa is the point of attachment to R 5 .
  • X 1 is X 2 -L'-
  • L 1 is C1-6 alkylene optionally substituted with from 1-3 R a ;
  • R 5 is H, halo, C1-6 alkoxy optionally substituted with from 1-3 R a , or -OH.
  • R 5 is H.
  • R 5 is halo (e.g., -F).
  • R 5 is C1-6 alkoxy optionally substituted with from 1-3 R a , such as wherein R 5 is C1-3 alkoxy such as methoxy.
  • R 5 is -OH.
  • X 2 is -N(R N )- (e.g., N(H)).
  • X 2 is -O-.
  • X 2 is -N(R N )S(O)2-, such as -N(H)S(O)2-*.
  • L 1 is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or - CH(Me)-).
  • L 1 is branched C3-6 alkylene.
  • L 1 can be wherein aa is the point of attachment to R 5 .
  • X 1 is -L ⁇ R 5 , wherein L 1 is C1-6 alkylene optionally substituted with from 1-3 R a ; and R 5 is -L 5 -R g .
  • R 5 is -O-R g .
  • R 5 is -O-(phenyl), wherein the phenyl is optionally substituted with from 1-2 R c .
  • L 1 is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or - CH(Me)-).
  • R la is H.
  • R 2a and R 2b are both H.
  • R 2a is a substituent that is other than H.
  • R 2a is C1-6 alkyl which is optionally substituted with from 1-6 R a , such as wherein R 2a is C1-3 alkyl, such as methyl or ethyl.
  • R 2b is H.
  • R 3a and R 3b are both H.
  • R 3a is a substituent that is other than H.
  • R 3a is C1-6 alkyl which is optionally substituted with from 1-6 R a , such as wherein R 3a is C1-3 alkyl, such as methyl or ethyl.
  • R 3a is C1-3 alkyl substituted with from 1-3 independently selected halo.
  • R 3a is -CH2F, -CHF2, -CF3, -CH2CHF2, or - CH2CH2F.
  • R 3a is C1-3 alkyl substituted with C1-4 alkoxy, C1-4 haloalkoxy, or NR e R f .
  • R 3a in these embodiments include -CFFOMe, -CHoCHoOMe, -CH(Me)CH20Me, CH 2 CH(Me)OMe, -CH 2 OEt, -CH2CH2OCHF2, -CH 2 NR e R f (e g., -CH 2 N(CF 3 )Me), or - CH 2 CH2NR e R f (e g., -CH 2 CH 2 NMe2).
  • R 3a is C1-3 alkyl substituted with C1-4 alkoxy, C1-4 haloalkoxy, or NR e R f and further substituted with from 1-3 independently selected halo. In certain of these embodiments, R 3a is C1-3 alkyl substituted with C1-4 alkoxy and further substituted with from 1-3 independently selected
  • Non-limiting examples of R 3a in these embodiments include: -J- (e.g.,
  • R 3a is selected from the group consisting of: heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and R c ; and
  • R 3a is — (C1-3 alkylene)-R g or -(C1-3 alkylene)-O-R g , and optionally the R g group of R 3a is:
  • R 3a is -CH2-R g , or - CtFCFFR 8 , wherein R g is 1,4-dioxanyl.
  • R 3a is-(L g ) g -R w .
  • R 3a is -(L g ) g -
  • R 3a is -CH2-R g2 -
  • R 3b is H.
  • R 3b is C1-3 alkyl.
  • R 3b is methyl, ethyl, or propyl.
  • R 3b is methyl.
  • R 3b is H.
  • R 3b is halo.
  • R 3b can be -F.
  • R 3a and R 3b together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-8 ring atoms;
  • each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(R d ), O, and S(0)o-2;
  • R 3a and R 3b together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-6 ring atoms; • wherein from 1 -2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(R d ), O, and S(0)o-2; and
  • R 3a and R 3b together with the Ring B ring atom to which each is attached, form a fused C3-6 cycloalkyl, wherein the fused C3-6 cycloalkyl is optionally substituted with from 1-2 R c .
  • R 3a and R 3b together with the Ring B ring atom to which each is attached, form
  • R 3a and R 3b together with the Ring B ring atom to which each is attached, form: , which is optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and R c , wherein: pl and p2 are independently 0, 1, or 2;
  • R 3a and R 3b together with the Ring B ring atom to which each is attached, form a fused ring selected
  • R la , R lb , R 2a , and R 2b are each H; and R 3a and R 3b taken together with the Ring B ring carbon atom to which each is attached form a fused C3-6 (such as C3 or C4) cycloalkyl, wherein the fused cycloalkyl ring is optionally substituted with from 1 -2 R c .
  • R la , R lb , R 2a , and R 2b are each H; and R 3a and R 3b together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-6 ring atoms;
  • R 2a and R 2b such as R 2a
  • R 3a and R 3b taken together with the Ring B ring atoms to which each is attached, form a fused saturated or unsaturated ring of 3-12 ring atoms
  • each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(R d ), O, and S(0)o-2;
  • R 2a and R 2b taken together with the Ring B ring atoms to which each is attached, form a fused saturated ring of 3-8 ring atoms;
  • each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(R d ), O, and S(0)o-2;
  • one of R 2a and R 2b (such as R 2a ) and one of R 3a and R 3b (such as R 3a ) taken together with the Ring B ring atoms to which each is attached form a fused C3-6 cycloalkyl which is optionally substituted with from 1-2 R c .
  • one of R 2a and R 2b (such as R 2a ) and one of R 3a and R 3b (such as R 3a ) taken together with the Ring B ring atoms to which each is attached form a fused cyclopropyl or cyclobutyl ring, e.g.,
  • R la , R lb is H; and R 2b is H; and R 3b is -(L g ) g -R g .
  • R la , R lb is H;
  • R la , R lb is H
  • R 2b and R 3b are each
  • R 2b and R 3b are each H.
  • R la , R lb , R 2a , and R 2b are each H, and R 3a is C1-3 alkyl optionally substituted with from 1-3 R a .
  • R la , R lb , R 2a , and R 2b are each H; R 3a , is C1-3 alkyl optionally substituted with from 1-3 R a ; and R 3b is H, optionally each R a substituent present in R 3a is independently selected from the group consisting of: halo, C1-4 alkoxy, and C1-4 haloalkoxy.
  • R la , R lb , R 2a , and R 2b are each H; and R 3a and R 3b are independently selected C1-3 alkyl.
  • R la , R lb is H; R 2a and R 3a taken together with the Ring B ring atoms to which each is attached, form a fused C3-6 (e.g., C3 or C4) cycloalkyl which is optionally substituted with from 1-2 R c ; and R 2b and R 3b are each H.
  • R la , R lb , R 2a , R 2b , R 3a , and R 3b are each H.
  • R 3b is H, and each optionally present R a substituent in R 3a is independently selected from the group consisting of: halo, C1-4 alkoxy, and Ci -4 haloalkoxy.
  • R 3b is -F, and each optionally present R a substituent in R 3a is independently selected from the group consisting of: halo, C1-4 alkoxy, and CM haloalkoxy.
  • R 3b is C1-3 alkyl (e.g., methyl), and each optionally present R a substituent in R 3a is independently selected from the group consisting of: halo, Ci -4 alkoxy, and C1-4 haloalkoxy.
  • R la , R lb , R 2a , and R 2b are each H;
  • R 3a is -R g , -(C1-3 alkylene)-R g , or -(C1-3 alkylene)-O-R g , optionally wherein the R g group of R 3a is:
  • R 3b is H.
  • R la , R lb , R 2a , and R 2b are each H; and R 3a is C1-3 alkyl optionally substituted with from 1-3 R a ; and R 3b is H, optionally each R a substituent present in R 3a is independently selected from the group consisting of: halo, C1-4 alkoxy, and C1-4 haloalkoxy.
  • R la , R lb , R 2a , and R 2b are each H and R 3a , is -R g , -(C1-3 alkylene)-R g , or -(C1-3 alkylene)-O-R g , optionally wherein the R g group of R 3a is:
  • R 3b is H.
  • R la , R lb , R 2a , and R 2b are each H, and R 3a and R 3b taken together with the Ring B ring carbon atom to which each is attached form a fused C3-6 (such as C3 or C4) cycloalkyl, wherein the fused cycloalkyl ring is optionally substituted with from 1 -2 R c .
  • R la , R lb , R 2a , and R 2b are each H, and R 3a and R 3b together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-6 ring atoms;
  • R la , R lb is H
  • R 2a and R 3a taken together with the Ring B ring atoms to which each is attached, form a fused C3-6 (e.g., C3 or C4) cycloalkyl which is optionally substituted with from 1-2 R c , and R 2b and R 3b are each H.
  • R la , R lb , R 2a , R 2b , R 3a , and R 3b are each H.
  • R 4 is H.
  • Ring A is
  • R cB (R cB )mi , wherein each R cB is an independently selected R c ; and ml is 0, 1, 2, 3, or 4.
  • ml is 1, 2, or 3, such as 1 or 2.
  • Ring A is wherein each R cB is an independently selected R c .
  • Ring A is selected from the group consisting wherein each R cB is an independently selected R c .
  • each R cB is independently selected from the group consisting of: -halo, such as -Cl and -F; -CN; C1-4 alkoxy; C1-4 haloalkoxy; C1-3 alkyl; and C1-3 alkyl substituted with from 1-6 independently selected halo.
  • Ring A is bicyclic heteroaryl including from 9-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(R d ), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 substituents independently selected from the group consisting of R c and oxo, such as wherein: Ring A each of which is further optionally substituted with R c .
  • the compound is selected from the group consisting of the compounds delineated in Table Cl, or a pharmaceutically acceptable salt thereof.
  • the symbol * at a chiral center denotes that this chiral center has been resolved (i.e., is a single epimer) and the absolute stereochemistry at that center has not been determined.
  • a chemical entity e.g., a compound that inhibits EGFR and/or HER2, or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination thereof
  • a pharmaceutical composition that includes the chemical entity and one or more pharmaceutically acceptable excipients, and optionally one or more additional therapeutic agents as described herein.
  • the chemical entities can be administered in combination with one or more conventional pharmaceutical excipients.
  • Pharmaceutically acceptable excipients include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-a-tocopherol polyethylene glycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens, poloxamers or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, tris, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium-chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium, sodium
  • Cyclodextrins such as a-, 0, and y-cyclodextrin, or chemically modified derivatives such as hydroxyalkyl cyclodextrins, including 2- and 3- hydroxypropyl-0-cyclodextrins, or other solubilized derivatives can also be used to enhance delivery of compounds described herein.
  • Dosage forms or compositions containing a chemical entity as described herein in the range of 0.005% to 100% with the balance made up from non-toxic excipient may be prepared.
  • the contemplated compositions may contain 0.001%- 100% of a chemical entity provided herein, in one embodiment 0.1-95%, in another embodiment 75-85%, in a further embodiment 20-80%.
  • Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 22 nd Edition (Pharmaceutical Press, London, UK. 2012).
  • the chemical entities described herein or a pharmaceutical composition thereof can be administered to subject in need thereof by any accepted route of administration.
  • Acceptable routes of administration include, but are not limited to, buccal, cutaneous, endocervical, endosinusial, endotracheal, enteral, epidural, interstitial, intra-abdominal, intra-arterial, intrabronchial, intrabursal, intracerebral, intracisternal, intracoronary, intradermal, intraductal, intraduodenal, intradural, intraepidermal, intraesophageal, intragastric, intragingival, intraileal, intralymphatic, intramedullary, intrameningeal, intramuscular, intraovarian, intraperitoneal, intraprostatic, intrapulmonary, intrasinal, intraspinal, intrasynovial, intratesticular, intrathecal, intratubular, intratumoral, intrauterine, intravascular, intravenous, nasal, nasogastric
  • compositions can be formulated for parenteral administration, e.g., formulated for injection via the intravenous, intramuscular, sub-cutaneous, or even intraperitoneal routes.
  • parenteral administration e.g., formulated for injection via the intravenous, intramuscular, sub-cutaneous, or even intraperitoneal routes.
  • such compositions can be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for use to prepare solutions or suspensions upon the addition of a liquid prior to injection can also be prepared; and the preparations can also be emulsified.
  • injectables either as liquid solutions or suspensions
  • solid forms suitable for use to prepare solutions or suspensions upon the addition of a liquid prior to injection can also be prepared; and the preparations can also be emulsified.
  • the preparation of such formulations will be known to those of skill in the art in light of the present disclosure.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil, or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that it may be easily injected. It also should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier also can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques, which yield a powder of the active ingredient, plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Intratumoral injections are discussed, e.g., in Lammers, et al., “Effect of Intratumoral Injection on the Biodistribution and the Therapeutic Potential of HPMA Copolymer-Based Drug Delivery Systems” Neoplasia. 2006, 10, 788-795.
  • Pharmacologically acceptable excipients usable in the rectal composition as a gel, cream, enema, or rectal suppository include, without limitation, any one or more of cocoa butter glycerides, synthetic polymers such as polyvinylpyrrolidone, PEG (like PEG ointments), glycerine, glycerinated gelatin, hydrogenated vegetable oils, poloxamers, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol Vaseline, anhydrous lanolin, shark liver oil, sodium saccharinate, menthol, sweet almond oil, sorbitol, sodium benzoate, anoxid SBN, vanilla essential oil, aerosol, parabens in phenoxyethanol, sodium methyl p-oxybenzoate, sodium propyl p- oxybenzoate, diethylamine, carbomers, carbopol, methyloxybenzoate, macrogol cetostearyl ether, cocoyl caprylo
  • suppositories can be prepared by mixing the chemical entities described herein with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum and release the active compound.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum and release the active compound.
  • compositions for rectal administration are in the form of an enema.
  • the compounds described herein or a pharmaceutical composition thereof are suitable for local delivery to the digestive or GI tract by way of oral administration (e.g., solid or liquid dosage forms.).
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the chemical entity is mixed with one or more pharmaceutically acceptable excipients, such as sodium citrate or dicalcium phosphate and/or: a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol mono
  • the dosage form may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the compositions will take the form of a unit dosage form such as a pill or tablet and thus the composition may contain, along with a chemical entity provided herein, a diluent such as lactose, sucrose, dicalcium phosphate, or the like; a lubricant such as magnesium stearate or the like; and a binder such as starch, gum acacia, polyvinylpyrrolidine, gelatin, cellulose, cellulose derivatives or the like.
  • a diluent such as lactose, sucrose, dicalcium phosphate, or the like
  • a lubricant such as magnesium stearate or the like
  • a binder such as starch, gum acacia, polyvinylpyrrolidine, gelatin, cellulose, cellulose derivatives or the like.
  • a powder, marume, solution or suspension (e.g., in propylene carbonate, vegetable oils, PEG’s, poloxamer 124 or triglycerides) is encapsulated in a capsule (gelatin or cellulose base capsule).
  • Unit dosage forms in which one or more chemical entities provided herein or additional active agents are physically separated are also contemplated; e.g., capsules with granules (or tablets in a capsule) of each drug; two-layer tablets; two- compartment gel caps, etc. Enteric coated or delayed release oral dosage forms are also contemplated.
  • physiologically acceptable compounds include wetting agents, emulsifying agents, dispersing agents or preservatives that are particularly useful for preventing the growth or action of microorganisms.
  • Various preservatives are well known and include, for example, phenol and ascorbic acid.
  • the excipients are sterile and generally free of undesirable matter. These compositions can be sterilized by conventional, well-known sterilization techniques. For various oral dosage form excipients such as tablets and capsules sterility is not required. The USP/NF standard is usually sufficient.
  • solid oral dosage forms can further include one or more components that chemically and/or structurally predispose the composition for delivery of the chemical entity to the stomach or the lower GI; e.g., the ascending colon and/or transverse colon and/or distal colon and/or small bowel.
  • Exemplary formulation techniques are described in, e.g., Filipski, K.J., et al., Current Topics in Medicinal Chemistry, 2013, 13, 776-802, which is incorporated herein by reference in its entirety.
  • Examples include upper-GI targeting techniques, e.g., Accordion Pill (Intec Pharma), floating capsules, and materials capable of adhering to mucosal walls.
  • Upper-GI targeting techniques e.g., Accordion Pill (Intec Pharma)
  • floating capsules e.g., floating capsules, and materials capable of adhering to mucosal walls.
  • enteric/pH-responsive coatings and excipients are available. These materials are typically polymers that are designed to dissolve or erode at specific pH ranges, selected based upon the GI region of desired drug release. These materials also function to protect acid labile drugs from gastric fluid or limit exposure in cases where the active ingredient may be irritating to the upper GI (e.g., hydroxypropyl methylcellulose phthalate series, Coateric (polyvinyl acetate phthalate), cellulose acetate phthalate, hydroxypropyl methylcellulose acetate succinate, Eudragit series (methacrylic acid-methyl methacrylate copolymers), and Marcoat).
  • Other techniques include dosage forms that respond to local flora in the GI tract, Pressure-controlled colon delivery capsule, and Pulsincap.
  • Ocular compositions can include, without limitation, one or more of any of the following: viscogens (e.g., Carboxymethylcellulose, Glycerin, Polyvinylpyrrolidone, Polyethylene glycol); Stabilizers (e.g., Pluronic (triblock copolymers), Cyclodextrins); Preservatives (e.g., Benzalkonium chloride, ETDA, SofZia (boric acid, propylene glycol, sorbitol, and zinc chloride; Alcon Laboratories, Inc.), Purite (stabilized oxy chloro complex; Allergan, Inc.)).
  • viscogens e.g., Carboxymethylcellulose, Glycerin, Polyvinylpyrrolidone, Polyethylene glycol
  • Stabilizers e.g., Pluronic (triblock copolymers), Cyclodextrins
  • Preservatives e.g., Benzalkonium chloride, ETDA, SofZi
  • Topical compositions can include ointments and creams.
  • Ointments are semisolid preparations that are typically based on petrolatum or other petroleum derivatives.
  • Creams containing the selected active agent are typically viscous liquid or semisolid emulsions, often either oil-in-water or water-in-oil.
  • Cream bases are typically water-washable, and contain an oil phase, an emulsifier and an aqueous phase.
  • the oil phase also sometimes called the “internal” phase, is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol; the aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant.
  • the emulsifier in a cream formulation is generally a nonionic, anionic, cationic or amphoteric surfactant.
  • an ointment base should be inert, stable, nonirritating and nonsensitizing.
  • compositions described herein can include one or more one or more of the following: lipids, interbilayer crosslinked multilamellar vesicles, biodegradeable poly(D,L-lactic-co-glycolic acid) [PLGA]-based or poly anhydride-based nanoparticles or microparticles, and nanoporous particle-supported lipid bilayers.
  • lipids interbilayer crosslinked multilamellar vesicles
  • biodegradeable poly(D,L-lactic-co-glycolic acid) [PLGA]-based or poly anhydride-based nanoparticles or microparticles and nanoporous particle-supported lipid bilayers.
  • the dosages may be varied depending on the requirement of the patient, the severity of the condition being treating and the particular compound being employed. Determination of the proper dosage for a particular situation can be determined by one skilled in the medical arts.
  • the total daily dosage may be divided and administered in portions throughout the day or by means providing continuous delivery.
  • the compounds described herein are administered at a dosage of from about 0.001 mg/Kg to about 500 mg/Kg (e.g., from about 0.001 mg/Kg to about 200 mg/Kg; from about 0.01 mg/Kg to about 200 mg/Kg; from about 0.01 mg/Kg to about 150 mg/Kg; from about 0.01 mg/Kg to about 100 mg/Kg; from about 0.01 mg/Kg to about 50 mg/Kg; from about 0.01 mg/Kg to about 10 mg/Kg; from about 0.01 mg/Kg to about 5 mg/Kg; from about 0.01 mg/Kg to about 1 mg/Kg; from about 0.01 mg/Kg to about 0.5 mg/Kg; from about 0.01 mg/Kg to about 0.1 mg/Kg; from about 0.
  • the foregoing dosages can be administered on a daily basis (e.g., as a single dose or as two or more divided doses) or non-daily basis (e.g., every other day, every two days, every three days, once weekly, twice weeks, once every two weeks, once a month).
  • a daily basis e.g., as a single dose or as two or more divided doses
  • non-daily basis e.g., every other day, every two days, every three days, once weekly, twice weeks, once every two weeks, once a month.
  • the period of administration of a compound described herein is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 1 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 1 1 months, 12 months, or more.
  • a period of during which administration is stopped is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 1 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 1 1 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 1 1 months, 12 months, or more.
  • a therapeutic compound is administered to an individual for a period of time followed by a separate period of time.
  • a therapeutic compound is administered for a first period and a second period following the first period, with administration stopped during the second period, followed by a third period where administration of the therapeutic compound is started and then a fourth period following the third period where administration is stopped.
  • the period of administration of a therapeutic compound followed by a period where administration is stopped is repeated for a determined or undetermined period of time.
  • a period of administration is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more.
  • a period of during which administration is stopped is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more.
  • EGFR epidermal growth factor receptor tyrosine kinase
  • HER2 human epidermal growth factor receptor 2
  • inhibitors of EGFR useful for treating or preventing diseases or disorders associated with dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same (i.e., an EGFR-associated disease or disorder), such as a central nervous system diseases, a pulmonary disorder, cardiovascular disease, ischemia, liver disease, a gastrointestinal disorder, a viral or bacterial infection, an inflammatory and/or autoimmune disease, or cancer (e.g., EGFR-associated cancer).
  • inhibitors of HER2 useful for treating or preventing diseases or disorders associated with dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, such as cancer (e.g., HER2- associated cancer).
  • cancer e.g., HER2- associated cancer
  • an “EGFR inhibitor” as used herein includes any compound exhibiting EGFR inactivation activity (e.g., inhibiting or decreasing).
  • an EGFR inhibitor can be selective for an EGFR kinase having one or more mutations.
  • an EGFR inhibitor can bind to the adenosine triphosphate (ATP)-binding site in the tyrosine kinase domain.
  • an EGFR inhibitor is an allosteric inhibitor.
  • the compounds provided herein can inhibit EGFR.
  • the compounds can bind to the EGFR adenosine triphosphate (ATP)-binding site in the tyrosine kinase domain.
  • ATP adenosine triphosphate
  • test compounds to act as inhibitors of EGFR may be demonstrated by assays known in the art.
  • the activity of the compounds and compositions provided herein as EGFR inhibitors can be assayed in vitro, in vivo, or in a cell line.
  • In vitro assays include assays that determine inhibition of the kinase and/or ATPase activity.
  • Alternate in vitro assays quantitate the ability of the inhibitor to bind to the protein kinase and can be measured either by radio labelling the compound prior to binding, isolating the compound/kinase complex and determining the amount of radio label bound, or by running a competition experiment where new compounds are incubated with the kinase bound to known radioligands.
  • an EGFR inhibitor can be evaluated by its effect on the initial velocity of EGFR tyrosine kinase catalyzed peptide phosphorylation (e.g., Yun et al. Cancer Cell. 2007;l l(3):217-227).
  • the binding constant of an EGFR inhibitor can be determined using fluorescence kinetics (e.g., Yun et al. Cancer Cell. 2007;l l(3):217-227).
  • SPR surface plasmon resonance
  • Assays can include, for example, proliferation inhibition assays such as those that measure cell growth inhibition, such as an MTS assay or by Cell Titer Gio Luminescent Cell viability assay (Promega®).
  • proliferation inhibition assays such as those that measure cell growth inhibition, such as an MTS assay or by Cell Titer Gio Luminescent Cell viability assay (Promega®).
  • MTS assay or by Cell Titer Gio Luminescent Cell viability assay (Promega®).
  • MTS assay assay or by Cell Titer Gio Luminescent Cell viability assay (Promega®).
  • MTS assay assay
  • Cell Titer Gio Luminescent Cell viability assay Promega®
  • a Western Blot analysis can be used. In such assays cells are seeded and grown in culture plates and then treated with a test compound the following day for varying durations.
  • Additional assays can include, for example, assays based on ALPHALISA TECHNOLOGY® (e g., see the ALPHALISA® EGF/EGFR binding kit from Promega). Such assays use a luminescent oxygen-channeling chemistry to detect molecules of interest in, for example, buffer, cell culture media, serum, and plasma. For example, a biotinylated EGF is bound to streptavidin-coated Alpha donor beads, and EGFR-Fc is captured by antihuman IgG Fc-specific AlphaLISA acceptor beads.
  • donor beads and acceptor beads come into close proximity, and the excitation of the donor beads provokes the release of singlet oxygen molecules that triggers a cascade of energy transfers in the acceptor beads. This results in a sharp peak of light emission at 615 nm.
  • assays can be used, for example, in competitive binding experiments.
  • assays can include assays based on Sox technology (e.g., see the PHOSPHOSENS® Sox-based Homogeneous, Kinetic or Endpoint/Red Fluorescencebased Assays from ASSAYQUANT®).
  • Sox chelation-enhanced fluorescence
  • Sox sulfonamido-oxine
  • Potency of an EGFR inhibitor as provided herein can be determined by ECso value.
  • a compound with a lower ECso value, as determined under substantially similar conditions, is a more potent inhibitor relative to a compound with a higher ECso value.
  • the substantially similar conditions comprise determining an EGFR- dependent phosphorylation level, in vitro or in vivo (e.g., in tumor cells, A431 cells, Ba/F3 cells, or 3T3 cells cells expressing a wild type EGFR, a mutant EGFR, or a fragment of any thereof).
  • Potency of an EGFR inhibitor as provided herein can also be determined by IC50 value.
  • a compound with a lower IC50 value, as determined under substantially similar conditions, is a more potent inhibitor relative to a compound with a higher IC50 value.
  • the substantially similar conditions comprise determining an EGFR- dependent phosphorylation level, in vitro or in vivo (e.g., in tumor cells, A431 cells, Ba/F3 cells, or 3T3 cells expressing a wild type EGFR, a mutant EGFR, or a fragment of any thereof).
  • the selectivity between wild type EGFR and EGFR containing one or more mutations as described herein can also be measured using cellular proliferation assays where cell proliferation is dependent on kinase activity.
  • murine Ba/F3 cells transfected with a suitable version of wild type EGFR such as VIII; containing a wild type EGFR kinase domain
  • H773_V774insX e.g., A767_V769dupASV, V769_D770insASV, D770_N771insNPG, D770_N771insNPY, D770_N771insSVD, D770_N771insGL, N771_H773dupNPH, N771_P772insN, N771_P772insH, N771_P772insV, P772_H773insDNP,
  • H773_V774insAH, or P772_H773insPNP can be used.
  • Proliferation assays are performed at a range of inhibitor concentrations (e.g., 10 pM, 3 pM, 1.1 pM, 330 nM, 110 nM, 33 nM, 11 nM, 3 nM, 1 nM) and an ECso is calculated.
  • An alternative method to measure effects on EGFR activity is to assay EGFR phosphorylation. Wildtype or mutant (L858R/T790M, Del/T790M, Del/T790M/L718Q, L858R/T790M/C797S, Del/T790M/C797S, L858R/T790M/I941R, or
  • EGFR can be transfected into cells which do not normally express endogenous EGFR and the ability of the inhibitor (e.g., using concentrations as above) to inhibit EGFR phosphorylation can be assayed.
  • Cells are exposed to increasing concentrations of inhibitor and stimulated with EGF.
  • the effects on EGFR phosphorylation are assayed by Western Blotting using phospho-specific EGFR antibodies.
  • the compounds provided herein can exhibit potent and selective inhibition of EGFR.
  • the compounds provided herein can bind to the EGFR adenosine triphosphate (ATP)-binding site in the tyrosine kinase domain.
  • the compounds provided herein can exhibit nanomolar potency against an EGFR kinase including an activating mutation or an EGFR inhibitor resistance mutation, including, for example, the resistance mutations in Table 2a and 2b (e.g., L747S, D761Y, T790M, and T854A), with minimal activity against related kinases (e.g., wild type EGFR).
  • Inhibition of wild type EGFR can cause undesireable side effects (e.g., diarrhea and skin rashes) that can impact quality of life and compliance.
  • the inhibititon of wild type EGFR can lead to dose limiting toxicities. See, e.g., Morphy. J. Med. Chem. 2010, 53, 4, 1413-1437 and Peters. J. Med. Chem. 2013, 56, 22, 8955-8971.
  • the compounds of Formula (I) can selectively target an EGFR kinase.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a pharmaceutically acceptable salt thereof can selectively target an EGFR kinase over another kinase or non-kinase target.
  • a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I- c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, can exhibit greater inhibition of EGFR containing one or more mutations as described herein (e.g., one or more mutations as described in Table la and lb) relative to inhibition of wild type EGFR.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit at least 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or 100-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit up to 1000-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit up to 10000-fold greater inhibition of EGFR having a combination of mutations described herein relative to inhibition of wild type EGFR.
  • a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I- c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, can exhibit from about 2- fold to about 10-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit from about 10-fold to about 100-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit from about 1 OO-fold to about 1 OOO-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit from about 1 OOO-fold to about 1 OOOO-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a pharmaceutically acceptable salt thereof in combination with a second EGFR inhibitor can exhibit greater inhibition of EGFR containing one or more mutations as described herein (e.g., one or more mutations as described in Table la and lb) relative to inhibition of wild type EGFR.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit at least 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or 100-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit up to 1000-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit up to 1 OOOO-fold greater inhibition of EGFR having a combination of mutations described herein relative to inhibition of wild type EGFR.
  • a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit from about 2-fold to about 10-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit from about 10-fold to about 100-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit from about 100-fold to about 1000-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit from about 1000-fold to about lOOOO-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR.
  • Compounds of Formula (I) are useful for treating diseases and disorders which can be treated with an EGFR inhibitor, such as EGFR-associated diseases and disorders, e.g., central nervous system diseases (e.g., neurodegenerative diseases), pulmonary disorders, cardiovascular disease, ischemia, liver disease, gastrointestinal disorders, viral or bacterial infections, inflammatory and/or autoimmune diseases (e.g., psoriasis and atopic dermatitis), and proliferative disorders such as cancers, including hematological cancers and solid tumors (e.g., advanced solid tumors).
  • EGFR-associated diseases and disorders e.g., central nervous system diseases (e.g., neurodegenerative diseases), pulmonary disorders, cardiovascular disease, ischemia, liver disease, gastrointestinal disorders, viral or bacterial infections, inflammatory and/or autoimmune diseases (e.g., psoriasis and atopic dermatitis), and proliferative disorders such as cancers, including hematological cancers and solid tumors (e
  • a “HER2 inhibitor” as used herein includes any compound exhibiting HER2 inactivation activity (e.g., inhibiting or decreasing).
  • a HER2 inhibitor can be selective for a HER2 kinase having one or more mutations.
  • a HER2 inhibitor can bind to the HER2 adenosine triphosphate (ATP)- binding site in the tyrosine kinase domain.
  • ATP adenosine triphosphate
  • the compounds provided herein can inhibit HER2.
  • the compounds can bind to the HER2 adenosine triphosphate (ATP)-binding site in the tyrosine kinase domain.
  • the compounds provided herein can inhibit wild type HER2.
  • the compounds provided herein can inhibit HER2 having one or more mutations as described herein.
  • test compounds to act as inhibitors of HER2 may be demonstrated by assays known in the art.
  • the activity of the compounds or compositions provided herein as HER2 inhibitors can be assayed in vitro, in vivo, or in a cell line.
  • In vitro assays include assays that determine inhibition of the kinase and/or ATPase activity.
  • Alternate in vitro assays quantitate the ability of the inhibitor to bind to the protein kinase and can be measured either by radio labelling the compound prior to binding, isolating the compound/kinase complex and determining the amount of radio label bound, or by running a competition experiment where new compounds are incubated with the kinase bound to known radioligands.
  • a HER2 inhibitor can be evaluated by its effect on the initial velocity of HER2 tyrosine kinase catalyzed peptide phosphorylation (e.g., Yun et al. Cancer Cell. 2007;l l(3):217-227).
  • an assay that indirectly measures ADP formed from the HER2 kinase reaction can be used (see, e.g., ATP/NADH coupled assay systems and luminescent kinase assays such as ADP-GLOTM Kinase Assay from Promega). See, e.g., Hanker et al. Cancer Discov. 2017 Jun;7(6): 575-585; Robichaux et al. Nat Med.
  • an assay that detects substrate phosphorylation using a labeled anti-phospho-tyrosine antibody can be used (see, e.g., Rabindran et al. Cancer Res. 2004 Jun 1 ;64(11):3958-65).
  • the binding constant of a HER2 inhibitor can be determined using fluorescence kinetics (e.g., Yun et al. Cancer Cell. 2007;l l(3):217-227). Examples of SPR binding assays include those disclosed in Li, Shiqing, et al.
  • covalent binding of a HER2 inhibitor to HER2 can be detected using mass spectrometry, see, e.g., Irie et al. Mol Cancer Ther. 2019 Apr;18(4):733-742. Additional HER2 inhibitor assays can be found, for example, in U.S. Patent No. 9,920,060, WO 2019/241715, and U.S. Publication No. 2017/0166598, each of which are incorporated by reference in their entireties.
  • Potency of a HER2 inhibitor as provided herein can be determined by ECso value.
  • a compound with a lower ECso value, as determined under substantially similar conditions, is a more potent inhibitor relative to a compound with a higher ECso value.
  • the substantially similar conditions comprise determining an HER2- dependent phosphorylation level, in vitro or in vivo (e.g., in tumor cells or Ba/F3 cells expressing a wild type HER2, a mutant HER2, or a fragment of any thereof).
  • Potency of an HER2 inhibitor as provided herein can also be determined by IC50 value.
  • a compound with a lower IC50 value, as determined under substantially similar conditions, is a more potent inhibitor relative to a compound with a higher IC50 value.
  • the substantially similar conditions comprise determining an HER2- dependent phosphorylation level, in vitro or in vivo (e.g., in tumor cells or Ba/F3 cells expressing a wild type HER2, a mutant HER2, or a fragment of any thereof).
  • Assays can include, for example, proliferation inhibition assays such as those that measure cell growth inhibition, such as an MTS assay or by Cell Titer Gio Luminescent Cell viability assay (Promega®).
  • cells are seeded and grown in cell culture plates before being exposed to a test compound for varying durations. Assessment of the viability of the cells following this exposure is then performed. Data are normalized with respect to untreated cells and can be displayed graphically. Growth curves can be fitted using a nonlinear regression model with sigmoidal dose response. As another example, a Western Blot analysis can be used. In such assays cells are seeded and grown in culture plates and then treated with a test compound the following day for varying durations. Cells are washed with PBS and lysed.
  • SDS-PAGE gels are used to separate the lysates which are transferred to nitrocellulose membranes, and probed with appropriate antibodies (e.g., phospho-HER2(Tyrl248)(2247), phospho-EGFR-Tyrl 173 phospho- HER2-Tyr877, phospho-HER2-Tyrl221, total HER2, phospho-AKT-Thr308, phospho- AKT-Ser374, total AKT, phospho-p44/42 MAPK-Thr202/Tyr204, and p44/42 MAPK).
  • appropriate antibodies e.g., phospho-HER2(Tyrl248)(2247), phospho-EGFR-Tyrl 173 phospho- HER2-Tyr877, phospho-HER2-Tyrl221, total HER2, phospho-AKT-Thr308, phospho- AKT-Ser374, total AKT, phospho-p44/42 MAPK-Thr202/Tyr204, and p44/42 MA
  • the selectivity between wild type HER2 and HER2 containing one or more mutations as described herein can also be measured using cellular proliferation assays where cell proliferation is dependent on kinase activity.
  • murine Ba/F3 cells transfected with a suitable version of wild type HER2, or Ba/F3 cells transfected with HER2 having one or more mutations such as S310F, S310Y, R678Q, R678W, R678P, I767M, V773M, V777L, V842I, M774AYVM, M774del insWLV, A775_G776insYVMA, A775_G776insAVMA, A775_G776insSVMA, A775_G776insVAG, A775insV G776C, A775_G776insI, G776del insVC2, G776del insW, G776del insLC, G776C V777
  • Proliferation assays are performed at a range of inhibitor concentrations (e.g., 10 pM, 3 pM, 1.1 pM, 330 nM, 110 nM, 33 nM, 11 nM, 3 nM, 1 nM) and an ECso is calculated.
  • inhibitor concentrations e.g. 10 pM, 3 pM, 1.1 pM, 330 nM, 110 nM, 33 nM, 11 nM, 3 nM, 1 nM
  • the compounds provided herein can exhibit potent and selective inhibition of HER2.
  • the compounds provided herein can bind to the HER2 adenosine triphosphate (ATP)-binding site in the tyrosine kinase domain.
  • the compounds provided herein can exhibit nanomolar potency against a HER2 kinase including an activating mutation or a HER2 inhibitor resistance mutation, including, for example, exon 20 insertions and/or the resistance mutations in Table 5 (e.g., L755S, L755P, T798I, and T798M), with minimal activity against related kinases (e.g., wild type EGFR).
  • the compounds of Formula (I) can selectively target a HER2 kinase.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can selectively target a HER2 kinase over another kinase (e.g., wild type EGFR) or non-kinase target.
  • a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I- c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, can exhibit greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein (e.g., one or more mutations as described in Table 3) relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I- c), (I-d) or (I-e)> or a pharmaceutically acceptable salt thereof, can exhibit greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein (e.g., one or more mutations as described in Table 3) relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit at least 2-fold, 3- fold, 5-fold, 10-fold, 25-fold, 50-fold or 100-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit up to 1000-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or nonkinase target.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit up to lOOOO-fold greater inhibition of wild type HER2 or HER2 having a combination of mutations described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I- c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, can exhibit from about 2- fold to about 10-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • non-kinase target e.g., wild type EGFR
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit from about 10-fold to about 100-fold greater inhibition of wild type HER2 or containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit from about 100-fold to about 1000-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit from about 1000-fold to about 1 OOOO-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein (e.g., one or more mutations as described in Table 3) relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a pharmaceutically acceptable salt thereof in combination with a second EGFR inhibitor can exhibit greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein (e.g., one or more mutations as described in Table 3) relative to inhibition of another kinase (e.g.,
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second HER2 inhibitor can exhibit at least 2-fold, 3 -fold, 5- fold, 10-fold, 25 -fold, 50-fold or 100-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • non-kinase target e.g., wild type EGFR
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second HER2 inhibitor can exhibit up to 1 OOO-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second HER2 inhibitor can exhibit up to 1 OOOO-fold greater inhibition of wild type HER2 or HER2 having a combination of mutations described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a pharmaceutically acceptable salt thereof in combination with a second HER2 inhibitor can exhibit from about 2-fold to about 10-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second HER2 inhibitor can exhibit from about 10-fold to about 1 OO- fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non- kinase target.
  • another kinase e.g., wild type EGFR
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second HER2 inhibitor can exhibit from about 100-fold to about 1000-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second HER2 inhibitor can exhibit from about 1 OOO-fold to about 1 OOOO-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • Compounds of Formula (I) are useful for treating diseases and disorders which can be treated with a HER2 inhibitor, such as HER2-associated diseases and disorders, e.g., proliferative disorders such as cancers (e.g., a HER2-associated cancer), including hematological cancers and solid tumors (e.g., advanced solid tumors).
  • HER2-associated diseases and disorders e.g., proliferative disorders such as cancers (e.g., a HER2-associated cancer), including hematological cancers and solid tumors (e.g., advanced solid tumors).
  • the compounds provided herein can also inhibit EGFR and HER2 as described herein.
  • the compounds provided herein can exhibit potent and selective inhibition of EGFR and HER2.
  • the compounds provided herein can exhibit nanomolar potency against an EGFR kinase having one or more mutations, including, for example, one or more of the mutations in Tables la, lb and 2a, 2b, and a HER2 kinase having one or more mutations, including, for example, the mutations in Table 3, with minimal activity against related kinases (e.g., wild type EGFR).
  • the compounds of Formula (I) can selectively target an EGFR and a HER2 kinase.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can selectively target an EGFR kinase and a HER2 kinase over another kinase or non-kinase target.
  • a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I- c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, can exhibit greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein (e.g., one or more mutations as described in Tables 3-5) relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I- c), (I-d) or (I-e)
  • a pharmaceutically acceptable salt thereof can exhibit greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein (e.g., one or more mutations as described in Tables 3-5)
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit at least 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or 100-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non- kinase target.
  • another kinase e.g., wild type EGFR
  • non- kinase target e.g., wild type EGFR
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit up to 1000-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit up to 1 OOOO-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 having one or more mutations described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I- c), (I d) or (I-e))> or a pharmaceutically acceptable salt thereof, can exhibit from about 2- fold to about 10-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non- kinase target.
  • another kinase e.g., wild type EGFR
  • non- kinase target e.g., wild type EGFR
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit from about 10-fold to about 100-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit from about 1 OO-fold to about 1 OOO-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • non-kinase target e.g., wild type EGFR
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit from about 1 OOO-fold to about 1 OOOO- fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • non-kinase target e.g., wild type EGFR
  • a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR and/or second HER2 inhibitor can exhibit greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein (e.g., one or more mutations as described in Table 3) relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a pharmaceutically acceptable salt thereof in combination with a second EGFR and/or second HER2 inhibitor can exhibit greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR and/or second HER2 inhibitor can exhibit at least 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or 100-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR and/or second HER2 inhibitor can exhibit up to 1000-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • non-kinase target e.g., wild type EGFR
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR and/or HER2 inhibitor can exhibit up to lOOOO-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 having a combination of mutations described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • non-kinase target e.g., wild type EGFR
  • a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR and/or second HER2 inhibitor can exhibit from about 2-fold to about 10-fold greater inhibition of EGFR containing one or more mutations as described herein and HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • non-kinase target e.g., wild type EGFR
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR and/or second HER2 inhibitor can exhibit from about 10-fold to about 100-fold greater inhibition of EGFR containing one or more mutations as described herein and HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • non-kinase target e.g., wild type EGFR
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR and/or second HER2 inhibitor can exhibit from about 100-fold to about 1000-fold greater inhibition of EGFR containing one or more mutations as described herein and second HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • non-kinase target e.g., wild type EGFR
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR and/or second HER2 inhibitor can exhibit from about 1000-fold to about 10000-fold greater inhibition of EGFR containing one or more mutations as described herein and HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
  • another kinase e.g., wild type EGFR
  • non-kinase target e.g., wild type EGFR
  • BUB buffered uninhibited by benzimidazole, BUB1-3
  • BUB1-3 benzimidazole, BUB1-3
  • inhibitors of BUB1 kinase useful for treating or preventing diseases or disorders associated with enhanced uncontrolled proliferative cellular processes such as, for example, cancer, inflammation, arthritis, viral diseases, cardiovascular diseases, or fungal diseases.
  • the disease or disorder is cancer.
  • a “BUB1 inhibitor” as used herein includes any compound exhibiting BUB1 inactivation activity (e.g., inhibiting or decreasing).
  • a BUB1 inhibitor can be selective for BUB1 over other kinases (e.g., wildtype EGFR).
  • the compounds provided herein can inhibit a Bub kinase. In some embodiments, the compounds provided herein can inhibit BUB1 kinase.
  • test compounds to act as inhibitors of BUB 1 may be demonstrated by assays known in the art.
  • the activity of the compounds and compositions provided herein as BUB1 inhibitors can be assayed in vitro, in vivo, or in a cell line.
  • In vitro assays include assays that determine inhibition of the kinase.
  • BUB1 inhibition of a compound provided herein can be determined using a time-resolved fluorescence energy transfer (TR-FRET) assay which measures phosphorylation of a synthetic peptide (e.g., Biotin-AHX-VLLPKKSFAEPG (C-terminus in amide form) by the (recombinant) catalytic domain of human BUB1 (amino acids 704-1085), expressed in Hi5 insect cells with an N-terminal His6-tag and purified by affinity- (Ni-NTA) and size exclusion chromatography.
  • TR-FRET time-resolved fluorescence energy transfer
  • the compounds provided herein exhibit central nervous system (CNS) penetrance.
  • CNS central nervous system
  • such compounds can be capable of crossing the blood brain barrier (BBB) and inhibiting an EGFR and/or HER2 kinase in the brain and/or other CNS structures.
  • the compounds provided herein are capable of crossing the blood brain barrier in a therapeutically effective amount.
  • treatment of a patient with cancer e.g., an EGFR-associated cancer or a HER2-associated cancer such as an EGFR- or HER2-associated brain or CNS cancer or an EGFR-associated or a HER2-associated cancer that has metastasized to the brain or CNS
  • administration e.g., oral administration
  • BBB models such as the transwell system, the hollow fiber (dynamic in vitro BBB) model, other microfluidic BBB systems, the BBB spheroid platform, and other cell aggregate-based BBB models. See, e.g., Cho et al. Nat Commun. 2017; 8: 15623; Bagchi et al. Drug Des Devel Ther. 2019; 13: 3591-3605; Gastfriend et al. Curr Opin Biomed Eng. 2018 Mar; 5: 6-12; and Wang et al. Biotechnol Bioeng. 2017 Jan; 114(1): 184-194.
  • the compounds described herein are fluorescently labeled, and the fluorescent label can be detected using microscopy (e.g., confocal microscopy).
  • microscopy e.g., confocal microscopy
  • the ability of the compound to penetrate the surface barrier of the model can be represented by the fluorescence intensity at a given depth below the surface.
  • the fluorescent label is non-fluorescent until it permeates live cells and is hydrolyzed by intracellular esterases to produce a fluorescent compound that is retained in the cell and can be quantified with a spectrophotometer.
  • Non-limiting examples of fluorescent labels that can be used in the assays described herein include Cy5, rhodamine, infrared IRDye® CW-800 (LICOR #929-71012), far-red IRDye® 650 (LICOR #929- 70020), sodium fluorescein (Na-F), lucifer yellow (LY), 5 ’carboxyfluorescein, and calcein-acetoxymethylester (calcein-AM).
  • the BBB model e.g., the tissue or cell aggregate
  • a compound described herein can be detected in one or more sections using mass spectrometry (e.g., MALDI-MSI analyses).
  • the ability of a compound described herein to cross the BBB through a transcellular transport system can be demonstrated by assays known in the art. See, e.g., Wang et al. Drug Deliv. 2019; 26(1): 551-565.
  • assays to determine if compounds can be effluxed by the P-glycoprotein (Pgp) include monolayer efflux assays in which movement of compounds through Pgp is quantified by measuring movement of digoxin, a model Pgp substrate (see, e.g., Doan et al. 2002. J Pharmacol Exp Ther.
  • binding of the compounds described herein to brain tissue is quantified.
  • a brain tissue binding assay can be performed using equilibrium dialysis, and the fraction of a compound described herein unbound to brain tissue can be detected using LC-MS/MS (Cyprotex: Brain Tissue Binding Assay www.cyprotex.com/admepk/protein_binding/brain-tissue-binding/).
  • Compounds of Formula (I) are useful for treating diseases and disorders which can be treated with an EGFR inhibitor, a HER2 inhibitor, a dual EGFR and HER2 inhibitor, and/or a BUB1 inhibitor, such as those described herein, e.g., cancer.
  • an EGFR inhibitor e.g., a HER2 inhibitor, a dual EGFR and HER2 inhibitor, and/or a BUB1 inhibitor, such as those described herein, e.g., cancer.
  • a method for treating a disease or disorder as provided herein in a subject in need thereof the method comprising administering to the subject a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof.
  • the disease or disorder is cancer.
  • treat or “treatment” refer to therapeutic or palliative measures.
  • Beneficial or desired clinical results include, but are not limited to, alleviation, in whole or in part, of symptoms associated with a disease or disorder or condition, diminishment of the extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state (e.g., one or more symptoms of the disease), and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • the terms “subject,” “individual,” or “patient,” are used interchangeably, refers to any animal, including mammals such as mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, primates, and humans.
  • the subject is a human.
  • the subject has experienced and/or exhibited at least one symptom of the disease or disorder to be treated and/or prevented.
  • the subject has been identified or diagnosed as having a cancer with a dysregulation of an EGFR gene, an EGFR protein, or expression or activity, or level of any of the same (an EGFR-associated cancer) (e.g., as determined using a regulatory agency-approved, e.g., FDA-approved, assay or kit).
  • the subject has a tumor that is positive for a dysregulation of an EGFR gene, an EGFR protein, or expression or activity, or level of any of the same (e.g., as determined using a regulatory agency-approved assay or kit).
  • the subject has a tumor that is positive for a mutation as described in Table la and lb.
  • the subject can be a subject with a tumor(s) that is positive for a dysregulation of an EGFR gene, an EGFR protein, or expression or activity, or level of any of the same (e.g., identified as positive using a regulatory agency-approved, e.g., FDA-approved, assay or kit).
  • the subject can be a subject whose tumors have a dysregulation of an EGFR gene, an EGFR protein, or expression or activity, or a level of the same (e.g., where the tumor is identified as such using a regulatory agency-approved, e.g., FDA-approved, kit or assay).
  • the subject is suspected of having an EGFR-associated cancer.
  • the subject has a clinical record indicating that the subject has a tumor that has a dysregulation of an EGFR gene, an EGFR protein, or expression or activity, or level of any of the same (and optionally the clinical record indicates that the subject should be treated with any of the compositions provided herein).
  • the subject has been identified or diagnosed as having a cancer with a dysregulation of a HER2 gene, a HER2 protein, or expression or activity, or level of any of the same (a HER2-associated cancer) (e.g., as determined using a regulatory agency-approved, e.g., FDA-approved, assay or kit).
  • the subject has a tumor that is positive for a dysregulation of a HER2 gene, a HER2 protein, or expression or activity, or level of any of the same (e.g., as determined using a regulatory agency- approved assay or kit).
  • the subject has a tumor that is positive for a mutation as described in Table 3.
  • the subject can be a subject with a tumor(s) that is positive for a dysregulation of a HER2 gene, a HER2 protein, or expression or activity, or level of any of the same (e.g., identified as positive using a regulatory agency-approved, e.g., FDA- approved, assay or kit).
  • the subject can be a subject whose tumors have a dysregulation of a HER2 gene, a HER2 protein, or expression or activity, or a level of the same (e.g., where the tumor is identified as such using a regulatory agency-approved, e.g., FDA-approved, kit or assay).
  • the subject is suspected of having a HER2-associated cancer.
  • the subject has a clinical record indicating that the subject has a tumor that has a dysregulation of a HER2 gene, a HER2 protein, or expression or activity, or level of any of the same (and optionally the clinical record indicates that the subject should be treated with any of the compositions provided herein).
  • the subject is a pediatric subject.
  • the term “pediatric subject” as used herein refers to a subject under the age of 21 years at the time of diagnosis or treatment.
  • the term “pediatric” can be further be divided into various subpopulations including: neonates (from birth through the first month of life); infants (1 month up to two years of age); children (two years of age up to 12 years of age); and adolescents (12 years of age through 21 years of age (up to, but not including, the twenty-second birthday)).
  • Berhman RE Kliegman R, Arvin AM, Nelson WE. Nelson Textbook of Pediatrics, 15th Ed. Philadelphia: W.B. Saunders Company, 1996; Rudolph AM, et al. Rudolph ’s Pediatrics, 21st Ed.
  • a pediatric subject is from birth through the first 28 days of life, from 29 days of age to less than two years of age, from two years of age to less than 12 years of age, or 12 years of age through 21 years of age (up to, but not including, the twenty-second birthday).
  • a pediatric subject is from birth through the first 28 days of life, from 29 days of age to less than 1 year of age, from one month of age to less than four months of age, from three months of age to less than seven months of age, from six months of age to less than 1 year of age, from 1 year of age to less than 2 years of age, from 2 years of age to less than 3 years of age, from 2 years of age to less than seven years of age, from 3 years of age to less than 5 years of age, from 5 years of age to less than 10 years of age, from 6 years of age to less than 13 years of age, from 10 years of age to less than 15 years of age, or from 15 years of age to less than 22 years of age.
  • compounds of Formula (I) e.g., Formula (I-a), (I-b), (I- c), (I d) or (I-e)
  • diseases and disorders as defined herein (for example, autoimmune diseases, inflammatory diseases, pulmonary disorders, cardiovascular disease, ischemia, liver disease, gastrointestinal disorders, viral or bacterial infections, central nervous system diseases (e.g., neurodegenerative diseases), and cancer).
  • preventing means to delay the onset, recurrence or spread, in whole or in part, of the disease or condition as described herein, or a symptom thereof.
  • EGFR-associated disease or disorder refers to diseases or disorders associated with or having a dysregulation of an EGFR gene, an EGFR kinase (also called herein an EGFR kinase protein), or the expression or activity or level of any (e.g., one or more) of the same (e.g., any of the types of dysregulation of an EGFR gene, an EGFR kinase, an EGFR kinase domain, or the expression or activity or level of any of the same described herein).
  • Non-limiting examples of an EGFR-associated disease or disorder include, for example, cancer, a central nervous system disease, a pulmonary disorder, cardiovascular disease, ischemia, liver disease, a gastrointestinal disorder, a viral or bacterial infection, and an inflammatory and/or autoimmune disease (e.g., psoriasis, eczema, atopic dermatitis, and atherosclerosis).
  • a central nervous system disease e.g., a central nervous system disease, a pulmonary disorder, cardiovascular disease, ischemia, liver disease, a gastrointestinal disorder, a viral or bacterial infection
  • an inflammatory and/or autoimmune disease e.g., psoriasis, eczema, atopic dermatitis, and atherosclerosis.
  • the inflammatory and/or autoimmune disease is selected from arthritis, systemic lupus erythematosus, atherosclerosis, and skin related disorders such as psoriasis, eczema, and atopic dermatitis.
  • arthritis systemic lupus erythematosus
  • atherosclerosis and skin related disorders such as psoriasis, eczema, and atopic dermatitis.
  • the central nervous system disease is a neurodegenerative disease.
  • the central nervous system disease is selected from Alzheimer's disease, Parkinson's disease, Huntington’s disease, amyotrophic lateral sclerosis, spinal cord injury, peripheral neuropathy, brain ischemia, and a psychiatric disorder such as schizophrenia. See, e.g., Iwakura and Nawa. Front Cell Neurosci. . 2013 Feb 13;7:4; and Chen et al. Sci Rep. 2019 Feb 21;9(1):2516.
  • EGFR-associated cancer refers to cancers associated with or having a dysregulation of an EGFR gene, an EGFR kinase (also called herein an EGFR kinase protein), or expression or activity, or level of any of the same.
  • an EGFR-associated cancer are described herein.
  • the phrase “dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same” refers to a genetic mutation (e.g., a mutation in an EGFR gene that results in the expression of an EGFR protein that includes a deletion of at least one amino acid as compared to a wild type EGFR protein, a mutation in an EGFR gene that results in the expression of an EGFR protein with one or more point mutations as compared to a wild type EGFR protein, a mutation in an EGFR gene that results in the expression of an EGFR protein with at least one inserted amino acid as compared to a wild type EGFR protein, a gene duplication that results in an increased level of EGFR protein in a cell, or a mutation in a regulatory sequence (e.g., a promoter and/or enhancer) that results in an increased level of EGFR protein in a cell), an alternative spliced version of an EGFR mRNA that
  • a dysregulation of an EGFR gene, an EGFR protein, or expression or activity, or level of any of the same can be a mutation in an EGFR gene that encodes an EGFR protein that is constitutively active or has increased activity as compared to a protein encoded by an EGFR gene that does not include the mutation.
  • Non-limiting examples of EGFR kinase protein point mutations/insertions/deletions are described in Table la and lb. Additional examples of EGFR kinase protein mutations (e.g., point mutations) are EGFR inhibitor resistance mutations (e.g., EGFR inhibitor mutations).
  • EGFR inhibitor resistance mutations are described in Table 2a and 2b.
  • the one or more EGFR inhibitor resistance mutations can include a substitution at amino acid position 718, 747, 761, 790, 797, or 854 (e.g., L718Q, L747S, D761Y, T790M, C797S, or T854A).
  • a substitution at amino acid position 718, 747, 761, 790, 797, or 854 e.g., L718Q, L747S, D761Y, T790M, C797S, or T854A.
  • dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same can be caused by an activating mutation in an EGFR gene.
  • dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same can be caused by a genetic mutation that results in the expression of an EGFR kinase that has increased resistance to an EGFR inhibitor, a tyrosine kinase inhibitor (TKI), and/or a multi-kinase inhibitor (MKI), e.g., as compared to a wild type EGFR kinase (see, e.g., the amino acid substitutions in Table 2a and 2b).
  • TKI tyrosine kinase inhibitor
  • MKI multi-kinase inhibitor
  • dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same can be caused by a mutation in a nucleic acid encoding an altered EGFR protein (e.g., an EGFR protein having a mutation (e.g., a primary mutation)) that results in the expression of an altered EGFR protein that has increased resistance to inhibition by an EGFR inhibitor, a tyrosine kinase inhibitor (TKI), and/or a multi-kinase inhibitor (MKI), e.g., as compared to a wild type EGFR kinase (see, e.g., the amino acid substitutions in Table 2a and 2b).
  • an altered EGFR protein e.g., an EGFR protein having a mutation (e.g., a primary mutation)
  • TKI tyrosine kinase inhibitor
  • MKI multi-kinase inhibitor
  • the exemplary EGFR kinase point mutations, insertions, and deletions shown in Tables la, lb and 2a, 2b can be caused by an activating mutation and/or can result in the expression of an EGFR kinase that has increased resistance to an EGFR inhibitor), tyrosine kinase inhibitor (TKI), and/or a multikinase inhibitor (MKI).
  • TKI tyrosine kinase inhibitor
  • MKI multikinase inhibitor
  • the individual has two or more EGFR inhibitor resistance mutations that increase resistance of the cancer to a first EGFR inhibitor.
  • the individual can have two EGFR inhibitor resistance mutations.
  • the two mutations occur in the same EGFR protein.
  • the two mutations occur in separate EGFR proteins.
  • the individual can have three EGFR inhibitor resistance mutations.
  • the three mutations occur in the same EGFR protein.
  • the three mutations occur in separate EGFR proteins.
  • the individual has two or more EGFR inhibitor resistance mutations selected from Del 19/L718Q, Del 19/T790M, Del 19/L844V, Del 19/T790M/L718Q, Del/T790M/C797S, Del 19/T790M/L844V, L858R/L718Q, L858R/L844V, L858R/T790M, L858R/T790M/L718Q, L858R/T790M/C797S, and
  • L858R/T790M/I941R or any combination thereof; e.g., any two of the aforementioned EGFR inhibitor resistance mutations.
  • activating mutation in reference to EGFR describes a mutation in an EGFR gene that results in the expression of an EGFR kinase that has an increased kinase activity, e.g., as compared to a wild type EGFR kinase, e.g., when assayed under identical conditions.
  • an activating mutation can be a mutation in an EGFR gene that results in the expression of an EGFR kinase that has one or more (e.g., two, three, four, five, six, seven, eight, nine, or ten) amino acid substitutions (e.g., any combination of any of the amino acid substitutions described herein) that has increased kinase activity, e.g., as compared to a wild type EGFR kinase, e.g., when assayed under identical conditions.
  • one or more e.g., two, three, four, five, six, seven, eight, nine, or ten amino acid substitutions (e.g., any combination of any of the amino acid substitutions described herein) that has increased kinase activity, e.g., as compared to a wild type EGFR kinase, e.g., when assayed under identical conditions.
  • an activating mutation can be a mutation in an EGFR gene that results in the expression of an EGFR kinase that has one or more (e.g., two, three, four, five, six, seven, eight, nine, or ten) amino acids deleted, e.g., as compared to a wild type EGFR kinase, e.g., when assayed under identical conditions.
  • an activating mutation can be a mutation in an EGFR gene that results in the expression of an EGFR kinase that has at least one (e.g., at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 12, at least 14, at least 16, at least 18, or at least 20) amino acid inserted as compared to a wild type EGFR kinase, e.g., the exemplary wild type EGFR kinase described herein, e.g., when assayed under identical conditions. Additional examples of activating mutations are known in the art.
  • wild type or wild-type describes a nucleic acid (e.g., an EGFR gene or an EGFR mRNA) or protein (e.g., an EGFR protein) sequence that is typically found in a subject that does not have a disease or disorder related to the reference nucleic acid or protein.
  • nucleic acid e.g., an EGFR gene or an EGFR mRNA
  • protein e.g., an EGFR protein
  • wild type EGFR or wild-type EGFR
  • an EGFR nucleic acid e.g., an EGFR gene or an EGFR mRNA
  • protein e.g., an EGFR protein
  • wild type EGFR or wild-type EGFR
  • an EGFR-associated disease e.g., an EGFR-associated cancer
  • protein e.g., an EGFR protein
  • an EGFR-associated disease e.g., an EGFR-associated cancer
  • a method of treating cancer e.g., an EGFR-associated cancer
  • the method comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I- b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof.
  • a compound of Formula (I) e.g., Formula (I-a), (I- b), (I-c), (I-d) or (I-e)
  • kits for treating an EGFR- associated cancer in a subject in need of such treatment comprising a) detecting a dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same in a sample from the subject; and b) administering a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof.
  • the dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same includes one or more EGFR kinase protein point mutations/insertions.
  • Non-limiting examples of EGFR kinase protein point mutations/insertions/deletions are described in Table la and lb.
  • the EGFR kinase protein point mutations/insertions/deletions are selected from the group consisting of G719S, G719C, G719A, L747S, D761Y, T790M, T854A, L858R, L861Q, a deletion in exon 19 (e.g., L747_A750del), and an insertion in exon 20 (e.g., V769_D770insX, D770_N771insX, N771_P772insX, P772_H773insX, or
  • the EGFR kinase protein point mutations/insertions/deletions are selected from the group consisting of L858R, deletions in exon 19 (e.g., L747_A750del), L747S, D761Y, T790M, and T854A.
  • the EGFR kinase protein insertion is an exon 20 insertion.
  • the EGFR kinase protein insertion is an exon 20 insertion selected from the group consisting of: V769_D770insX, D770_N771insX, N771_P772insX,
  • the EGFR kinase protein insertion is an exon 20 insertion selected from the group consisting of: A767_V769dupASV, V769_D770insASV, D770_N771insNPG, D770_N771insNPY, D770_N771insSVD, D770_N771insGL, N771_H773dupNPH, N771_P772insN, N771_P772insH,
  • the cancer e.g., EGFR-associated cancer
  • a hematological cancer e.g., acute lymphocytic cancer, Hodgkin lymphoma, non-Hodgkin lymphoma, and leukemia such as acute-myelogenous leukemia (AML), chronic-myelogenous leukemia (CML), acute- promyelocytic leukemia, and acute lymphocytic leukemia (ALL)
  • AML acute-myelogenous leukemia
  • CML chronic-myelogenous leukemia
  • ALL acute lymphocytic leukemia
  • central or peripheral nervous system tissue cancer an endocrine or neuroendocrine cancer including multiple neuroendocrine type I and type II tumors, Li-Fraumeni tumors, alveolar rhabdomyosarcoma, bone cancer, brain cancer, breast cancer, cancer of the anus, anal canal, or anorectum, cancer of the eye, cancer of the intrahepatic bile
  • the cancer is selected from the group consisting of: head and neck, ovarian, cervical, bladder and oesophageal cancers, pancreatic, gastrointestinal cancer, gastric, breast, endometrial and colorectal cancers, hepatocellular carcinoma, glioblastoma, bladder, lung cancer, e.g., non-small cell lung cancer (NSCLC), bronchioloalveolar carcinoma.
  • the cancer is pancreatic cancer, head and neck cancer, melanoma, colon cancer, renal cancer, leukemia, lung cancer, or breast cancer. In some cases, the cancer is melanoma, colon cancer, renal cancer, leukemia, or breast cancer.
  • the compounds provided herein are useful for treating a primary brain tumor or metastatic brain tumor.
  • the compounds can be used in the treatment of one or more of gliomas such as glioblastoma (also known as glioblastoma multiforme), astrocytomas, oligodendrogliomas, ependymomas, and mixed gliomas, meningiomas, medulloblastomas, gangliogliomas, schwannomas (neurilemmomas), and craniopharyngiomas (see, for example, Liu et al. J Exp Clin Cancer Res. 2019 May 23;38(1):219); and Ding et al. Cancer Res.
  • gliomas such as glioblastoma (also known as glioblastoma multiforme), astrocytomas, oligodendrogliomas, ependymomas, and mixed gliomas, meningiomas, medulloblastomas, gan
  • the brain tumor is a primary brain tumor.
  • the brain tumor is a metastatic brain tumor, e.g., a metastatic brain tumor from lung cancer, melanoma, breast cancer, ovarian cancer, colorectal cancer, kidney cancer, bladder cancer, or undifferentiated carcinoma.
  • the brain tumor is a metastatic brain tumor from lung cancer (e.g., non-small cell lung cancer).
  • the compounds provided herein exhibit brain and/or central nervous system (CNS) penetrance.
  • CNS central nervous system
  • the patient has previously been treated with another anticancer agent, e.g., another EGFR and/or HER2 inhibitor (e.g., a compound that is not a compound of Formula I) or a multi-kinase inhibitor.
  • another anticancer agent e.g., another EGFR and/or HER2 inhibitor (e.g., a compound that is not a compound of Formula I) or a multi-kinase inhibitor.
  • the cancer is a cancer of B cell origin. In some embodiments, the cancer is a lineage dependent cancer. In some embodiments, the cancer is a lineage dependent cancer where EGFR or the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, plays a role in the initiation and/or development of the cancer.
  • the cancer is an EGFR-associated cancer.
  • a method for treating a subject diagnosed with or identified as having an EGFR-associated cancer comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof as defined herein.
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same includes one or more deletions (e.g., deletion of an amino acid at position 4), insertions, or point mutation(s) in an EGFR kinase.
  • dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same includes at least one deletion, insertion, or point mutation in an EGFR gene that results in the production of an EGFR kinase that has one or more of the amino acid substitutions, insertions, or deletions in Table la and lb.
  • the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same includes a deletion of one or more residues from the EGFR kinase, resulting in constitutive activity of the EGFR kinase domain.
  • the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same includes at least one point mutation in an EGFR gene that results in the production of an EGFR kinase that has one or more amino acid substitutions, insertions, or deletions as compared to the wild type EGFR kinase (see, for example, the point mutations listed in Table la and lb).
  • dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same includes at least one point mutation in an EGFR gene that results in the production of an EGFR kinase that has one or more of the amino acid substitutions, insertions, or deletions in Table la and lb.
  • the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same includes an insertion of one or more residues in exon 20 of the EGFR gene (e.g., any of the exon 20 insertions described in Table la and lb).
  • Exon 20 of EGFR has two major regions, the c -helix (residues 762- 766) and the loop following the c-helix (residues 767-774).
  • a stabilized and ridged active conformation induces resistance to first generation EGFR inhibitors.
  • the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same includes an insertion of one or more residues in exon 20 selected from the group consisting of: V769_D770insX, D770_N771insX, N771_P772insX, P772_H773insX, and H773_V774insX.
  • the EGFR kinase protein insertion is an exon 20 insertion selected from the group consisting of: A767_V769dupASV, V769_D770insASV, D770_N771insNPG, D770_N771insNPY, D770_N771insSVD, D770_N771insGL, N771_H773dupNPH, N771_P772insN, N771_P772insH, N771_P772insV, P772_H773insDNP, P772_H773insPNP,
  • the EGFR mutations shown may be activating mutations and/or confer increased resistance of EGFR to an EGFR inhibitor and/or a multi-kinase inhibitor (MKI), e.g., as compared to a wild type EGFR
  • MKI multi-kinase inhibitor
  • the EGFR mutations shown may be activating mutations and/or confer increased resistance of EGFR to an EGFR inhibitor and/or a multi-kinase inhibitor (MKI), e.g., as compared to a wild type EGFR
  • MKI multi-kinase inhibitor
  • the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same includes a splice variation in an EGFR mRNA which results in an expressed protein that is an alternatively spliced variant of EGFR having at least one residue deleted (as compared to the wild type EGFR kinase) resulting in a constitutive activity of an EGFR kinase domain.
  • the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same includes at least one point mutation in an EGFR gene that results in the production of an EGFR kinase that has one or more amino acid substitutions or insertions or deletions in an EGFR gene that results in the production of an EGFR kinase that has one or more amino acids inserted or removed, as compared to the wild type EGFR kinase.
  • the resulting EGFR kinase is more resistant to inhibition (e.g., inhibition of its signaling activity) by one or more first EGFR inhibitors, as compared to a wild type EGFR kinase or an EGFR kinase not including the same mutation.
  • Such mutations optionally, do not decrease the sensitivity of the cancer cell or tumor having the EGFR kinase to treatment with a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof (e.g., as compared to a cancer cell or a tumor that does not include the particular EGFR inhibitor resistance mutation).
  • a compound of Formula (I) e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)
  • a pharmaceutically acceptable salt thereof e.g., as compared to a cancer cell or
  • the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same includes at least one point mutation in an EGFR gene that results in the production of an EGFR kinase that has one or more amino acid substitutions as compared to the wild type EGFR kinase, and which has increased resistance to a compound of Formula (I), or a pharmaceutically acceptable salt thereof, as compared to a wild type EGFR kinase or an EGFR kinase not including the same mutation.
  • an EGFR inhibitor resistance mutation can result in an EGFR kinase that has one or more of an increased Vmax, a decreased K m , and a decreased KD in the presence of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, as compared to a wild type EGFR kinase or an EGFR kinase not having the same mutation in the presence of the same compound of Formula (I), or a pharmaceutically acceptable salt thereof.
  • dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same includes at least one EGFR inhibitor resistance mutation in an EGFR gene that results in the production of an EGFR kinase that has one or more of the amino acid substitutions, insertions, or deletions as described in Table 2a and 2b.
  • compounds of Formula (I) are useful in treating subjects that develop cancers with EGFR inhibitor resistance mutations (e.g., that result in an increased resistance to a first EGFR inhibitor, e.g., a substitution at ammo acid position 718, 747, 761, 790, 797, or 854 (e.g., L718Q, L747S, D761 Y, T790M, C797S, T854A), and/or one or more EGFR inhibitor resistance mutations listed in Table 2a and 2b) by either dosing in combination or as a subsequent or additional (e.g., followup) therapy to existing drug treatments (e.g., other inhibitors of EGFR; e.g., first and/or second EGFR inhibitors).
  • Table 2a EGFR Protein Amin

Abstract

This disclosure provides a compound of formula (I) or a pharmaceutically acceptable salt that inhibits epidermal growth factor receptor (EGER, ERBB1) and/or Human epidermal growth factor receptor 2 (HER2, ERBB2). These compounds of formula (I) are therefore useful for the treatment of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human). This disclosure also provides compositions containing the same as well as methods of using and making the same.

Description

TETRAHYDROINDOLE DERIVATIVES AS EGFR AND/OR HER2 INHIBTORS USEFUL FOR THE TREATMENT OF CANCER
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of United States Provisional Application No. 63/319,183, filed on March 11, 2022, which is incorporated herein by reference in its entirety.
TECHNICAL FIELD
This disclosure provides chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit epidermal growth factor receptor (EGFR, ERBB1) and/or Human epidermal growth factor receptor 2 (HER2, ERBB2). These chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) EGFR and/or HER2 activation contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human). This disclosure also provides compositions containing the same as well as methods of using and making the same.
BACKGROUND
Epidermal growth factor receptor (EGFR, ERBB1) and Human epidermal growth factor receptor 2 (HER2, ERBB2) are members of a family of proteins which regulate cellular processes implicated in tumor growth, including proliferation and differentiation. Several investigators have demonstrated the role of EGFR and HER2 in development and cancer (Reviewed in Salomon, et al., Crit. Rev. Oncol. Hematol. (1995) 19:183-232, Klapper, etal., Adv. Cancer Res. (2000) 77, 25-79 and Hynes and Stern, Biochim. Biophys. Acta (1994) 1198:165-184). EGFR overexpression is present in at least 70% of human cancers, such as non-small cell lung carcinoma (NSCLC), breast cancer, glioma, and prostate cancer. HER2 overexpression occurs in approximately 30% of all breast cancer. It has also been implicated in other human cancers including colon, ovary, bladder, stomach, esophagus, lung, uterus and prostate. HER2 overexpression has also been correlated with poor prognosis in human cancer, including metastasis, and early relapse. EGFR and HER2 are, therefore, widely recognized as targets for the design and development of therapies that can specifically bind and inhibit tyrosine kinase activity and its signal transduction pathway in cancer cells, and thus can serve as diagnostic or therapeutic agents. For example, EGFR tyrosine kinase inhibitors (TKIs) are effective clinical therapies for EGFR mutant advanced non-small cell lung cancer (NSCLC) patients. However, the vast majority of patients develop disease progression following successful treatment with an EGFR TKI. Common mechanisms of resistance include acquired, secondary mutation T790M, C797S, and EGFR exon 20 insertion mutations. For example, NSCLC tumors can have EGFR exon 20 insertion mutations that are intrinsically resistant to current EGFR TKIs.
Overexpression of another protein, BUB1 (Budding uninhibited by benzimidazole, BUB1) kinase, is often associated with proliferating cells, including cancer cells, and tissues (Bolanos-Garcia VM and Blundell TL, Trends Biochem. Sci. 36, 141, 2010). This protein is an essential part of the complex network of proteins that form the mitotic checkpoint. The major function of an unsatisfied mitotic checkpoint is to keep the anaphase-promoting complex/cyclosome (APC/C) in an inactive state. As soon as the checkpoint gets satisfied the APC/C ubiquitin-ligase targets cyclin B and securin for proteolytic degradation leading to separation of the paired chromosomes and exit from mitosis.
Incomplete mitotic checkpoint function has been linked with aneuploidy and tumourigenesis (see Weaver BA and Cleveland DW, Cancer Res. 67, 10103, 2007; King RW, Biochim Biophys Acta 1786, 4, 2008). In contrast, complete inhibition of the mitotic checkpoint has been recognized to result in severe chromosome missegregation and induction of apoptosis in tumor cells (see Kops GJ et al., Nature Rev. Cancer 5, 773, 2005; Schmidt M and Medema RH, Cell Cycle 5, 159, 2006; Schmidt M and Bastians H, Drug Res. Updates 10, 162, 2007). Thus, mitotic checkpoint inhibition through inhibition of BUB1 kinase represents an approach for the treatment of proliferative disorders, including solid tumors such as carcinomas, sarcomas, leukemias and lymphoid malignancies or other disorders, associated with uncontrolled cellular proliferation. SUMMARY
This disclosure provides chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit epidermal growth factor receptor (EGFR, ERBB1) and/or Human epidermal growth factor receptor 2 (HER2, ERBB2). These chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) EGFR and/or HER2 activation contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human). This disclosure also provides compositions containing the same as well as methods of using and making the same.
In one aspect, this disclosure features compounds of of Formula (I):
Figure imgf000004_0001
Formula (I) or a pharmaceutically acceptable salt thereof, wherein:
Ring C is selected from the group consisting of:
Figure imgf000004_0002
• bicyclic heteroaryl including 7-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with X1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc;
• bicyclic C5-10 cycloalkyl or C5-10 cycloalkenyl, each of which is optionally substituted with X1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc; • heterocyclyl or heterocycloalkenyl including from 5-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with X1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc; and
• C 10 or C 14 aryl optionally substituted with X1 and further optionally substituted with from 1-4 Rc;
X1 is selected from the group consisting of (a) -(X2)m-L1-R5; and (b)
Figure imgf000005_0001
; wherein: m is 0 or 1 ;
X2 is selected from the group consisting of:
Figure imgf000005_0002
• -C2-6 alkenyl ene optionally substituted with from 1-3 Ra;
• -C(=O)O-*, -C(=O)N(RN)-*, or -S(O)I-2N(RN)-*;
• -OC(=O)-*, -N(RN)C(=O)-*, or -N(RN)S(O)I-2-*; and
• -OC(=O)N(RN)-*, -N(RN)C(=O)O-*, -N(RN)C(=O)N(RN)-*, or - N(RN)S(O)I-2N(RN)-*, wherein the asterisk represents point of attachment to L1;
L1 and L2 is selected from the group consisting of: a bond and C1-10 alkylene optionally substituted with from 1-6 Ra;
R5 is selected from the group consisting of:
• H;
• halo;
• -OH;
• -NReRf; • -C1-6 alkoxy or -S(0)o-2(C1-6 alkyl), each optionally substituted with from 1-6 Ra;
• -Rg;
• -L5-Rg;
• -Rg2-Rw or -Rg2-RY; and
• -L5-Rg2-Rw or -L5-Rg2-RY;
R6 is selected from the group consisting of:
• H;
• halo;
• -OH;
• -NReRf;
• -Rg;
• -R"
• -L6-Rg;
• -Rg2-Rw or -Rg2-RY;
• -L6-Rg2-Rw or -L6-Rg2-RY; and
• -C1-6 alkoxy or -S(0)o-2(C1-6 alkyl), each optionally substituted with from 1-6 Ra; provided that: when L1 is a bond, then R5 is selected from the group consisting of: H, -Rg, -Rg2-
Rw, and -Rg2-RY; and
X1 is other than H, -OH, or NH2;
L5 is selected from the group consisting of: -O-, -S(0)o-2, -NH-, and -N(Rd)-;
Rw is -Lw-W, wherein Lw is C(=O), S(O)i-2, OC(=O)*, NHC(=O)*, NRdC(=O)*, NHS(O)I-2*, or NRdS(0)i-2*, wherein the asterisk represents point of attachment to W, and W is selected from the group consisting of:
• C2-6 alkenyl; C2-6 alkynyl; or C3-10 allenyl, each of which is optionally substituted with from 1-3 Ra and further optionally substituted with Rg, wherein W is attached to Lw via an sp2 or sp hybridized carbon atom, thereby providing an a, P- unsaturated system; and
• bicyclo [x.y.O] cycloalkyl optionally substituted with from 1 -2 Rc, wherein x is 1 or 2; and y is an integer from 1 to 6;
RY is selected from the group consisting of: -Rg and -(Lg)g-Rg; each of Rla, Rlb, R2a, R2b, R3a, and R3b is independently selected from the group consisting of:
• H;
• halo;
• -OH;
• -C(O)OH or -C(O)NH2;
• -CN;
• -Rb;
• -Lb-Rb;
• -C1-6 alkoxy or -C1-6 thioalkoxy, each optionally substituted with from 1-6 Ra;
• NReRf;
• Rg; and
. -(Lg)g-Rg;
. -(Lg)g-Rw;
• -(Lg)g-Rg2-Rw; or any two of variables Rla, Rlb, R2a, R2b, R3a, and R3b, together with the Ring B ring atoms to which each is attached, form a fused saturated or unsaturated ring of 3-12 ring atoms; • wherein from 0-2 of the ring atoms are each an independently selected heteroatom , wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated or unsaturated ring of 3-12 ring atoms is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rw;
Ring A is Rg;
R4 is selected from the group consisting of: H and Rd; each R7 is an independently selected Rc; n is 0, 1, 2, or 3; each occurrence of Ra is independently selected from the group consisting of: - OH; -halo; -NReRf; C1-4 alkoxy; C1-4 haloalkoxy; -C(=O)O(C1-4 alkyl); -C(=O)(C1-4 alkyl); -C(=O)OH; -CONR’R”; -S(O)I-2NR’R”; -S(O)I-2(CM alkyl); and cyano; each occurrence of Rb is independently C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each of which is optionally substituted with from 1-6 Ra; each occurrence of Lb is independently C(=O); S(O)i-2; C(=O)O*; C(=O)NH*; C(=O)NRd*; S(O)I-2NH*; or S(O)i-2N(Rd)*, wherein the asterisk represents point of attachment to Rb; each occurrence of Rc is independently selected from the group consisting of: halo; cyano; Ci-io alkyl which is optionally substituted with from 1-6 independently selected Ra; C2-6 alkenyl; C2-6 alkynyl; C1-4 alkoxy optionally substituted withC1-4 alkoxy or CM haloalkoxyC;1-4 haloalkoxy; -S(O)I-2(CM alkyl); -S(0)(=NH)(CM alkyl); -NReRf; -OH; -S(O)I-2NR’R”; -CM thioalkoxy; -NO2; -C(=0)(Ci-io alkyl); -C(=0)0(CM alkyl); - C(=O)OH; -C(=O)NR’R”; and -SF5; each occurrence of Rd is independently selected from the group consisting of: CM alkyl optionally substituted with from 1-3 independently selected Ra; -C(0)(CM alkyl); - C(0)0(C1-4 alkyl); -CONR’R”; -S(O)I-2NR’R”; - S(O)I-2(CM alkyl); -OH; and CM alkoxy; each occurrence of Re and Rf is independently selected from the group consisting of: H; C1-6 alkyl optionally substituted with from 1-3 substituents each independently selected from the group consisting of NR’R”, -OH, C1-6 alkoxy, C1-6 haloalkoxy, and halo; -C(O)(C1-4 alkyl); -C(O)O(CM alkyl); -CONR’R”; -S(O)I-2NR’R”; - S(O)I-2(CM alkyl); -OH; andC1-4 alkoxy; each occurrence of Rg is independently selected from the group consisting of:
• C3-8 cycloalkyl or C3-8 cycloalkenyl, each of which is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc;
• heterocyclyl or heterocycloalkenyl including from 3-8 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc;
• heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc; and
• Ce-io aryl optionally substituted with from 1-4 Rc; each occurrence of Lg is independently selected from the group consisting of: -O-, -NH-, -NRd, -S(0)o-2, C(O), and C1-3 alkylene optionally substituted with from 1-3 Ra; each g is independently 1, 2, or 3; each Rg2 is a divalent Rg group; each occurrence of R’ and R” is independently selected from the group consisting of: H; -OH; andC1-4 alkyl;
Also provided herein is a pharmaceutical composition comprising a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
Provided herein is a method for treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
Also provided herein is a method for treating cancer in a subject in need thereof, the method comprising (a) determining that the cancer is associated with a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same; and (b) administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
Provided herein is a method of treating an EGFR-associated disease or disorder in a subject, the method comprising administering to a subject identified or diagnosed as having an EGFR-associated disease or disorder a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
This disclosure also provides a method of treating an EGFR-associated disease or disorder in a subject, the method comprising: determining that the cancer in the subject is an EGFR-associated disease or disorder; and administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein. Further provided herein is a method of treating an EGFR-associated cancer in a subject, the method comprising administering to a subject identified or diagnosed as having an EGFR-associated cancer a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
This disclosure also provides a method of treating an EGFR-associated cancer in a subject, the method comprising: determining that the cancer in the subject is an EGFR- associated cancer; and administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
Provided herein is a method of treating a subject, the method comprising administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein, to a subject having a clinical record that indicates that the subject has a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same.
Also provided herein is a method of treating a subject having a cancer, wherein the method comprises:
(a) administering one or more doses of a first EGFR inhibitor to the subject for a period of time;
(b) after (a), determining whether a cancer cell in a sample obtained from the subject has at least one EGFR inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor of step (a); and
(c) administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has been determined to have a cancer cell that has at least one EGFR inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor of step (a); or
(d) administering additional doses of the first EGFR inhibitor of step (a) to the subject if the subject has not been determined to have a cancer cell that has at least one EGFR inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor of step (a).
Further provided herein is a method of treating a subject having a cancer, wherein the method comprises: (a) determining whether a cancer cell in a sample obtained from a subject having a cancer and previously administered one or more doses of a first EGFR inhibitor has one or more EGFR inhibitor resistance mutations that confer increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor that was previously administered to the subject; and
(b) administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has been determined to have a cancer cell that has at least one EGFR inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor that was previously administered to the subject; or
(c) administering additional doses of the first EGFR inhibitor to the subject if the subject has not been determined to have a cancer cell that has at least one EGFR inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor previously administered to the subject.
Also provided herein is a method of treating a subject having a cancer, wherein the method comprises:
(a) determining that a cancer cell in a sample obtained from a subject having a cancer and previously administered one or more doses of a first EGFR inhibitor has one or more EGFR inhibitor resistance mutations that confer increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor that was previously administered to the subject; and
(b) administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, as a monotherapy or in conjunction with another anti cancer agent to the subject.
Further provided herein is a method of treating a subject having a cancer, wherein the method comprises:
(a) determining that a cancer cell in a sample obtained from a subject having a cancer and previously administered one or more doses of a first EGFR inhibitor does not have one or more EGFR inhibitor resistance mutations that confer increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor that was previously administered to the subject; and
(b) administering additional doses of the first EGFR inhibitor to the subject.
This disclosure also provides a method for inhibiting EGFR in a mammalian cell, the method comprising contacting the mammalian cell with an effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof.
Also provided herein is a method for treating cancer in a subject in need thereof, the method comprising (a) determining that the cancer is associated with a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same; and (b) administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
Further provided herein is a method of treating a HER2-associated cancer in a subject, the method comprising administering to a subject identified or diagnosed as having a HER2-associated cancer a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
This disclosure also provides a method of treating a HER2-associated cancer in a subject, the method comprising: determining that the cancer in the subject is a HER2- associated cancer; and administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
Provided herein is a method of treating a subject having a cancer, the method comprising administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein, to a subject having a clinical record that indicates that the subject has a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same.
Also provided herein is a method of treating a subject having a cancer, wherein the method comprises:
(a) administering one or more doses of a first HER2 inhibitor to the subject for a period of time;
(b) after (a), determining whether a cancer cell in a sample obtained from the subject has at least one HER2 inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first HER2 inhibitor of step (a); and
(c) administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has been determined to have a cancer cell that has at least one HER2 inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first HER2 inhibitor of step (a); or
(d) administering additional doses of the first HER2 inhibitor of step (a) to the subject if the subject has not been determined to have a cancer cell that has at least one HER2 inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first HER2 inhibitor of step (a).
Further provided herein is a method of treating a subject having a cancer, wherein the method comprises:
(a) determining whether a cancer cell in a sample obtained from a subject having a cancer and previously administered one or more doses of a first HER2 inhibitor has one or more HER2 inhibitor resistance mutations that confer increased resistance to a cancer cell or tumor to treatment with the first HER2 inhibitor that was previously administered to the subject; and
(b) administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has been determined to have a cancer cell that has at least one HER2 inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first HER2 inhibitor that was previously administered to the subject; or (c) administering additional doses of the first HER2 inhibitor to the subject if the subject has not been determined to have a cancer cell that has at least one HER2 inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first HER2 inhibitor previously administered to the subject.
Also provided herein is a method of treating a subject having a cancer, wherein the method comprises:
(a) determining that a cancer cell in a sample obtained from a subject having a cancer and previously administered one or more doses of a first HER2 inhibitor has one or more HER2 inhibitor resistance mutations that confer increased resistance to a cancer cell or tumor to treatment with the first HER2 inhibitor that was previously administered to the subject; and
(b) administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, as a monotherapy or in conjunction with another anti cancer agent to the subject.
Further provided herein is a method of treating a subject having a cancer, wherein the method comprises:
(a) determining that a cancer cell in a sample obtained from a subject having a cancer and previously administered one or more doses of a first HER2 inhibitor does not have one or more HER2 inhibitor resistance mutations that confer increased resistance to a cancer cell or tumor to treatment with the first HER2 inhibitor that was previously administered to the subject; and
(b) administering additional doses of the first HER2 inhibitor to the subject.
This disclosure also provides a method for inhibiting HER2 in a mammalian cell, the method comprising contacting the mammalian cell with an effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof.
Also provided herein is a method for treating cancer in a subject in need thereof, the method comprising (a) determining that the cancer is associated with a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same and that the cancer is associated with a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same; and (b) administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
Further provided herein is a method of treating an EGFR-associated and HER2- associated cancer in a subject, the method comprising administering to a subject identified or diagnosed as having an EGFR-associated and a HER2-associated cancer a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
This disclosure also provides a method of treating a an EGFR-associated and HER2-associated cancer in a subject, the method comprising: determining that the cancer in the subject is an EGFR-associated and a HER2-associated cancer; and administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein.
Provided herein is a method of treating a subject, the method comprising administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as provided herein, to a subject having a clinical record that indicates that the subject has a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same and a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same.
This disclosure also provides a method for inhibiting EGFR and HER2 in a mammalian cell, the method comprising contacting the mammalian cell with an effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof.
In addition to the above, provided herein is a method for inhibiting a BUB (budding uninhibited by benzimidazole, BUB1-3) kinase. In some embodiments, the methods provided herein include methods for inhibiting BUB11. For example, a method for inhibiting BUB1 in a mammalian cell, the method comprising contacting the mammalian cell with an effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I- C), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof.
Other embodiments include those described in the Detailed Description and/or in the claims.
Additional Definitions
To facilitate understanding of the disclosure set forth herein, a number of additional terms are defined below. Generally, the nomenclature used herein and the laboratory procedures in organic chemistry, medicinal chemistry, and pharmacology described herein are those well-known and commonly employed in the art. Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Each of the patents, applications, published applications, and other publications that are mentioned throughout the specification and the attached appendices are incorporated herein by reference in their entireties.
The term “acceptable” with respect to a formulation, composition or ingredient, as used herein, means having no persistent detrimental effect on the general health of the subject being treated.
“API” refers to an active pharmaceutical ingredient.
The terms “effective amount” or “therapeutically effective amount,” as used herein, refer to a sufficient amount of a chemical entity being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result includes reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an “effective amount” for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms. An appropriate “effective” amount in any individual case is determined using any suitable technique, such as a dose escalation study.
The term “excipient” or “pharmaceutically acceptable excipient” means a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, carrier, solvent, or encapsulating material. In one embodiment, each component is “pharmaceutically acceptable” in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio. See, e.g., Remington: The Science and Practice of Pharmacy, 21st ed.; Lippincott Williams & Wilkins: Philadelphia, PA, 2005; Handbook of Pharmaceutical Excipients, 6th ed., ' Rowe el al., Eds.; The Pharmaceutical Press and the American Pharmaceutical Association: 2009; Handbook of Pharmaceutical Additives, 3rd ed.,' Ash and Ash Eds.; Gower Publishing Company: 2007; Pharmaceutical Preformulation and Formulation, 2nd ed.,' Gibson Ed.; CRC Press LLC: Boca Raton, FL, 2009.
The term “pharmaceutically acceptable salt” refers to a formulation of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound. In certain instances, pharmaceutically acceptable salts are obtained by reacting a compound described herein, with acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like. In some instances, pharmaceutically acceptable salts are obtained by reacting a compound having acidic group described herein with a base to form a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, A-methyl-D-glucamine, tris(hydroxymethyl)methylamine, and salts with amino acids such as arginine, lysine, and the like, or by other methods previously determined. The pharmacologically acceptable salt s not specifically limited as far as it can be used in medicaments. Examples of a salt that the compounds described hereinform with a base include the following: salts thereof with inorganic bases such as sodium, potassium, magnesium, calcium, and aluminum; salts thereof with organic bases such as methylamine, ethylamine and ethanolamine; salts thereof with basic amino acids such as lysine and ornithine; and ammonium salt. The salts may be acid addition salts, which are specifically exemplified by acid addition salts with the following: mineral acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, nitric acid, and phosphoric acid: organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, methanesulfonic acid, and ethanesulfonic acid; acidic amino acids such as aspartic acid and glutamic acid.
The term “pharmaceutical composition” refers to a mixture of a compound described herein with other chemical components (referred to collectively herein as “excipients”), such as carriers, stabilizers, diluents, dispersing agents, suspending agents, and/or thickening agents. The pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but not limited to: rectal, oral, intravenous, aerosol, parenteral, ophthalmic, pulmonary, and topical administration.
The term “subject” refers to an animal, including, but not limited to, a primate (e.g., human), monkey, cow, pig, sheep, goat, horse, dog, cat, rabbit, rat, or mouse. The terms “subject” and “patient” are used interchangeably herein in reference, for example, to a mammalian subject, such as a human.
The term "halo" refers to fluoro (F), chloro (Cl), bromo (Br), or iodo (I).
The term “oxo” refers to a divalent doubly bonded oxygen atom (i.e., “=O”). As used herein, oxo groups are attached to carbon atoms to form carbonyls.
The term "alkyl" refers to a saturated acyclic hydrocarbon radical that may be a straight chain or branched chain, containing the indicated number of carbon atoms. For example, Ci-io indicates that the group may have from 1 to 10 (inclusive) carbon atoms in it. Alkyl groups can either be unsubstituted or substituted with one or more substituents. Non-limiting examples include methyl, ethyl, Ao-propyl, tert-butyl, /7-hexyl. The term “saturated” as used in this context means only single bonds present between constituent carbon atoms and other available valences occupied by hydrogen and/or other substituents as defined herein.
The term "haloalkyl" refers to an alkyl, in which one or more hydrogen atoms is/are replaced with an independently selected halo.
The term "alkoxy" refers to an -O-alkyl radical (e.g., -OCH3).
The term "alkylene" refers to a divalent alkyl (e.g., -CH2-). Similarly, terms such as “cycloalkylene” and “heterocyclylene” refer to divalent cycloalkyl and heterocyclyl respectively. For avoidance of doubt, in “cycloalkylene” and “heterocyclylene”, the two radicals can be on the same ring carbon atom (e.g., a geminal diradical such as
Figure imgf000020_0001
Figure imgf000020_0002
different ring atoms (e.g., ring carbon and/or nitrogen atoms (e.g., vicinal ring carbon and/or nitrogen atoms)) (e.g.,
Figure imgf000020_0003
Figure imgf000020_0004
The term "alkenyl" refers to an acyclic hydrocarbon chain that may be a straight chain or branched chain having one or more carbon-carbon double bonds. The alkenyl moiety contains the indicated number of carbon atoms. For example, C2-6 indicates that the group may have from 2 to 6 (inclusive) carbon atoms in it. Alkenyl groups can either be unsubstituted or substituted with one or more substituents.
The term "alkynyl" refers to an acyclic hydrocarbon chain that may be a straight chain or branched chain having one or more carbon-carbon triple bonds. The alkynyl moiety contains the indicated number of carbon atoms. For example, C2-6 indicates that the group may have from 2 to 6 (inclusive) carbon atoms in it. Alkynyl groups can either be unsubstituted or substituted with one or more substituents.
The term "aryl" refers to a 6-20 carbon mono-, bi-, tri- or polycyclic group wherein at least one ring in the system is aromatic (e.g., 6-carbon monocyclic, 10-carbon bicyclic, or 14-carbon tricyclic aromatic ring system); and wherein 0, 1, 2, 3, or 4 atoms of each ring may be substituted by a substituent. Examples of aryl groups include phenyl, naphthyl, tetrahydronaphthyl, and the like.
The term "cycloalkyl" as used herein refers to cyclic saturated hydrocarbon groups having, e.g., 3 to 20 ring carbons, preferably 3 to 16 ring carbons, and more preferably 3 to 12 ring carbons or 3-10 ring carbons or 3-6 ring carbons, wherein the cycloalkyl group may be optionally substituted. Examples of cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Cycloalkyl may include multiple fused and/or bridged rings. Non-limiting examples of fused/bridged cycloalkyl includes: bicyclo[1.1.0]butane, bicyclo[2.1.0]pentane, bicyclo [1.1.1] pentane, bicyclo[3.1.0]hexane, bicyclo[2.1.1]hexane, bicyclo [3.2.0] heptane, bicyclo[4.1.0]heptane, bicyclo[2.2.1]heptane, bicyclo[3.1.1]heptane, bicyclo[4.2.0]octane, bicyclo[3.2.1]octane, bicyclo[2.2.2]octane, and the like. Cycloalkyl also includes spirocyclic rings (e.g., spirocyclic bicycle wherein two rings are connected through just one atom). Non-limiting examples of spirocyclic cycloalkyls include spiro[2.2]pentane, spiro[2.5] octane, spiro[3.5]nonane, spiro[3.5]nonane, spiro[3.5]nonane, spiro [4.4] nonane, spiro[2.6]nonane, spiro [4.5] decane, spiro[3.6]decane, spiro[5.5]undecane, and the like. The term “saturated” as used in this context means only single bonds present between constituent carbon atoms.
The term "cycloalkenyl" as used herein means partially unsaturated cyclic hydrocarbon groups having 3 to 20 ring carbons, preferably 3 to 16 ring carbons, and more preferably 3 to 12 ring carbons or 3-10 ring carbons or 3-6 ring carbons, wherein the cycloalkenyl group may be optionally substituted. Examples of cycloalkenyl groups include, without limitation, cyclopentenyl, cyclohexenyl, cycloheptenyl, and cyclooctenyl. As partially unsaturated cyclic hydrocarbon groups, cycloalkenyl groups may have any degree of unsaturation provided that one or more double bonds is present in the ring, none of the rings in the ring system are aromatic, and the cycloalkenyl group is not fully saturated overall. Cycloalkenyl may include multiple fused and/or bridged and/or spirocyclic rings.
The term “heteroaryl”, as used herein, means a mono-, bi-, tri- or polycyclic group having 5 to 20 ring atoms, alternatively 5, 6, 9, 10, or 14 ring atoms; wherein at least one ring in the system contains one or more heteroatoms independently selected from the group consisting of N, O, and S and at least one ring in the system is aromatic (but does not have to be a ring which contains a heteroatom, e.g. tetrahydroisoquinolinyl, e.g., tetrahydroquinolinyl). Heteroaryl groups can either be unsubstituted or substituted with one or more substituents. Examples of heteroaryl include thienyl, pyridinyl, furyl, oxazolyl, oxadiazolyl, pyrrolyl, imidazolyl, triazolyl, thiodiazolyl, pyrazolyl, isoxazolyl, thiadiazolyl, pyranyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, thiazolyl benzothienyl, benzoxadiazolyl, benzofuranyl, benzimidazolyl, benzotriazolyl, cinnolinyl, indazolyl, indolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, purinyl, thienopyridinyl, pyrido[2,3- t7]pyrimidinyl, pyrrolo[2,3-/>]pyridinyl, quinazolinyl, quinolinyl, thieno[2,3-c]pyridinyl, pyrazolo[3,4-/>]pyridinyl, pyrazolo[3,4-c]pyridinyl, pyrazolo[4,3-c]pyridine, pyrazolo[4,3-/?]pyridinyl, tetrazolyl, chromane, 2,3-dihydrobenzo[/?][ l ,4]dioxine, benzo[</][l,3]dioxole, 2, 3 -dihydrobenzofuran, tetrahydroquinoline, 2,3- dihydrobenzo[/?][ l ,4]oxathiine, isoindoline, and others. In some embodiments, the heteroaryl is selected from thienyl, pyridinyl, furyl, pyrazolyl, imidazolyl, isoindolinyl, pyranyl, pyrazinyl, and pyrimidinyl. For purposes of clarification, heteroaryl also includes aromatic lactams, aromatic cyclic ureas, or vinylogous analogs thereof, in which each ring nitrogen adjacent to a carbonyl is tertiary (i.e., all three valences are occupied by non-
Figure imgf000022_0001
hydrogen substituents), such as one or more of pyridone (e.g., -J— , I
Figure imgf000022_0002
o
Figure imgf000022_0003
wherein each ring nitrogen adjacent to a carbonyl is tertiary (i.e., the oxo group (i.e., “=0”) herein is a constituent part of the heteroaryl ring).
The term "heterocyclyl" refers to a mono-, bi-, tri-, or polycyclic saturated ring system with 3-16 ring atoms (e.g., 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system) having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic or polycyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2 or 3 atoms of each ring may be substituted by a substituent. Examples of heterocyclyl groups include piperazinyl, pyrrolidinyl, dioxanyl, morpholinyl, tetrahydrofuranyl, and the like. Heterocyclyl may include multiple fused and bridged rings. Non-limiting examples of fused/bridged heteorocyclyl includes: 2-azabicyclo[ 1.1.0] butane, 2-azabicyclo[2.1.0]pentane, 2- azabicyclo[l. l. l]pentane, 3-azabicyclo[3.1.0]hexane, 5-azabicyclo[2.1.1]hexane, 3- azabicyclo[3.2.0]heptane, octahydrocyclopenta[c]pyrrole, 3-azabicyclo[4.1.0]heptane, 7- azabicyclo[2.2. l]heptane, 6-azabicyclo[3.1.1]heptane, 7-azabicyclo[4.2.0]octane, 2- azabicyclo[2.2.2]octane, 3-azabicyclo[3.2.1]octane, 2-oxabicyclo[ 1.1.0] butane, 2- oxabicyclo[2.1.0]pentane, 2-oxabicyclo[l. l. l]pentane, 3-oxabicyclo[3.1.0]hexane, 5- oxabicyclo[2.1.1]hexane, 3-oxabicyclo[3.2.0]heptane, 3 -oxabicyclo [4. 1.0]heptane, 7- oxabicyclo[2.2.1]heptane, 6-oxabicyclo[3.1.1]heptane, 7-oxabicyclo[4.2.0]octane, 2- oxabicyclo[2.2.2]octane, 3-oxabicyclo[3.2.1]octane, and the like. Heterocyclyl also includes spirocyclic rings (e.g., spirocyclic bicycle wherein two rings are connected through just one atom). Non-limiting examples of spirocyclic heterocyclyls include 2- azaspiro[2.2]pentane, 4-azaspiro[2.5 ] octane, 1 -azaspiro [3.5] nonane, 2- azaspiro [3.5] nonane, 7-azaspiro[3.5]nonane, 2-azaspiro[4.4] nonane, 6- azaspiro[2.6]nonane, 1 , 7-diazaspir o [4.5] decane, 7-azaspiro[4.5] decane 2,5- diazaspiro [ 3.6] decane, 3 -azaspiro [5.5] undecane, 2-oxaspiro[2.2]pentane, 4- oxaspiro[2.5] octane, 1 -oxaspiro [3.5] nonane, 2-oxaspiro[3.5]nonane, 7- oxaspiro [3.5] nonane, 2-oxaspiro[4.4] nonane, 6-oxaspiro[2.6] nonane, 1,7- dioxaspiro[4.5]decane, 2, 5 - di oxaspiro [3.6] decane, 1 - oxaspiro [5.5] undecane, 3- oxaspiro [5.5] undecane, 3 -oxa-9-azaspiro [5.5] undecane and the like. The term “saturated” as used in this context means only single bonds present between constituent ring atoms and other available valences occupied by hydrogen and/or other substituents as defined herein.
The term "heterocycloalkenyl" as used herein means partially unsaturated cyclic ring system with 3-16 ring atoms (e.g., 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system) having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic or polycyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2 or 3 atoms of each ring may be substituted by a substituent. Examples of heterocycloalkenyl groups include, without limitation, tetrahydropyridyl, dihydropyrazinyl, dihydropyridyl, dihydropyrrolyl, dihydrofuranyl, dihydrothiophenyl. As partially unsaturated cyclic groups, heterocycloalkenyl groups may have any degree of unsaturation provided that one or more double bonds is present in the ring, none of the rings in the ring system are aromatic, and the heterocycloalkenyl group is not fully saturated overall. Heterocycloalkenyl may include multiple fused and/or bridged and/or spirocyclic rings.
As used herein, examples of aromatic rings include: benzene, pyridine, pyrimidine, pyrazine, pyridazine, pyridone, pyrrole, pyrazole, oxazole, thioazole, isoxazole, isothiazole, and the like.
As used herein, when a ring is described as being “partially unsaturated”, it means said ring has one or more additional degrees of unsaturation (in addition to the degree of unsaturation attributed to the ring itself; e.g., one or more double or tirple bonds between constituent ring atoms), provided that the ring is not aromatic. Examples of such rings include: cyclopentene, cyclohexene, cycloheptene, dihydropyridine, tetrahydropyridine, dihydropyrrole, dihydrofuran, dihydrothiophene, and the like.
For the avoidance of doubt, and unless otherwise specified, for rings and cyclic groups (e.g., aryl, heteroaryl, heterocyclyl, heterocycloalkenyl, cycloalkenyl, cycloalkyl, and the like described herein) containing a sufficient number of ring atoms to form bicyclic or higher order ring systems (e.g., tricyclic, polycyclic ring systems), it is understood that such rings and cyclic groups encompass those having fused rings, including those in which the points of fusion are located (i) on adjacent ring atoms (e.g., [x.x.O] ring systems, in
Figure imgf000024_0003
of ring atoms (bridged ring systems having all bridge lengths > 0) (e.g.,
Figure imgf000024_0001
Figure imgf000024_0002
In addition, atoms making up the compounds of the present embodiments are intended to include all isotopic forms of such atoms. Isotopes, as used herein, include those atoms having the same atomic number but different mass numbers. By way of general example and without limitation, isotopes of hydrogen include tritium and deuterium, and isotopes of carbon include 13C and 14C.
In addition, the compounds generically or specifically disclosed herein are intended to include all tautomeric forms. Thus, by way of example, a compound containing the encompasses the tautomeric form containing the moiety:
Figure imgf000025_0001
. y, a pyridinyl or pyrimidinyl moiety that is described to be optionally substituted with hydroxyl encompasses pyridone or pyrimidone tautomeric forms.
The compounds provided herein may encompass various stereochemical forms. The compounds also encompass diastereomers as well as optical isomers, e.g., mixtures of enantiomers including racemic mixtures, as well as individual enantiomers and diastereomers, which arise as a consequence of structural asymmetry in certain compounds. Unless otherwise indicated, when a disclosed compound is named or depicted by a structure without specifying the stereochemistry and has one or more chiral centers, it is understood to represent all possible stereoisomers of the compound.
The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features and advantages of the invention will be apparent from the description and drawings, and from the claims.
DETAILED DESCRIPTION
This disclosure provides chemical entities (e.g., a compound or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination of the compound) that inhibit epidermal growth factor receptor (EGFR, ERBB1) and/or Human epidermal growth factor receptor 2 (HER2, ERBB2). These chemical entities are useful, e.g., for treating a condition, disease or disorder in which increased (e.g., excessive) EGFR and/or HER2 activation contributes to the pathology and/or symptoms and/or progression of the condition, disease or disorder (e.g., cancer) in a subject (e.g., a human). In some embodiments, the chemical entities provided herein can inhibit an EGFR kinase and/or a HER2 kinase that has an exon 20 mutation (e.g., any of the exon 20 mutations described herein). Exon 20 mutations can confer intrinsic resistance to EGFR and/or HER2 inhibitors, and there are currently only limited targeted therapies that have been approved for subjects with these mutations. This disclosure also provides compositions containing the chemical entities provided herein as well as methods of using and making the same.
Formulae (I) Compounds
In one aspect, this disclosure features compounds of of Formula (I):
Figure imgf000026_0001
Formula (I) or a pharmaceutically acceptable salt thereof, wherein:
Ring C is selected from the group consisting of:
Figure imgf000026_0002
• bicyclic heteroaryl including 7-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with X1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc;
• bicyclic C5-10 cycloalkyl or C5-10 cycloalkenyl, each of which is optionally substituted with X1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc; • heterocyclyl or heterocycloalkenyl including from 5-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with X1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc; and
• C io or C 14 aryl optionally substituted with X1 and further optionally substituted with from 1-4 Rc;
X1 is selected from the group consisting of (a) -(X2)m-L1-R5; and (b)
Figure imgf000027_0001
; wherein: m is 0 or 1 ;
X2 is selected from the group consisting of:
Figure imgf000027_0002
• -C2-6 alkenyl ene optionally substituted with from 1-3 Ra;
• -C(=O)O-*, -C(=O)N(RN)-*, or -S(O)I-2N(RN)-*;
• -OC(=O)-*, -N(RN)C(=O)-*, or -N(RN)S(O)I-2-*; and
• -OC(=O)N(RN)-*, -N(RN)C(=O)O-*, -N(RN)C(=O)N(RN)-*, or - N(RN)S(O)I-2N(RN)-*, wherein the asterisk represents point of attachment to L1;
L1 and L2 is selected from the group consisting of: a bond and C1-10 alkylene optionally substituted with from 1-6 Ra;
R5 is selected from the group consisting of:
• H;
• halo;
• -OH;
• -NReRf; • -C1-6 alkoxy or -S(0)o-2(C1-6 alkyl), each optionally substituted with from 1-6 Ra;
• -Rg;
• -L5-Rg;
• -Rg2-Rw or -Rg2-RY; and
• -L5-Rg2-Rw or -L5-Rg2-RY;
R6 is selected from the group consisting of:
• H;
• halo;
• -OH;
• -NReRf;
• -Rg;
• -R"
• -L6-Rg;
• -Rg2-Rw or -Rg2-RY;
• -L6-Rg2-Rw or -L6-Rg2-RY; and
• -C1-6 alkoxy or -S(0)o-2(C1-6 alkyl), each optionally substituted with from 1-6 Ra; provided that: when L1 is a bond, then R5 is selected from the group consisting of: H, -Rg, -Rg2-
Rw, and -Rg2-RY; and
X1 is other than H, -OH, or NH2;
L5 is selected from the group consisting of: -O-, -S(0)o-2, -NH-, and -N(Rd)-;
Rw is -Lw-W, wherein Lw is C(=O), S(O)i-2, OC(=O)*, NHC(=O)*, NRdC(=O)*, NHS(O)I-2*, or NRdS(0)i-2*, wherein the asterisk represents point of attachment to W, and W is selected from the group consisting of:
• C2-6 alkenyl; C2-6 alkynyl; or C3-10 allenyl, each of which is optionally substituted with from 1-3 Ra and further optionally substituted with Rg, wherein W is attached to Lw via an sp2 or sp hybridized carbon atom, thereby providing an a, P- unsaturated system; and
• bicyclo [x.y.O] cycloalkyl optionally substituted with from 1 -2 Rc, wherein x is 1 or 2; and y is an integer from 1 to 6;
RY is selected from the group consisting of: -Rg and -(Lg)g-Rg; each of Rla, Rlb, R2a, R2b, R3a, and R3b is independently selected from the group consisting of:
• H;
• halo;
• -OH;
• -C(O)OH or -C(O)NH2;
• -CN;
• -Rb;
• -Lb-Rb;
• -C1-6 alkoxy or -C1-6 thioalkoxy, each optionally substituted with from 1-6 Ra;
• NReRf;
• Rg; and
. -(Lg)g-Rg;
. -(Lg)g-Rw;
• -(Lg)g-Rg2-Rw; or any two of variables Rla, Rlb, R2a, R2b, R3a, and R3b, together with the Ring B ring atoms to which each is attached, form a fused saturated or unsaturated ring of 3-12 ring atoms; • wherein from 0-2 of the ring atoms are each an independently selected heteroatom , wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated or unsaturated ring of 3-12 ring atoms is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rw;
Ring A is Rg;
R4 is selected from the group consisting of: H and Rd; each R7 is an independently selected Rc; n is 0, 1, 2, or 3; each occurrence of Ra is independently selected from the group consisting of: - OH; -halo; -NReRf; C1-4 alkoxy; C1-4 haloalkoxy; -C(=O)O(C1-4 alkyl); -C(=O)(C1-4 alkyl); -C(=O)OH; -CONR’R”; -S(O)I-2NR’R”; -S(O)I-2(CM alkyl); and cyano; each occurrence of Rb is independently C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each of which is optionally substituted with from 1-6 Ra; each occurrence of Lb is independently C(=O); S(O)i-2; C(=O)O*; C(=O)NH*; C(=O)NRd*; S(O)I-2NH*; or S(O)i-2N(Rd)*, wherein the asterisk represents point of attachment to Rb; each occurrence of Rc is independently selected from the group consisting of: halo; cyano; Ci-io alkyl which is optionally substituted with from 1-6 independently selected Ra; C2-6 alkenyl; C2-6 alkynyl; C1-4 alkoxy optionally substituted withC1-4 alkoxy or CM haloalkoxyC;1-4 haloalkoxy; -S(O)I-2(CM alkyl); -S(0)(=NH)(CM alkyl); -NReRf; -OH; -S(O)I-2NR’R”; -CM thioalkoxy; -NO2; -C(=0)(Ci-io alkyl); -C(=0)0(CM alkyl); - C(=O)OH; -C(=O)NR’R”; and -SF5; each occurrence of Rd is independently selected from the group consisting of: CM alkyl optionally substituted with from 1-3 independently selected Ra; -C(0)(CM alkyl); - C(0)0(C1-4 alkyl); -CONR’R”; -S(O)I-2NR’R”; - S(O)I-2(CM alkyl); -OH; and CM alkoxy; each occurrence of Re and Rf is independently selected from the group consisting of: H; C1-6 alkyl optionally substituted with from 1-3 substituents each independently selected from the group consisting of NR’R”, -OH, C1-6 alkoxy, C1-6 haloalkoxy, and halo; -C(O)(C1-4 alkyl); -C(0)0(CM alkyl); -CONR’R”; -S(O)I-2NR’R”; - S(O)I-2(CM alkyl); -OH; andC1-4 alkoxy; each occurrence of Rg is independently selected from the group consisting of:
• C3-8 cycloalkyl or C3-8 cycloalkenyl, each of which is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc;
• heterocyclyl or heterocycloalkenyl including from 3-8 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc;
• heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc; and
• Ce-io aryl optionally substituted with from 1-4 Rc; each occurrence of Lg is independently selected from the group consisting of: -O-, -NH-, -NRd, -S(0)o-2, C(O), and C1-3 alkylene optionally substituted with from 1-3 Ra; each g is independently 1, 2, or 3; each Rg2 is a divalent Rg group; each occurrence of R’ and R” is independently selected from the group consisting of: H; -OH; andC1-4 alkyl; In some embodiments, it is provided that when Ring A is unsubstituted phenyl; each of R2a, R2b, R3a, and R3b is H, and -O-L-R5 is -O-(unsubstituted C1-4 alkyl); OCH2CHF2; OCH2CF3; OCH2-cyclopropyl; OCH2CH2OCH3; or OCH2CH2OH, then one of the following applies: (i) Rla and Rlb cannot simultaneously be H; (ii) Rla and Rlb cannot simultaneously be CH3; and (iii) when one of Rla and Rlb is C3 alkenyl, -C(O)OH, -C(O)OMe, -C(O)N(Me)2, or -C(O)N(Me)(CH2CH2OH), then the other of Rla and Rlb cannot be H;
Variable Ring C
Figure imgf000032_0001
In some embodiments, Ring C is X1 (R7)n .
In some embodiments, Ring C is bicyclic heteroaryl including 7-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with X1 and further optionally substituted with from 1-4 RcA, wherein each RcA is an independently selected Rc.
In certain of the foregoing embodiments, Ring C is bicyclic heteroaryl including 9-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with X1 and further optionally substituted with from 1-4 RcA, wherein each RcA is an independently selected Rc.
In certain of the foregoing embodiments, Ring C is bicyclic heteroaryl including 9-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 RcA, wherein each RcA is an independently selected Rc In certain of these embodiments, Ring C is connected to
Figure imgf000033_0001
6-membered ring.
In certain embodiments, Ring
Figure imgf000033_0002
Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA; n is 0, 1, or 2; and each RcA is an independently selected Rc.
In certain of these embodiments, Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
As non-limiting examples of the foregoing embodiments, Ring C can be selected from the group consisting
Figure imgf000033_0003
Figure imgf000033_0004
wherein each RcA is an independently selected Rc. For example, Ring C can
Figure imgf000034_0001
Figure imgf000034_0002
As non-limiting examples of the foregoing embodiments, Ring C is selected from
Figure imgf000034_0003
optionally substituted with RcA, wherein each RcA is an independently selected Rc.
In certain of these embodiments, Ring
Figure imgf000034_0004
wherein RcA is an independently selected Rc.
In certain of these embodiments, Ring
Figure imgf000034_0005
RCA is an independently selected Rc.
Figure imgf000035_0001
5 p y g p consisting of: halo, NReRf, C1-4 alkoxy, C1-4 haloalkoxy, C1-3 alkyl, C1-3 alkyl substituted with from 1-3 independently selected halo, C1-3 alkyl substituted withC1-4 alkoxy, and C1- 4 alkoxy substituted with C1-4 alkoxy, and wherein each occurrence of RcA is independently selected from the group consisting of:C1-4 alkoxyC; 1-4 haloalkoxy; C1-3 alkyl; and C1-3 alkyl substituted with from 1-3 independently selected halo.
In certain embodiments (when Ring
Figure imgf000035_0002
Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
As non-limiting examples of the foregoing embodiments, Ring C can be selected
Figure imgf000035_0003
Figure imgf000036_0001
each further optionally substituted with RcA, wherein each
RCA is an independently selected Rc. For example, Ring C can
Figure imgf000036_0002
Figure imgf000036_0003
In certain embodiments, Ring
Figure imgf000036_0004
Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA; n2 is 0 or 1; and each RCA is an independently selected Rc.
In certain of these embodiments, Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA. As non-limiting examples of the foregoing embodiments, Ring C can be selected
Figure imgf000037_0001
RCA is an independently selected Rc. For example, Ring C can
Figure imgf000037_0002
Figure imgf000037_0003
In certain embodiments (when Ring
Figure imgf000037_0004
Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
As non-limiting examples of the foregoing embodiments, Ring C can be selected from the group consisting of:
Figure imgf000037_0005
Figure imgf000038_0001
each further optionally substituted with RcA, wherein each
RCA is an independently selected Rc.
In certain embodiments, Ring C is selected from the group consisting of:
Figure imgf000038_0002
Figure imgf000038_0003
each further optionally substituted with RcA, wherein each RcA is an independently selected Rc.
In certain embodiments (when Ring C is bicyclic heteroaryl including 9-10 ring atoms, wherein from 1 -4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 RcA, wherein each RcA is an independently selected
Rc), Ring C is connected t
Figure imgf000038_0004
via a 5-membered ring.
As non-limiting examples of the foregoing embodiments, Ring C can be selected from the group consisting of:
Figure imgf000038_0005
Figure imgf000039_0001
Figure imgf000039_0002
, each further optionally substituted with RcA, wherein each RcA is an independently selected Rc. In certain embodiments, Ring C is bicyclic heteroaryl including 9-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is substituted with X1 and further optionally substituted with from 1-4 RcA, wherein each RcA is an independently selected Rc. In certain of these embodiments, Ring
Figure imgf000039_0003
Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA; n is 0, 1, or 2; and each RcA is an independently selected Rc.
In certain of the foregoing embodiments, Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA. As non-limiting examples of the foregoing embodiments, Ring C can be selected from the group consisting of:
Figure imgf000040_0001
each further optionally substituted with RcA, wherein each RcA is an independently selected
Rc. For example, Ring C can
Figure imgf000040_0002
As non-limiting examples of the foregoing embodiments, Ring
Figure imgf000040_0003
Figure imgf000040_0004
each of which is further optionally substituted with from 1-2 RcA, wherein each RcA is an independently selected Rc.
In certain of these embodiments, Ring
Figure imgf000040_0005
In certain of these embodiments, Ring
Figure imgf000040_0006
In certain of these embodiments, Ring
Figure imgf000041_0001
wherein RcA is an independently selected Rc.
In certain embodiments (when Ring
Figure imgf000041_0002
Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
As non-limiting examples of the foregoing embodiments, Ring C can be selected from the group consisting of:
Figure imgf000041_0003
and
Figure imgf000041_0004
each further optionally substituted with RcA, wherein each RcA is an independently selected Rc. For example, Ring C can
Figure imgf000041_0005
Figure imgf000041_0006
In certain embodiments, Ring
Figure imgf000042_0001
Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA; n2 is 0 or 1; and each RCA is an independently selected Rc.
In certain of these embodiments, Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
As non-limiting examples of the foregoing embodiments, Ring C can be selected
Figure imgf000042_0002
optionally substituted with RcA, wherein each RcA is an independently selected Rc.
In certain embodiments (when Ring
Figure imgf000042_0003
unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA. In some embodiments, Ring C is heterocyclyl or heterocycloalkenyl including from 5-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with X1 and further optionally substituted with from 1 -4 substituents independently selected from the group consisting of oxo and RcA, wherein each RcA is an independently selected Rc.
In certain of these embodiments, Ring C is heterocyclyl including from 4-8, such as 5-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with X1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and RcA, wherein each RCA is an independently selected Rc. For example, Ring C can be
Figure imgf000043_0001
Figure imgf000043_0002
In certain embodiments, m is 1. In some embodiments, m is 0.
In some embodiments,
Figure imgf000043_0003
In certain of these embodiments, R6 is -Rg2-Rw.
In certain of the foregoing embodiments, -R6 is
Figure imgf000043_0004
, wherein Ring
D is heterocyclylene including from 3-10 ring atoms, wherein from 0-2 ring atoms (in addition to the ring nitrogen atom bonded to Rw) are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclylene is optionally substituted with from 1-3 substituents each independently selected from the group consisting of: oxo and -Rc. In certain of the foregoing embodiments, -R6 is
Figure imgf000044_0001
, wherein Ring
D is heterocyclylene including from 3-10 ring atoms, wherein from 0-2 ring atoms (in addition to the ring nitrogen atom bonded to Rw) are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclylene is optionally substituted with from 1-3 substituents each independently selected from the group consisting of: oxo and -Rc; optionally wherein -R6 is a monocyclic heterocyclylene ring including from 3-10 ring atoms as defined above with a nitrogen atom bonded t
Figure imgf000044_0002
Figure imgf000044_0003
optionally wherein -R6 is a bicyclic heterocyclylene ring including from 3-10 ring atoms as defined above with a nitrogen atom bonded to Rw (e.g.
Figure imgf000044_0004
wherein xl and x2 are each independently 0, 1 , or 2.
In certain embodiments, xl = 0, and x2 = 0; or xl = 0, and x2 = 1; or xl = 0, and x2 = 2. In certain embodiments, R6 is selected from the group consisting of:
Figure imgf000045_0001
Figure imgf000045_0002
In certain embodiments (e.g., when R6 is -Rg2-Rw), -Rw is -Lw-W; and Lw is C(=O).
In certain embodiments, (e.g., when R6 is -Rg2-Rw), -Rw is -Lw-W; and Lw is C(=O) NHC(=O)*, NRdC(=O)* (e.g., NMeC(=O)*), or NHS(O)I-2* wherein the asterisk represents point of attachment to W.
In certain of these embodiments, W is C2-6 alkenyl optionally substituted with from 1-3 Ra and further optionally substituted with Rg, wherein W is attached to Lw via an sp2 hybridized carbon atom.
In certain of these embodiments, W is C2-6 alkenyl or C2-6 optionally substituted with from 1 -3 Ra and further optionally substituted with Rg, wherein W is attached to Lw via an sp2 or sp hybridized carbon atom.
In certain of the foregoing embodiments, W is C2-4 alkenyl optionally substituted with from 1-3 Ra and further optionally substituted with Rg, wherein W is attached to Lw via an sp2 hybridized carbon atom. For example, W can be CH=CH2.
In certain of the foregoing embodiments, W is C2 -4 alkenyl (e.g., CH=CH2) or C2-4 alkynyl alkynyl (e.g.,
Figure imgf000045_0003
optionally substituted with from 1-3 Ra and further optionally substituted with Rg, wherein W is attached to Lw via an sp2 or sp hybridized carbon atom.
In certain embodiments, -Lw-W is -C(=0)CH=CH2.
In certain embodiments, -Lw-W is -C(=0)CH=CH2; -C(=0)NHCH=CH2; C(=0)CH=CHCH2NReRf (e g., C(=O)CH=CHCH2N(HMe), C(=O)CH=CHCH2NMe2,
Figure imgf000046_0001
In certain embodiments, X2 is selected from the group consisting of: -O-, -N(RN)-, and -S(0)o-2. In certain of these embodiments, X2 is -N(RN)-. For example, X2 can be - N(H)-. As another non-limiting example, X2 can be -O-.
In certain embodiments, X2 is selected from the group consisting of: -OC(=O)-*, - N(RN)C(=O)-*, and -N(RN)S(O)I-2-*. In certain of these embodiments, X2 is - N(RN)C(=O)-*. For example, X2 can be -N(H)C(=O)-*. In certain embodiments, X2 is - N(RN)S(O)2-*. For example, X2 can be -NHS(0)2-.
In certain embodiments, X2 is selected from the group consisting of: - OC(=O)N(RN)-*, -N(RN)C(=O)O-*, -N(RN)C(=O)N(RN)-*, and -N(RN)S(O)I-2N(RN)-*. In certain of these embodiments, X2 is -N(RN)C(=O)O-*. For example, X2 can be - N(H)C(=O)O-*. X2 is -N(RN)C(=O)N(RN)-*, such as -N(H)C(=O)N(H)-*.
In certain embodiments,
Figure imgf000046_0002
certain of these embodiments, X2 is -C(=O)N(RN)-*. For example, X2 can be -C(=O)N(H)- *
In certain embodiments,
Figure imgf000046_0003
In certain embodiments, X2 is C2-6 alkenyl ene optionally substituted with from 1-3
Ra. For example, X2 can
Figure imgf000046_0004
In certain embodiments, L1 is a bond.
In certain embodiments, L1 is C1-10 alkylene optionally substituted with from 1-6 Ra
In certain of these embodiments, L1 is C1-3 alkylene optionally substituted with from 1-6 Ra. In certain of the foregoing embodiments, L1 is unsubstituted C1-3 alkylene. As non-limiting examples of the foregoing embodiments, L1 can be -CH2-, -CH2CH2-, - CH2CF2-, or -CH(Me)-. For example, L1 can be -CH2-, -CH2CH2-, or -CH(Me)-.
In certain embodiments, L1 is branched C3-6 alkylene optionally substituted with from 1-6 Ra. For example, L1 can be
Figure imgf000047_0001
wherein aa is the point of attachment to R5.
In certain embodiments, R5 is -C1-6 alkoxy or -S(0)o-2(C1-6 alkyl), each optionally substituted with from 1-6 Ra. In certain of these embodiments, R5 is -C1-6 alkoxy optionally substituted with from 1-6 Ra. As a non-limiting example of the foregoing embodiments, R5 can be -C1-3 alkoxy. For example, R5 can be methoxy.
In certain embodiments, R5 is H or halo. As non-limiting examples of the foregoing embodiments, R5 can be H or -F. For example, R5 can be H.
In certain embodiments, R5 is -OH or -NReRf. For example, R5 can be -OH.
In certain embodiments, R5 is -Rg.
In certain of these embodiments, R5 is selected from the group consisting of:
• heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc; and
• Ce-io aryl optionally substituted with from 1-4 Rc.
In certain of the foregoing embodiments, R5 is Ce-io aryl optionally substituted with from 1-4 Rc. In certain of these embodiments, R5 is phenyl optionally substituted with from 1-4 Rc. As non-limiting examples of the foregoing embodiments, R5 can be phenyl optionally substituted with from 1-2 independently selected halo, such as -F.
In certain embodiments, R5 is heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc.
In certain of the foregoing embodiments, R5 is heteroaryl including from 5-6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc.
In certain of these embodiments, R5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(Rd), and wherein the heteroaryl is optionally substituted with
Figure imgf000048_0001
In certain embodiments, R5 is heteroaryl including 5 ring atoms, wherein from 1 -4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc. For example, R5 can
Figure imgf000048_0002
Figure imgf000048_0003
In certain embodiments, R5 is selected from the group consisting of: • C3-10 cycloalkyl or C3-10 cycloalkenyl, each of which is optionally substituted with from 1 -4 substituents independently selected from the group consisting of oxo and Rc; and
• heterocyclyl or heterocycloalkenyl including from 3-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
In certain of these embodiments, R5 is C3-10 cycloalkyl or C3-10 cycloalkenyl, each of which is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
In certain of the foregoing embodiments, R5 is C3-10 cycloalkyl (e.g., C3-6 cycloalkyl) optionally substituted with from 1-4 Rc, such as wherein R5 is cyclopropyl.
In certain embodiments, R5 is heterocyclyl or heterocycloalkenyl including from 3- 10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
In certain of these embodiments, R5 is heterocyclyl including from 4-8, such as 4- 6, ring atoms, wherein from 1 -3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc. For example, R5 can be
Figure imgf000049_0001
Figure imgf000049_0002
In certain embodiments, R5 is selected from the group consisting of: -Rg2-Rw and -Rg2-RY. In certain of these embodiments, R5 is -Rg2-RY.
In certain embodiments, the -Rg2 group present in R5 is Ce-io arylene optionally substituted with from 1-4 Rc.
In certain of these embodiments, the -Rg2 group present in R5 is phenylene optionally substituted with from 1-4 Rc.
In certain of the foregoing embodiments, the -Rg2 group present in R5 is 1,3- phenylene or 1 ,4-phenylene, each optionally substituted with from 1-4 Rc. As non-limiting examples of the foregoing embodiments, -Rg2 can be
Figure imgf000050_0001
Figure imgf000050_0002
wherein bb is the point of attachment to RY.
In certain embodiments, the RY group present in R5 is -Rg.
In certain of these embodiments, the RY group present in R5 is heterocyclyl or heterocycloalkenyl including from 3-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
In certain of the foregoing embodiments, the RY group present in R5 is heterocyclyl including from 4-8, such as 4-6, ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc, such as wherein RY is
Figure imgf000050_0003
In certain embodiments, R5 is -L5-Rg.
In certain of these embodiments, R5 is -O-Rg. In certain embodiments, R5 is -0-(Ce-io aryl) wherein the Ce-io aryl is optionally substituted with from 1-4 Rc.
As a non-limiting example of the foregoing embodiments, R5 can be -O-phenyl wherein the phenyl is optionally substituted with from 1-2 Rc. For example, R5 can be
Figure imgf000051_0001
Non-Limiting Combinations of m, X2, L1, and Rs
[AA]:
In certain embodiments, X1 is -(X2)m-L1-R5, wherein:
• m is 0 or 1;
• X2 is -N(RN)- or -O-;
• L1 is a bond or C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is - R"
In certain embodiments of [AA], R5 is phenyl optionally substituted with from 1-4 Rc, such as wherein R5 is phenyl optionally substituted with from 1-2 independently selected halo, such as -F.
In certain embodiments of [AA], R5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(Rd), and wherein the heteroaryl is optionally substituted with from 1 -4 Rc, such as wherein
Figure imgf000051_0002
In certain embodiments of [AA], R5 is heteroaryl including 5 ring atoms, wherein from 1-4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc, such as wherein
Figure imgf000052_0001
Figure imgf000052_0002
In certain embodiments of [AA], R5 is C3-10 cycloalkyl, such as C3-6 cycloalkyl, optionally substituted with from 1-4 Rc, such as wherein R5 is cyclopropyl.
In certain embodiments of [AA], R5 is heterocyclyl including from 4-8, such as 4-
6, ring atoms, wherein from 1 -3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc. For example, R5 can
Figure imgf000052_0003
Figure imgf000052_0004
In certain embodiments of [AA], m is 0.
In certain embodiments of [AA], m is 1.
In certain embodiments of [AA], X2 is -N(RN)- (e.g., N(H)).
In certain embodiments of [AA], X2 is -O-.
In certain embodiments of [AA], L1 is a bond.
In certain embodiments of [AA], L1 is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or -
CH(Me)-).
In certain embodiments of [AA], L1 is branched C3-6 alkylene. For example, L1 can
Figure imgf000052_0005
[BB]:
In certain embodiments, X1 is -X -L -R5, wherein:
• X2 is -N(RN)C(=O)-*, -N(RN)S(O)2-*, -N(RN)C(=O)O-*, or N(RN)C(=O)N(RN)*;
• L1 is a bond or C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is - R"
In certain embodiments of [BB], R5 is phenyl optionally substituted with from 1-4 Rc, such as wherein R5 is phenyl optionally substituted with from 1-2 independently selected halo, such as -F.
In certain embodiments of [BB], R5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(Rd), and wherein the heteroaryl is optionally substituted with
Figure imgf000053_0001
In certain embodiments of [BB], R5 is heteroaryl including 5 ring atoms, wherein from 1-4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc, such as wherein
Figure imgf000053_0002
Figure imgf000053_0003
In certain embodiments of [BB], R5 is C3-10 cycloalkyl, such as C3-6 cycloalkyl, optionally substituted with from 1-4 Rc, such as wherein R5 is cyclopropyl.
In certain embodiments of [BB], R5 is heterocyclyl including from 4-8, such as 4- 6, ring atoms, wherein from 1 -3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc. For example, R5 can
Figure imgf000054_0001
Figure imgf000054_0002
In certain embodiments of [BB], X2 is -N(RN)C(=O)-* (e.g., -N(H)C(=O)-*).
In certain embodiments of [BB], X2 is -N(RN)S(O)2-, such as -N(H)S(O)2-*.
In certain embodiments of [BB], X2 is -N(RN)C(=O)O-*, or -N(RN)C(=O)N(RN)-* (e g., -N(H)C(=O)O-*; e g., -N(H)C(=O)N(H)-*).
In certain embodiments of [BB], L1 is a bond.
In certain embodiments of [BB], L1 is Ci-3 alkylene (e g., -CH2-, -CH2CH2-, or -
CH(Me)-).
In certain embodiments of [BB], L1 is branched C3-6 alkylene. For example, L1 can
Figure imgf000054_0003
wherein aa is the point of attachment to R5.
[CC]:
In certain embodiments, X1 is -X2-L1-R5, wherein:
Figure imgf000054_0004
• L1 is a bond or C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is - R" In certain embodiments of [CC], R5 is phenyl optionally substituted with from 1-4
Rc, such as wherein R5 is phenyl optionally substituted with from 1-2 independently selected halo, such as -F.
In certain embodiments of [CC], R5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(Rd), and wherein the heteroaryl is optionally substituted with
Figure imgf000055_0001
In certain embodiments of [CC], R5 is heteroaryl including 5 ring atoms, wherein from 1-4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc, such as wherein
Figure imgf000055_0002
Figure imgf000055_0003
In certain embodiments of [CC], R5 is C3-10 cycloalkyl, such as C3-6 cycloalkyl, optionally substituted with from 1-4 Rc, such as wherein R5 is cyclopropyl.
In certain embodiments of [CC], R5 is heterocyclyl including from 4-8, such as 4- 6, ring atoms, wherein from 1 -3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc. For example, R5 can
Figure imgf000056_0001
such as
Figure imgf000056_0002
In some embodiments
In some embodiments
Figure imgf000056_0003
In certain embodiments of [CC], L1 is a bond.
In certain embodiments of [CC], L1 is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or -
CH(Me)-).
In certain embodiments of [CC], L1 is branched C3-6 alkylene. For example, L1 can
Figure imgf000056_0004
wherein aa is the point of attachment to R5.
[DD]:
In certain embodiments, X1 is -(X2)m-L1-R5, wherein:
• m is 0 or 1;
• X2 is -N(RN)- or -O-;
• L1 is a bond or C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is R"2-Rv
In certain embodiments of [DD], the -Rg2 group present in R5 is 1,3-phenylene or 1,4-phenylene, each optionally substituted with from 1-4 Rc, such as wherein -Rg2 is
Figure imgf000057_0001
wherein bb is the point of attachment to RY.
In certain embodiments of [DD], the RY group present in R5 is -Rg.
In certain embodiments of [DD], the RY group present in R5 is heterocyclyl including from 4-8, such as 4-6, ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc. For example, RY can be
Figure imgf000057_0002
In certain embodiments of [DD], X2 is -N(RN)- (e.g., N(H)).
In certain embodiments of [DD], X2 is -O-.
In certain embodiments of [DD], L1 is a bond.
In certain embodiments of [DD], L1 is Ci-3 alkylene (e g., -CH2-, -CH2CH2-, or -
CH(Me)-).
In certain embodiments of [DD], L1 is branched C3-6 alkylene. For example, L1 can
Figure imgf000057_0003
, wherein aa is the point of attachment to R5.
[EE]:
In certain embodiments, X1 is -X2-L1-R5, wherein:
• X2 is -N(RN)-, -O-, -N(RN)C(=O)-*, -N(RN)S(O)2-, -N(RN)C(=O)O-*, or -N(RN)C(=O)N(RN)-*;
• L1 is C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is H, halo, C1-6 alkoxy optionally substituted with from 1-3 Ra, or -OH. In certain embodiments of [EE], R5 is H.
In certain embodiments of [EE], R5 is halo (e.g., -F).
In certain embodiments of [EE], R5 is C1-6 alkoxy optionally substituted with from 1-3 Ra, such as wherein R5 is C1-3 alkoxy such as methoxy.
In certain embodiments of [EE], R5 is -OH.
In certain embodiments of [EE], X2 is -N(RN)- (e.g., N(H)).
In certain embodiments of [EE], X2 is -O-.
In certain embodiments of [EE], X2 is -N(RN)C(=O)-* (e.g., -N(H)C(=O)-*).
In certain embodiments of [EE], X2 is -N(RN)S(O)2-, such as -N(H)S(O)2-*.
In certain embodiments of [EE], X2 is -N(RN)C(=O)O-*, or -N(RN)C(=O)N(RN)-* (e.g., -N(H)C(=O)O-*; e g., -N(H)C(=O)N(H)-*).
In certain embodiments of [EE], L1 is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or - CH(Me)-).
In certain embodiments of [EE], L1 is branched C3-6 alkylene. For example, L1 can
Figure imgf000058_0001
, wherein aa is the point of attachment to R5.
[FF]:
In certain embodiments, X1 is -L1-R5, wherein L1 is C1-6 alkylene optionally substituted with from 1-3 Ra; and R5 is -L5-Rg.
In certain embodiments of [FF], R5 is O-R"
In certain embodiments of [FF], R5 is -O-(phenyl), wherein the phenyl is optionally substituted with from 1-2 Rc.
In certain embodiments of [FF], L1 is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or - CH(Me)-). Variable RcA
In certain embodiments, each occurrence of RcA is independently selected from the group consisting of: halo; cyano; Ci-io alkyl which is optionally substituted with from 1-6 independently selected Ra; C1-4 alkoxy optionally substituted with C1-4 alkoxy or C1-4 haloalkoxy; C1-4 haloalkoxy; -S(0)I-2(CM alkyl); -NReRf; -OH; -S(0)I-2NR’R”; -CM thioalkoxy; -C(=0)(Ci-io alkyl); -C(=O)O(CM alkyl); -C(=O)OH; and -C(=O)NR’R”
In certain embodiments, one occurrence of RcA is -NReRf.
In certain of these embodiments, one occurrence of RcA is -NH2.
In certain of the foregoing embodiments, one occurrence of RcA is -NH(CI-6 alkyl), wherein the C1-6 alkyl is optionally substituted with from 1-3 substituents each independently selected from the group consisting of NR’R”, -OH, C1-6 alkoxy, C1-6 haloalkoxy, and halo. For example, one occurrence of RcA can be -NHMe, -NHCH2CF3, -NHCH2CH2OH, or -NHiPr.
In certain embodiments, one occurrence of RcA is -NHC(=O)CM alkyl, such as NHC(=O)CH3
In certain embodiments, one occurrence of RcA is N(CI-3 alkyl)2 such as NMe2.
In certain embodiments, one occurrence of RcA is C a1l-4koxy optionally substituted withC1-4 alkoxy or C1-4 haloalkoxy. For example, RcA can be OMe or OCH2CH2OMe.
In certain embodiments, one occurrence of RcA is C1-4 haloalkoxy (e.g., - OCH2CF3).
In certain embodiments, one occurrence of RcA is C1 t-h4ioalkoxy (e.g., -SCH3).
In certain embodiments, one occurrence of RcA is C1-4 alkyl, such as methyl; or wherein one occurrence of RcA is C1-4 alkyl substituted with from 1-6 independently selected halo. For example, one occurrence of RcA can be -CF3.
In certain embodiments, one occurrence of RcA is C1-4 alkyl substituted with Ra, such asC1-4 alkyl substituted with C1-3 alkoxy or C(=O)NR’R”. For example, one occurrence of RCA can
Figure imgf000059_0001
In certain embodiments, one occurrence of RcA is halo (e.g., -F). In certain embodiments, one occurrence of RcA is -OH.
In certain embodiments, one occurrence of RcA is C(=O)NR’R” (e.g., C(=O)NHMe).
Variables Rla> Rlb, R2a, R2b, R3a, and R3b
In some embodiments, Rla and Rlb are both H.
In some embodiments, R2a and R2b are both H.
In some embodiments, from 1-2 (e.g., 1 or 2) of R2a and R2b is an independently selected substituent that is other than H.
In certain of these embodiments, one of R2a and R2b (e.g., R2a), is a substituent that is other than H.
In certain of the foregoing embodiments, one of R2a and R2b (e.g., R2a), is Rb. In certain of these embodiments, one of R2a and R2b (e.g., R2a) is C1-6 alkyl which is optionally substituted with from 1-6 Ra. In certain of these embodiments, one of R2a and R2b (e.g., R2a) is Ci -3 alkyl, such as methyl or ethyl. In certain embodiments (when one of R2a and R2b is as defined supra), the other of R2a and R2b (e.g., R2b) is H.
In some embodiments, R3a and R3b are both H.
In some embodiments, from 1-2 (e.g., 1 or 2) of R3a and R3b is an independently selected substituent that is other than H.
In certain of the foregoing embodiments, one of R3a and R3b (e.g., R3a) is a substituent that is other than H. In certain of these embodiments, one of R3a and R3b (e.g., R3a) is Rb. In certain of these embodiments, one of R3a and R3b (e.g., R3a) is C1-6 alkyl which is optionally substituted with from 1-6 Ra. For example, one of R3a and R3b (e.g., R3a) can be C1-3 alkyl, such as methyl or ethyl. In certain embodiments (when one of R3a and R3b is as defined supra), the other of R3a and R3b (e.g., R3b) is H.
In some embodiments, one of R3a and R3b, such as R3a, is C1-3 alkyl optionally substituted with from 1-3 independently selected halo. As non-limiting examples of the foregoing embodiments, one of R3a and R3b, such as R3a , is -CH3, -CH2CH3, -CH2F, - CHF2, -CF3, -CH2CHF2, or -CH2CH2F. In some embodiments, one of R3a and R3b, such as R3a, is C1-3 alkyl substituted with C1-4 alkoxy, C1-4 haloalkoxy, or NReRf. As non-limiting examples of the foregoing embodiments, one of R3a and R3b, such as R3a, is -CH2OMe, -CH2CH2OMe, - CH(Me)CH2OMe, -CH2CH(Me)OMe, -CH2OEt, -CH2CH2OCHF2, -CH2NReRf (e g., - CH2N(CF3)Me), or -CH2CH2NReRf (e g., -CH2CH2NMe2).
In some embodiments, one of R3a and R3b, such as R3a, is C1-3 alkyl substituted withC1-4 alkoxy. As non-limiting examples of the foregoing embodiments, one of R3a and R3b, such as R3a, is -CH2OMe, -CH2CH2OMe, -CH(Me)CH2OMe, -CH2CH(Me)OMe, or -CH2OEt, such as -CH2OMe.
In some embodiments, one of R3a and R3b, such as R3a, is C1-3 alkyl substituted withC1-4 alkoxy. As non-limiting examples of the foregoing embodiments, one of R3a and R3b, such as R3a, is -CH2OMe, -CH2CH2OMe, -CH(Me)CH2OMe, -CH2CH(Me)OMe, or -CH2OEt, such as -CH2OMe; such as -CH2CH2OMe; optionally the other one of R3a and R3b, such as R3b is H.
In some embodiments, one of R3a and R3b, such as R3a, is C1-3 alkyl substituted withC1-4 alkoxy, C1-4 haloalkoxy, or NReRf and further substituted with from 1-3 independently selected halo. In certain embodiments, one of R3a and R3b, such as R3a, is C1-3 alkyl substituted with C1-4 alkoxy and further substituted with from 1-3 independently '''O selected halo. For example, one of R3a and R3b, such as R3a, can be V —1—F (e.g., V — I—
Figure imgf000061_0001
In some embodiments, one of R3a and R3b, such as R3a, is C3-6 alkyl substituted withC1-4 alkoxy, C1-4 haloalkoxy, or NReRf. In certain of these embodiments, one of R3a and R3b, such as R3a, is branched C3-6 alkyl substituted with C a1-l4koxy, C h1-a4loalkoxy, or NReRf. In certain of the foregoing embodiments, one of R3a and R3b, such as R3a, is branched C3-6 alkyl substituted with C1-4 alkoxy. For example, one of R3a and R3b, such as
R3a, can
Figure imgf000062_0001
In some embodiments, one of R3a and R3b, such as R3a, is Rg or -(Lg)g-Rg.
In certain of the foregoing embodiments, one of R3a and R3b, such as R3a, is selected from the group consisting of: heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc; and
C3-6 cycloalkyl optionally substituted with from 1-4 Rc.
As non-limiting examples of the foregoing embodiments, one of R3a and R3b, such as R3a, is selected from the group consisting of: cyclopropyl, cyclobutyl, oxetanyl, and azetidinyl, each of which is optionally substituted with from 1 -2 substituents independently selected from the group consisting of: C1-3 alkyl and halo, wherein the ring nitrogen of the azetidinyl is optionally substituted with Rd.
In certain of the foregoing embodiments, one of R3a and R3b, such as R3a, is -(C1-3 alkylene)-Rg or -(C1-3 alkylene)-O-Rg, and optionally the Rg group of R3a or R3b is:
C3-6 cycloalkyl optionally substituted with from 1-4 Rc, or heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
In certain of the foregoing embodiments, one of R3a and R3b, such as R3a, is -CH2- Rg, -CHoCHoR8, or -CH2-0-Rg, wherein the Rg group of R3a or R3b is selected from the group:
C3-6 cycloalkyl (e.g., cyclopropyl, cyclobutyl) optionally substituted with from 1-4 Rc, or heterocyclyl including from 4-6 ring atoms (e.g., oxetanyl, azetidinyl), wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1 -4 substituents independently selected from the group consisting of oxo and Rc (e.g., C1-3 alkyl, halo).
In certain of the foregoing embodiments, one of R3a and R3b, such as R3a, is -CH2- Rg, -CH2CH2Rg, or -CH2-0-Rg, wherein the Rg group of R3a or R3b is selected from the group consisting of: cyclopropyl, cyclobutyl, oxetanyl, and azetidinyl, each of which is optionally substituted with from 1-2 substituents independently selected from the group consisting of: C1-3 alkyl and halo, wherein the ring nitrogen of the azetidinyl is optionally substituted with Rd
In certain of the foregoing embodiments, one of R3a and R3b, such as R3a, is -CH2-Rg, -CHzCHzR8, or -CH2-0-Rg, wherein the Rg group of R3a or R3b is selected from the group consisting of: cyclopropyl, cyclobutyl, oxetanyl, 1 ,4-dioxanyl, and azetidinyl, each of which is optionally substituted with from 1-2 substituents independently selected from the group consisting of: C1-3 alkyl and halo, wherein the ring nitrogen of the azetidinyl is optionally substituted with Rd.
As non-limiting examples of the foregoing embodiments, one of R3a and R3b, such as R3a, can be selected from the group consisting of:
Figure imgf000063_0001
, such as
Figure imgf000063_0002
As further non-limiting examples of the forgoing embodiments, one of R3a and R3b, such as R3a, can be selected from the group consisting of:
Figure imgf000064_0001
In some embodiments, one of R3a and R3b, such as R3a, is-(Lg)g-Rw.
In certain embodiments, one of R3a and R3b, such as R3a, is -(C1-3 alkylene)-Rw ; optionally one of R3a and R3b, such as R3a, is -CH2-RW, or -CH2CH2-RW.
In certain embodiments, the Rw group of R3a or R3b is: C(=0)-CH=CH2, or - NHC(=O)-CH=CH2.
As a non-limiting example, one of R3a and R3b, such as R3a, can be
Figure imgf000064_0002
In some embodiments, one of R3a and R3b, such as R3a, is -(Lg)g-Rg2-Rw.
In some embodiments, one of R3a and R3b, such as R3a, is -(C1-3 alkyl ene)-Rg2-Rw, and optionally one of R3a and R3b, such as R3a, is -CH2-Rg2-Rw, or -CH2CH2-Rg2-Rw. In certain of these embodiments, the Rg2 group
Figure imgf000065_0001
such as
Figure imgf000065_0002
wherein the waveline represents the point of attachment to Lg (e.g., -CH2- or -CH2CH2-) and the asterisk represents the point of attachment to Rw; and wherein the Rw group of R3a or R3b is C(=0)-CH=CH2, or -
NHC(=O)-CH=CH2.
In certain of these embodiments, one of R3a and R3b, such as R3a, is -CH2-Rg2-Rw, and wherein the Rg2 group
Figure imgf000065_0003
Figure imgf000065_0004
wherein the waveline represents the point of attachment to Lg
(e.g., -CH2- or -CH2CH2-) and the asterisk represents the point of attachment to Rw; and wherein the Rw group of R3a or R3b is C(=0)-CH=CH2, or -NHC(=0)-CH=CH2.
As a non-limiting example, one of R3a and R3b, such
Figure imgf000065_0005
such as
Figure imgf000065_0006
In some embodiments, the other of R3a and R3b is -H.
In some embodiments, the other of R3a and R3b is C1-3 alkyl, such as methyl.
In some embodiments, the other of R3a and R3b is halo, such as -F.
In certain embodiments (when one of R3a and R3b is as defined anywhere supra), the other of R3a and R3b is selected from the group consisting of: -H; C1-3 alkyl (e.g., methyl); and -F.
In some embodiments, R3a and R3b, together with the Ring B ring atom to which each is attached, form a fused saturated or unsaturated ring of 3-12 ring atoms;
• wherein from 0-2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated or unsaturated ring of 3-12 ring atoms is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rw.
In certain of these embodiments, R3a and R3b, together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-8 ring atoms;
• wherein from 0-2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated ring of 4-8 ring atoms is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rw.
In certain of these embodiments, R3a and R3b, together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-6 ring atoms;
• wherein from 1-2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and wherein the fused saturated ring of 4-6 ring atoms is optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and
Rc
As non-limiting examples of the foregoing embodiments, R3a and R3b, together with the Ring B ring atom to which each is attached, form
Figure imgf000067_0001
In certain embodiments, R3a and R3b, together with the Ring B ring atom to which each is attached, form:
Figure imgf000067_0002
, which is optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc, wherein: pl and p2 are independently 0, 1, or 2;
Rz is H, Rd, C(=0)-W, or S(O)2W; and cc represents the point of attachment to C(R2aR2b).
In certain of these embodiments, R3a and R3b, together with the Ring B ring atom to which each is attached, form
Figure imgf000067_0003
, wherein Rz is H, Rd, C(=0)-W, or S(O)2W; and cc represents the point of attachment to C(R2aR2b).
In certain embodiments, R3a and R3b, together with the Ring B ring atom to which each is attached, form a fused ring selected from the group consisting of:
Figure imgf000067_0004
Figure imgf000068_0001
Figure imgf000068_0002
attachment to C(R2aR2b).
In certain embodiments, Rz is H.
In certain embodiments, Rz is Rd. In certain of these embodiments, Rz is C1-6 alkyl optionally substituted with from 1-3 independently selected Ra. In certain embodiments, Rz is C(=0)-W or S(O)2W. In certain embodiments, W is C2-4 alkenyl. As a non-limiting example of the foregoing embodiments, Rz can be
C(=O)-CH2=CH2.
In certain embodiments, R3a and R3b, together with the Ring B ring atom to which each is attached, form a fused C3-6 cycloalkyl, wherein the fused C3-6 cycloalkyl is optionally substituted with from 1-2 Rc.
As non-limiting examples of the foregoing embodiments, R3a and R3b, together with the Ring B ring atom to which each is attached, form
Figure imgf000068_0003
In certain of these foregoing embodiments, Rla, Rlb, R2a, and R2b are each H; and
R3a and R3b taken together with the Ring B ring carbon atom to which each is attached form a fused C3-6 (such as C3 or C4) cycloalkyl, wherein the fused cycloalkyl ring is optionally substituted with from 1-2 Rc.
In certain embodiments, one of R2a and R2b, such as R2a, and one of R3a and R3b, such as R3a, taken together with the Ring B ring atoms to which each is attached, form a fused saturated or unsaturated ring of 3-12 ring atoms;
• wherein from 0-2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated or unsaturated ring of 3-12 ring atoms is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
In certain the the foregoing embodiments, one of R2a and R2b (such as R2a) and one of R3a and R3b (such as R3a) taken together with the Ring B ring atoms to which each is attached, form a fused saturated ring of 3-8 ring atoms;
• wherein from 0-2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated ring of 3-8 ring atoms is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc
In certain of these foregoing embodiments, one of R2a and R2b , such as R2a, and one of R3a and R3b , such as R3a, taken together with the Ring B ring atoms to which each is attached, form a fused C3-6 cycloalkyl which is optionally substituted with from 1-2 Rc.
As non-limiting examples of the foregoing embodiments, one of R2a and R2b (such as R2a) and one of R3a and R3b (such as R3a) taken together with the Ring B ring atoms to which each is attached, form a fused cyclobutyl or cyclopropyl ring, e.g.,
Figure imgf000070_0001
In certain embodiments, the other one of R3a and R3b is Rg or -(Lg)g-Rg.
In certain embodiments, the other one of R3a and R3b is -(Lg)g-Rg.
In certain embodiment, the other one of R3a and R3b is -(C1-3 alkylene)-Rg or -(C1-3 alkylene)-O-Rg, and optionally the Rg group of R3a or R3b is:
C3-6 cycloalkyl optionally substituted with from 1-4 Rc, or heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
In certain embodiments, the other one of R3a and R3b, such as R3a, is -CH2-Rg, - CHiCHiR8, or -CH2-0-Rg, wherein the Rg group of R3a or R3b is:
C3-6 cycloalkyl optionally substituted with from 1-4 Rc, or heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
In certain enbodiments, the other one of R3a and R3b, such as R3a, is -CH2-Rg, - CH2CH2Rg, or -CH2-0-Rg, wherein the Rg group of R3a or R3b is selected from the group consisting of: cyclopropyl, cyclobutyl, oxetanyl, 1 ,4-dioxanyl, and azetidinyl, each of which is optionally substituted with from 1-2 substituents independently selected from the group consisting of: C1-3 alkyl and halo, wherein the ring nitrogen of the azetidinyl is optionally substituted with Rd.
In certain embodiments, the other one of R3a and R3b, such as R3a, is selected from the
Figure imgf000071_0001
In certain embodiments, Rla, Rlb, R2a, and R2b are each H, and R3a and R3b are independently selected C1-3 alkyl.
In certain embodiments, Rla, Rlb, R2a, and R2b are each H; one of R3a and R3b, such as R3a, is C1-3 alkyl optionally substituted with from 1-3 Ra; and the other of R3a and R3b is H, optionally each Ra substituent present in R3a or R3b is independently selected from the group consisting of: halo, C1-4 alkoxy, and C1-4 haloalkoxy.
In certain embodiments, Rla, Rlb, R2a, and R2b are each H; one of R3a and R3b, such as R3a, is C1-3 alkyl optionally substituted with from C1-4 alkoxy; optionally one of R3a and R3b, such as R3a, is -CHiCHi-OMe; and the other of R3a and R3b is H.
In certain embodiments, Rla, Rlb, R2a, and R2b are each H; one of R3a and R3b, such as R3a, is C1-3 alkyl optionally substituted with from 1-3 Ra; and the other of R3a and R3b is -F, optionally each Ra substituent present in R3a or R3b is independently selected from the group consisting of: halo, C1-4 alkoxy, and C1-4 haloalkoxy.
In certain embodiments, Rla, Rlb, R2a, and R2b are each H; one of R3a and R3b, such as R3a, is C1-3 alkyl optionally substituted with from 1-3 Ra; and the other of R3a and R3b is C1-3 alkyl (e.g., methyl), optionally each Ra substituent present in R3a or R3b is independently selected from the group consisting of: halo, C1 a-4lkoxy, and C h1-a4loalkoxy.
In certain embodiments, Rla, Rlb, R2a, and R2b are each H; one of R3a and R3b, such as R3a, is C3-6 (e.g., C4) alkyl optionally substituted with from 1-3 Ra; and the other of R3a and R3b is H, -F, or C1-3 alkyl (e.g., methyl), optionally each Ra substituent present in R3a or R3b is independently selected from the group consisting of: halo, C1-4 alkoxy, and CM haloalkoxy.
In certain embodiments, Rla, Rlb, R2a, and R2b are each H, and one of R3a and R3b, such as R3a, is -Rg, -(C1-3 alkylene)-Rg, or -(C1-3 alkylene)-O-Rg, optionally wherein the Rg group of R3a or R3b is:
C3-6 cycloalkyl optionally substituted with from 1-4 Rc, or heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc; and the other of R3a and R3b is H.
In some embodiments, Rla, Rlb, R2a, and R2b are each H; and R3a and R3b together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-6 ring atoms;
• wherein from 1 -2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated ring of 4-6 ring atoms is optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc In certain embodoments, Rla, Rlb, R2a, and R2b are each H; and R3a and R3b taken together with the Ring B ring carbon atom to which each is attached form a fused C3-6 (such as C3 or C4) cycloalkyl, wherein the fused cycloalkyl ring is optionally substituted with from 1 -2 Rc.
In certain embodiments, Rla, Rlb, R2a, and R2b are each H; and R3a and R3b are independently selected C1-3 alkyl.
In some embodiments, Rla, Rlb is H, and one of R2a and R2b (such as R2a) and one of R3a and R3b (such as R3a) taken together with the Ring B ring atoms to which each is attached, form a fused C3-6 (such as C3 or C4) cycloalkyl which is optionally substituted with from 1 -2 Rc; and the other of R2a and R2b and the other of R3a and R3b are each H.
In some embodiments, the other of R2a and R2b and the other of R3a and R3b are each H.
In the some embodiments, the other of R3a and R3b is C1-3 alkyl. As non-limiting examples of the foregoing embodiments, the other of R3a and R3b is -CH3, -CH2CH3.
In some embodiments, Rla, Rlb is H; one of R2a and R2b (such as R2a) and one of R3a and R3b (such as R3a) taken together with the Ring B ring atoms to which each is attached, form a fused C3-6 (such as C3 or C4) cycloalkyl which is optionally substituted with from 1 -2 Rc; and the other of R2a and R2b and the other of R3a and R3b are each H.
In some embodiments, Rla, Rlb, R2a, R2b, R3a, and R3b are each H.
Figure imgf000073_0001
Figure imgf000074_0001
Variable R4, and Ring A
In some embodiments, R4 is hydrogen.
In certain embodiments, R4 is hydrogen; and R4 is hydrogen.
(RcB)mi
In some embodiments, Ring A is
Figure imgf000074_0002
, wherein each RcB is an independently selected Rc; and ml is 0, 1, 2, 3, or 4.
In certain of these embodiments, ml is 1, 2, or 3. For example, ml can be 1 or 2 (e g., 2).
In certain embodiments, Ring
Figure imgf000074_0003
), wherein each RcB is an independently selected Rc.
As non-limiting examples, Ring A can be
Figure imgf000074_0004
In certain embodiments, Ring A is selected from the group consisting of:
Figure imgf000075_0001
In certain embodiments, each RcB is independently selected from the group consisting of: -halo, such as -Cl and -F; -CN; C1-4 alkoxy; C1-4 haloalkoxy; C1-3 alkyl; and C1-3 alkyl substituted with from 1-6 independently selected halo.
In certain embodiments, Ring A is
Figure imgf000075_0002
, wherein RcB1 is Rc; and RcB2 is H or Rc, optionally wherein RcB1 and RcB2 are each independently selected from the group consisting of: -halo, such as -Cl and -F; -CN; C1-4 alkoxy; C1-4 haloalkoxy; C1-3 alkyl; and C1-3 alkyl substituted with from 1-6 independently selected halo.
In certain embodiments (when Ring
Figure imgf000075_0003
halo, such as -F or
-Cl, such as -F.
In certain embodiments, RcB1 is C1-3 alkyl or C1-3 alkyl substituted with from 1-6 independently selected halo. For example, RcB1 can be methyl, -CHF2, or -CF3.
In certain embodiments, RcB2 is selected from the group consisting of: halo; -CN; CM alkoxy;C1-4 haloalkoxy; C1-3 alkyl; and C1-3 alkyl substituted with from 1-6 independently selected halo. In certain of these embodiments, RcB2 is C1-4 alkoxy or CM haloalkoxy.
In certain embodiments, RcB2 is selected from the group consisting of cyano; C1-3 alkyl; and C1-3 alkyl substituted with from 1-6 independently selected halo. For example, RcB2 can be cyano, methyl, ethyl, -CHF2, -CF3, or -CH2CHF2. In some embodiments, Ring A is heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1 -4 substituents independently selected from the group consisting of Rc and oxo.
In certain of these embodiments, Ring A is bicyclic heteroaryl including from 9-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 substituents independently selected from the group consisting of Rc and oxo.
In certain embodiments, Ring A is selected from the group consisting of:
Figure imgf000076_0001
each of which is further optionally substituted with Rc.
Non-Limiting Combinations
In certain embodiments, the compound is a compound of Formula (I-a):
Figure imgf000076_0002
or a pharmaceutically acceptable salt thereof, wherein: each RcA is an independently selected Rc; n is 0, 1, or 2; and
Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
In certain embodiments of Formula (I-a), Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
Figure imgf000077_0001
Figure imgf000077_0002
each further optionally substituted with RcA, wherein each RcA is an independently selected Rc.
In certain of these embodiments,
Figure imgf000077_0003
selected from the group consisting
Figure imgf000077_0004
Figure imgf000078_0001
Figure imgf000078_0002
each further optionally substituted with RcA, wherein each RcA is an independently selected Rc.
In certain of the foregoing embodiments,
Figure imgf000078_0003
wherein RcA is an independently selected Rc.
In certain of the foregoing embodiments,
Figure imgf000078_0004
wherein each RcA is an independently selected Rc.
In certain of the foregoing embodiments,
Figure imgf000078_0005
selected from the group consisting of:
Figure imgf000079_0001
each occurrence of RcA is independently selected from the group consisting of: halo; NReRf; C1-4 alkoxy; C1- h4aloalkoxy; C1-3 alkyl; C1-3 alkyl substituted with from 1-3 independently selected halo; C1-3 alkyl substituted with C1-4 alkoxy; and C1-4 alkoxy substituted with C1-4 alkoxy; such as wherein each occurrence of RcA is independently selected from the group consisting of: C1-4 alkoxy; C1-4 haloalkoxy; C1-3 alkyl; and C1-3 alkyl substituted with from 1-3 independently selected halo. In certain embodiments of Formula (I-a), Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
In certain of these embodiments,
Figure imgf000079_0002
selected from the group consisting
Figure imgf000079_0003
each further optionally substituted with RcA, wherein each RcA is an independently selected
Rc In certain of these embodiments,
Figure imgf000080_0001
selected from the group consisting
Figure imgf000080_0002
Figure imgf000080_0003
each further optionally substituted with RcA, wherein each RcA is an independently selected Rc.
In certain embodiments, the compound is a compound of Formula (I-b):
Figure imgf000080_0004
Formula (I-b) or a pharmaceutically acceptable salt thereof, wherein: each RcA is an independently selected Rc; n is 0 or 1 ; and
Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA. In certain embodiments of Formula (I-b), Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
In certain of these embodiments,
Figure imgf000081_0001
selected from the group consisting
Figure imgf000081_0002
, each further optionally substituted with RcA, wherein each RcA is an independently selected Rc.
In certain embodiments of Formula (I-b), Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
In certain of these embodiments,
Figure imgf000081_0003
selected from the group consisting of:
Figure imgf000081_0004
further optionally substituted with RcA, wherein each RcA is an independently selected Rc. In certain embodiments, the compound is a compound of Formula (I-c):
Figure imgf000082_0001
Formula (I-c) or a pharmaceutically acceptable salt thereof; wherein n is 0, 1, or 2; each RCA is an independently selected Rc; and
Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
In certain embodiments of Formula (I-c), Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
In certain of these embodiments,
Figure imgf000082_0002
selected from the group consisting
Figure imgf000082_0003
each further optionally substituted with RcA, wherein each RcA is an independently selected Rc.
Figure imgf000083_0001
which is further optionally substituted with from 1-2 RCA, wherein each RcA is an independently selected Rc.
Figure imgf000083_0002
In certain embodiments of Formula (I-c), Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA. In certain of these embodiments,
Figure imgf000084_0001
selected from the group consisting
Figure imgf000084_0002
optionally substituted with RcA, wherein each RcA is an independently selected Rc.
Figure imgf000084_0003
X1 Formula (I-d) or a pharmaceutically acceptable salt thereof; wherein n is 0 or 1 ; each RCA is an independently selected Rc; and Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA. In certain of these embodiments, Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA. In certain embodiments of Formula
Figure imgf000085_0001
selected from the group
Figure imgf000085_0002
each further optionally substituted with RCA, wherein each RcA is an independently selected Rc. In certain embodiments of Formula (I-d), Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA. In certain embodiments, the compound of Formula (I) is a compound of Formula
(I-e)
Figure imgf000085_0003
Formula (I-e), or a pharmaceutically acceptable salt thereof, wherein Ring D is heterocyclylene including from 3-10 ring atoms, wherein from 0-2 ring atoms (in addition to the ring nitrogen atom bonded to Rw) are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclylene is optionally substituted with from 1-3 substituents each independently selected from the group consisting of: oxo and -Rc. In certain embodiments of Formula (I-e), Rw is -Lw-W; and Lw is C(=O).
In certain embodiments of Formula (I-e), W is C2-6 alkenyl optionally substituted with from 1-3 Ra and further optionally substituted with Rg, wherein W is attached to Lw via an sp2 hybridized carbon atom. As a non-limiting example of the foregoing embodiments, W can be CH=CH2.
In certain embodiments of Formula (I-e), W is C2-6 alkenyl or C2-6 alkynyl optionally substituted with from 1-3 Ra and further optionally substituted with Rg, wherein W is attached to Lw via an sp2 or sp hybridized carbon atom. As non-limiting examples of
Figure imgf000086_0002
In certain embodiments of Formula (I-e), Ring
Figure imgf000086_0001
which is optionally substituted with from 1-2 Rc, wherein xl and x2 are each independently 0, 1, or 2.
In certain of these embodiments, xl is 0. As non-limiting examples of the foregoing embodiments, Ring D can be selected
Figure imgf000087_0001
As further non-limiting examples of the foregoing embodiments, Ring D can be
Figure imgf000087_0002
In certain embodiments of Formula
Figure imgf000087_0003
heterocyclylene bonded to Rw including from 3-10 ring atoms, wherein from 0-2 ring atoms (in addition to the ring nitrogen atom bonded to Rw) are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclylene is optionally substituted with from 1-3 substituents each independently selected from the group consisting of: oxo and -Rc; optionally wherein
Figure imgf000087_0004
is a monocyclic heterocyclylene ring including from 3-10 ring atoms as defined above with a nitrogen atom bonded to Rw (e.g.,
Figure imgf000087_0005
, such
Figure imgf000087_0006
, such as
Figure imgf000088_0001
heterocyclylene ring including from 3-10 ring atoms as defined above with a nitrogen atom
Figure imgf000088_0002
In certain embodiments of Formula (I-e), n is 0.
In certain embodiments of Formula (I-e), n is 1 or 2, such as wherein n is 1.
Figure imgf000088_0003
In certain embodiments of Formula (I-e), R7 is NReRf, such as NH2, NH(CI-3 alkyl), or N(CI-3 alkyl)2, such as wherein R7 is NH2. In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), each occurrence of RCA is independently selected from the group consisting of: halo; cyano; Ci-io alkyl which is optionally substituted with from 1-6 independently selected Ra; C1-4 alkoxy optionally substituted with C1-4 alkoxy or C1-4 haloalkoxy; C1-4 haloalkoxy; -S(O)i-2(Ci-4 alkyl); -NReRf; -OH; -S(O)I-2NR’R”; -C1-4 thioalkoxy; -C(=0)(Ci-io alkyl); -C(=O)O(Ci- 4 alkyl); -C(=O)OH; and -C(=O)NR’R”
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), one occurrence of RCA IS -NReRf
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), one occurrence ofRcA is -NH2.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), one occurrence of RCA is -NH(CI-6 alkyl), wherein the C1-6 alkyl is optionally substituted with from 1-3 substituents each independently selected from the group consisting of NR’R”, -OH, C1-6 alkoxy, C1-6 haloalkoxy, and halo. For example, one occurrence of RcA can be -NHMe, - NHCH2CF3, -NHCH2CH2OH, or -NHiPr.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), one occurrence of RCA is -NHC(=0)CI-4 alkyl, such as NHC(=0)CH3; or wherein one occurrence of RcA is N(CI-3 alkyl)2 such as NMei.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), one occurrence of RCA is C1-4 alkoxy optionally substituted with C1-4 alkoxy or C1-4 haloalkoxy. For example, one occurrence of RcA can be OMe or OCFbCFFOMe. As another non-limiting example, RcA can be C1-4 haloalkoxy, such as -OCH2CF3.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), one occurrence of RCA is C1-4 thioalkoxy (e.g., SCH3).
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), one occurrence of RCA is C1-6 alkyl, such as methyl; or wherein one occurrence of RcA is C1-6 alkyl substituted with from 1-6 independently selected halo (e.g., RcA can be -CF3).
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), one occurrence of RCA is C1-6 alkyl substituted with Ra, such as C1-6 alkyl substituted with C1-3 alkoxy or C(=O)NR’R”. For example, one occurrence of RcA can be
Figure imgf000090_0001
, or
Figure imgf000090_0002
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), one occurrence of RCA is halo (e.g., -F).
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), one occurrence ofRcA is -OH.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), one occurrence of RCA IS C(=O)NR’R”, such as C(=O)NHMe.
In Formula (I-a), (I-b), (I-c), (I-d) or (I-e), X1 can be as defined anywhere herein. In certain embodiments, X1 can be as defined in [AA1], [BB1], [CC1], [DD1], [EE1], or [FF1], infra.
[AA1J:
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), X1 is -(X2)m- L1-R5, wherein:
• m is 0 or 1;
• X2 is -N(RN)- or -O-;
• L1 is a bond or C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is - R"
In certain embodiments of [AA1], R5 is phenyl optionally substituted with from 1 - 4 Rc, such as wherein R5 is phenyl optionally substituted with from 1-2 independently selected halo, such as -F.
In certain embodiments of [AA1], R5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(Rd), and wherein the heteroaryl is optionally substituted with from 1 -4 Rc, such as wherein
Figure imgf000091_0001
In certain embodiments of [AA1], R5 is heteroaryl including 5 ring atoms, wherein from 1-4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc, such as wherein
Figure imgf000091_0002
Figure imgf000091_0003
In certain embodiments of [AA1], R5 is C3-10 cycloalkyl, such as C3-6 cycloalkyl, optionally substituted with from 1-4 Rc, such as wherein R5 is cyclopropyl.
In certain embodiments of [AA1], R5 is heterocyclyl including from 4-8, such as 4-
6, ring atoms, wherein from 1 -3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc. For example, R5 can
Figure imgf000091_0004
Figure imgf000091_0005
In certain embodiments of [ AA 11, in is 0.
In certain embodiments of [ AA 11, in is 1.
In certain embodiments of [AA1], X2 is -N(RN)- (e.g., N(H)).
In certain embodiments of [AA1], X2 is -O-. In certain embodiments of [AA1], L1 is a bond.
In certain embodiments of [AA1], L1 is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or - CH(Me)-).
In certain embodiments of [AA1], L1 is branched C3-6 alkylene. For example, L1 can be
Figure imgf000092_0001
wherein aa is the point of attachment to R5.
[BB11:
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), X1 is -X2-L1- R5, wherein:
• X2 is -N(RN)C(=O)-*, -N(RN)S(O)2-*, -N(RN)C(=O)O-*, or N(RN)C(=O)N(RN)*;
• L1 is a bond or C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is - R"
In certain embodiments of [BB1], R5 is phenyl optionally substituted with from 1 -
4 Rc, such as wherein R5 is phenyl optionally substituted with from 1-2 independently selected halo, such as -F.
In certain embodiments of [BB1], R5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(Rd), and wherein the heteroaryl is optionally substituted with from 1 -4 Rc, such as wherein
Figure imgf000092_0002
In certain embodiments of [BB1], R5 is heteroaryl including 5 ring atoms, wherein from 1-4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc, such as wherein
Figure imgf000093_0001
Figure imgf000093_0002
In certain embodiments of [BB1], R5 is C3-10 cycloalkyl, such as C3-6 cycloalkyl, optionally substituted with from 1-4 Rc, such as wherein R5 is cyclopropyl.
In certain embodiments of [BB1], R5 is heterocyclyl including from 4-8, such as 4-
6, ring atoms, wherein from 1 -3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc. For example, R5 can
Figure imgf000093_0003
Figure imgf000093_0004
In certain embodiments of [BB1], X2 is -N(RN)C(=O)-* (e.g., -N(H)C(=O)-*).
In certain embodiments of [BB1], X2 is -N(RN)S(O)2-, such as -N(H)S(O)2-*.
In certain embodiments of [BB1], X2 is -N(RN)C(=O)O-*, or -N(RN)C(=O)N(RN)-
* (e g., -N(H)C(=O)O-*; e g., -N(H)C(=O)N(H)-*).
In certain embodiments of [BB1], L1 is a bond.
In certain embodiments of [BB1], L1 is Ci-3 alkylene (e g., -CH2-, -CH2CH2-, or -
CH(Me)-).
In certain embodiments of [BB1], L1 is branched C3-6 alkylene. For example, L1 can be
Figure imgf000093_0005
wherein aa is the point of attachment to R5. [CC1]:
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), X1 is -X2-L1-
R5, wherein:
Figure imgf000094_0001
L1 is a bond or C1-6 alkylene optionally substituted with from 1-3 Ra; and
R5 is - R"
In certain embodiments of [CC1], R5 is phenyl optionally substituted with from 1 - 4 Rc, such as wherein R5 is phenyl optionally substituted with from 1-2 independently selected halo, such as -F.
In certain embodiments of [CC1], R5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(Rd), and wherein the heteroaryl is optionally substituted with
Figure imgf000094_0002
In certain embodiments of [CC1], R5 is heteroaryl including 5 ring atoms, wherein from 1-4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc, such as wherein
Figure imgf000094_0003
Figure imgf000094_0004
In certain embodiments of [CC1], R5 is C3-10 cycloalkyl, such as C3-6 cycloalkyl, optionally substituted with from 1-4 Rc, such as wherein R5 is cyclopropyl. In certain embodiments of [CC11, R5 is heterocyclyl including from 4-8, such as 4-
6, ring atoms, wherein from 1 -3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc. For example, R5 can
Figure imgf000095_0001
Figure imgf000095_0002
In some embodiments
Figure imgf000095_0003
In certain embodiments of [CC1], L1 is a bond.
In certain embodiments of [CC1], L1 is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or - CH(Me)-).
In certain embodiments of [CC1], L1 is branched C3-6 alkylene. For example, L1 can be
Figure imgf000095_0004
wherein aa is the point of attachment to R5.
[DD1]:
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), X1 is -(X2)m- L'-R5, wherein:
• m is 0 or 1;
• X2 is -N(RN)- or -O-;
• L1 is a bond or C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is R"2-Rv In certain embodiments of [DD11, the -Rg2 group present in R5 is 1,3 -phenylene or
1,4-phenylene, each optionally substituted with from 1-4 Rc, such as wherein -Rg2 is
Figure imgf000096_0001
wherein bb is the point of attachment to RY.
In certain embodiments of [DD1], the RY group present in R5 is -Rg.
In certain embodiments of [DD1], the RY group present in R5 is heterocyclyl including from 4-8, such as 4-6, ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc. For example, RY can be
Figure imgf000096_0002
In certain embodiments of [DD1], X2 is -N(RN)- (e.g., N(H)).
In certain embodiments of [DD1], X2 is -O-.
In certain embodiments of [DD1], L1 is a bond.
In certain embodiments of [DD1], L1 is Ci-3 alkylene (e g., -CH2-, -CH2CH2-, or -
CH(Me)-).
In certain embodiments of [DD1], L1 is branched C3-6 alkylene. For example, L1 can be
Figure imgf000096_0003
, wherein aa is the point of attachment to R5.
[EE1]:
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), X1 is X2-L'-
R5, wherein:
• X2 is -N(RN)-, -O-, -N(RN)C(=O)-*, -N(RN)S(O)2-, -N(RN)C(=O)O-*, or -N(RN)C(=O)N(RN)-*; • L1 is C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is H, halo, C1-6 alkoxy optionally substituted with from 1-3 Ra, or -OH.
In certain embodiments of [EE1], R5 is H.
In certain embodiments of [EE1], R5 is halo (e.g., -F).
In certain embodiments of [EE1], R5 is C1-6 alkoxy optionally substituted with from 1-3 Ra, such as wherein R5 is C1-3 alkoxy such as methoxy.
In certain embodiments of [EE1], R5 is -OH.
In certain embodiments of [EE1], X2 is -N(RN)- (e.g., N(H)).
In certain embodiments of [EE1], X2 is -O-.
In certain embodiments of [EE1], X2 is -N(RN)C(=O)-* (e.g., -N(H)C(=O)-*).
In certain embodiments of [EE1], X2 is -N(RN)S(O)2-, such as -N(H)S(O)2-*.
In certain embodiments of [EE1], X2 is -N(RN)C(=O)O-*, or -N(RN)C(=O)N(RN)- * (e.g., -N(H)C(=O)O-*; e g., -N(H)C(=O)N(H)-*).
In certain embodiments of [EE1], L1 is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or - CH(Me)-).
In certain embodiments of [EE1], L1 is branched C3-6 alkylene. For example, L1 can be
Figure imgf000097_0001
wherein aa is the point of attachment to R5.
[FF1]:
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), X1 is -L^R5, wherein L1 is C1-6 alkylene optionally substituted with from 1-3 Ra; and R5 is -L5-Rg.
In certain embodiments of [FF1], R5 is -O-Rg.
In certain embodiments of [FF1], R5 is -O-(phenyl), wherein the phenyl is optionally substituted with from 1-2 Rc. In certain embodiments of [FF1], L1 is C1-3 alkylene (e.g., -CH2-, -CH2CH2-, or - CH(Me)-).
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb is H.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R2a and R2b are both H.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R2a is a substituent that is other than H. In certain of these embodiments, R2a is C1-6 alkyl which is optionally substituted with from 1-6 Ra, such as wherein R2a is C1-3 alkyl, such as methyl or ethyl.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R2b is H.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3a and R3b are both H.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3a is a substituent that is other than H. In certain of these embodiments, R3a is C1-6 alkyl which is optionally substituted with from 1-6 Ra, such as wherein R3a is C1-3 alkyl, such as methyl or ethyl.
In certain of foregoing embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3a is C1-3 alkyl substituted with from 1-3 independently selected halo. As non-limiting examples of the foregoing embodiments, R3a is -CH2F, -CHF2, -CF3, -CH2CHF2, or - CH2CH2F.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3a is C1-3 alkyl substituted with C1-4 alkoxy, C1-4 haloalkoxy, or NReRf. Non-limiting examples of R3a in these embodiments include -CFFOMe, -CHoCHoOMe, -CH(Me)CH20Me, CH2CH(Me)OMe, -CH2OEt, -CH2CH2OCHF2, -CH2NReRf (e g., -CH2N(CF3)Me), or - CH2CH2NReRf (e g., -CH2CH2NMe2).
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3a is C1-3 alkyl substituted with C1-4 alkoxy, C1-4 haloalkoxy, or NReRf and further substituted with from 1-3 independently selected halo. In certain of these embodiments, R3a is C1-3 alkyl substituted with C1-4 alkoxy and further substituted with from 1-3 independently selected
'''O
V halo. Non-limiting examples of R3a in these embodiments include: -J- (e.g.,
Figure imgf000099_0001
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3a is C3-6 alkyl substituted with C1-4 alkoxy, C1-4 haloalkoxy, or NReRf. In certain of these embodiments, R3a is branched C3-6 alkyl substituted with C1-4 alkoxy, C1-4 haloalkoxy, or NReRf. In certain of the foregoing embodiments, R3a is branched C3-6 alkyl substituted with C1-4 alkoxy. For example, R3a can
Figure imgf000099_0002
In certain of the foregoing embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I- e), R3a is selected from the group consisting of: heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc; and
C3-6 cycloalkyl optionally substituted with from 1-4 Rc.
In certain of the foregoing embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I- e), R3a is — (C1-3 alkylene)-Rg or -(C1-3 alkylene)-O-Rg, and optionally the Rg group of R3a is:
C3-6 cycloalkyl optionally substituted with from 1-4 Rc, or heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3a is -CH2-Rg, or - CtFCFFR8, wherein Rg is 1,4-dioxanyl.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3a is-(Lg)g-Rw.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3a is-CH2CH2-Rw, wherein the Rw group is C(=0)-CH=CH2, or -NHC(=0)-CH=CH2.
As a non-limiting example of certain embodiments of Formula (I-a), (I-b), (I-c), (I-d)
Figure imgf000100_0001
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3a is -(Lg)g-
Rg2-Rw
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3a is -CH2-Rg2-
Rw, wherein the Rg2 group
Figure imgf000100_0003
such
Figure imgf000100_0002
Figure imgf000100_0004
wherein the waveline represents the point of attachment to -CH2- and the asterisk represents the point of attachment to Rw; and optionally the Rw group is C(=O)-
CH=CH2.
Figure imgf000101_0001
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3b is H.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3b is C1-3 alkyl. As non-limiting examples of the foregoing embodiments, R3b is methyl, ethyl, or propyl. For example, R3b is methyl.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3b is H.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3b is halo. For example, R3b can be -F.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3a and R3b, together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-8 ring atoms;
• wherein from 0-2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated ring of 4-8 ring atoms is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rw.
In certain of the foregoing embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I- e), R3a and R3b together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-6 ring atoms; • wherein from 1 -2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated ring of 4-6 ring atoms is optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc
In certain embodiments, R3a and R3b, together with the Ring B ring atom to which each is attached, form a fused C3-6 cycloalkyl, wherein the fused C3-6 cycloalkyl is optionally substituted with from 1-2 Rc.
As non-limiting examples of the foregoing embodiments, R3a and R3b, together with the Ring B ring atom to which each is attached, form
Figure imgf000102_0001
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3a and R3b, together with the Ring B ring atom to which each is attached, form:
Figure imgf000102_0002
, which is optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc, wherein: pl and p2 are independently 0, 1, or 2;
Rz is H, Rd, C(=0)-W, or S(O)2W; and cc represents the point of attachment to C(R2aR2b).
In certain embodiments of Formula (I-a), (I-b), (I-c together with the Ring B ring atom to which each is attached,
Figure imgf000102_0003
, wherein Rz is H, Rd, C(=O)-W, or S(O)iW; and cc represents the point of attachment to C(R2aR2b).
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R3a and R3b, together with the Ring B ring atom to which each is attached, form a fused ring selected
Figure imgf000103_0001
W; and cc represents the point of attachment to C(R2aR2b).
In certain embodiments, Rz is H. In certain embodiments, Rz is C1-6 alkyl optionally substituted with from 1-3 independently selected Ra. In certain embodiments, Rz is C(=O)-W or S(0)2W, optionally wherein W is C2-4 alkenyl.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb, R2a, and R2b are each H; and R3a and R3b taken together with the Ring B ring carbon atom to which each is attached form a fused C3-6 (such as C3 or C4) cycloalkyl, wherein the fused cycloalkyl ring is optionally substituted with from 1 -2 Rc. In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb, R2a, and R2b are each H; and R3a and R3b together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-6 ring atoms;
• wherein from 1 -2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated ring of 4-6 ring atoms is optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), one of R2a and R2b (such as R2a) and one of R3a and R3b (such as R3a) taken together with the Ring B ring atoms to which each is attached, form a fused saturated or unsaturated ring of 3-12 ring atoms;
• wherein from 0-2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated or unsaturated ring of 3-12 ring atoms is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
In certain of the foregoing embodiments, one of R2a and R2b (such as R2a) and one of R3a and R3b (such as R3a) taken together with the Ring B ring atoms to which each is attached, form a fused saturated ring of 3-8 ring atoms;
• wherein from 0-2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated ring of 3-8 ring atoms is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc
In certain of these foregoing embodiments, one of R2a and R2b (such as R2a) and one of R3a and R3b (such as R3a) taken together with the Ring B ring atoms to which each is attached, form a fused C3-6 cycloalkyl which is optionally substituted with from 1-2 Rc. As non-limiting examples of the foregoing embodiments, one of R2a and R2b (such as R2a) and one of R3a and R3b (such as R3a) taken together with the Ring B ring atoms to which each is attached, form a fused cyclopropyl or cyclobutyl ring, e.g.,
Figure imgf000105_0001
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb is H; and R2b is H; and R3b is -(Lg)g-Rg.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb is H; and R2b
Figure imgf000105_0002
In certain of these foregoing embodiments , Rla, Rlb is H, and R2b and R3b are each
H.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R2b and R3b are each H.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb, R2a, and R2b are each H, and R3a is C1-3 alkyl optionally substituted with from 1-3 Ra.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb, R2a, and R2b are each H; R3a, is C1-3 alkyl optionally substituted with from 1-3 Ra; and R3b is H, optionally each Ra substituent present in R3a is independently selected from the group consisting of: halo, C1-4 alkoxy, and C1-4 haloalkoxy. In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb, R2a, and R2b are each H; and R3a and R3b are independently selected C1-3 alkyl.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb is H; R2a and R3a taken together with the Ring B ring atoms to which each is attached, form a fused C3-6 (e.g., C3 or C4) cycloalkyl which is optionally substituted with from 1-2 Rc; and R2b and R3b are each H.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb, R2a, R2b, R3a, and R3b are each H.
In certain of these foregoing emodiments, R3b is H, and each optionally present Ra substituent in R3a is independently selected from the group consisting of: halo, C1-4 alkoxy, and Ci -4 haloalkoxy.
In certain of emodiments, R3b is -F, and each optionally present Ra substituent in R3a is independently selected from the group consisting of: halo, C1-4 alkoxy, and CM haloalkoxy.
In certain of emodiments, R3b is C1-3 alkyl (e.g., methyl), and each optionally present Ra substituent in R3a is independently selected from the group consisting of: halo, Ci -4 alkoxy, and C1-4 haloalkoxy.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb, R2a, and R2b are each H; R3a, is -Rg, -(C1-3 alkylene)-Rg, or -(C1-3 alkylene)-O-Rg, optionally wherein the Rg group of R3a is:
C3-6 cycloalkyl optionally substituted with from 1-4 Rc, or heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc; and
R3b is H. In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb, R2a, and R2b are each H; and R3a is C1-3 alkyl optionally substituted with from 1-3 Ra; and R3b is H, optionally each Ra substituent present in R3a is independently selected from the group consisting of: halo, C1-4 alkoxy, and C1-4 haloalkoxy.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb, R2a, and R2b are each H and R3a, is -Rg, -(C1-3 alkylene)-Rg, or -(C1-3 alkylene)-O-Rg, optionally wherein the Rg group of R3a is:
C3-6 cycloalkyl optionally substituted with from 1-4 Rc, or heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc; and
R3b is H.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb, R2a, and R2b are each H, and R3a and R3b taken together with the Ring B ring carbon atom to which each is attached form a fused C3-6 (such as C3 or C4) cycloalkyl, wherein the fused cycloalkyl ring is optionally substituted with from 1 -2 Rc.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb, R2a, and R2b are each H, and R3a and R3b together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-6 ring atoms;
• wherein from 1 -2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated ring of 4-6 ring atoms is optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb is H, and R2a and R3a taken together with the Ring B ring atoms to which each is attached, form a fused C3-6 (e.g., C3 or C4) cycloalkyl which is optionally substituted with from 1-2 Rc, and R2b and R3b are each H.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Rla, Rlb, R2a, R2b, R3a, and R3b are each H.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), R4 is H.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Ring A is
(RcB)mi
Figure imgf000108_0001
, wherein each RcB is an independently selected Rc; and ml is 0, 1, 2, 3, or 4.
In certain of these embodiments, ml is 1, 2, or 3, such as 1 or 2.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Ring A is
Figure imgf000108_0002
wherein each RcB is an independently selected Rc.
As non-limiting example of certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or
(I-e), Ring A can
Figure imgf000108_0003
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Ring A is selected from the group consisting
Figure imgf000108_0004
Figure imgf000109_0001
wherein each RcB is an independently selected Rc.
In certain embodiments, each RcB is independently selected from the group consisting of: -halo, such as -Cl and -F; -CN; C1-4 alkoxy; C1-4 haloalkoxy; C1-3 alkyl; and C1-3 alkyl substituted with from 1-6 independently selected halo.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), Ring A is bicyclic heteroaryl including from 9-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 substituents independently selected from the group consisting of Rc and oxo, such as wherein: Ring A
Figure imgf000109_0002
Figure imgf000109_0003
each of which is further optionally substituted with Rc.
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), the
Figure imgf000109_0004
In certain embodiments of Formula (I-a), (I-b), (I-c), (I-d) or (I-e), the
Figure imgf000110_0001
Non-Limiting Exemplary Compounds In certain embodiments, the compound is selected from the group consisting of the compounds delineated in Table Cl, or a pharmaceutically acceptable salt thereof.
Table Cl
For certain compounds, the symbol * at a chiral center denotes that this chiral center has been resolved (i.e., is a single epimer) and the absolute stereochemistry at that center has not been determined.
Figure imgf000110_0002
Figure imgf000111_0001
Figure imgf000112_0001
ʼnll
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Pharmaceutical Compositions and Administration
General
In some embodiments, a chemical entity (e.g., a compound that inhibits EGFR and/or HER2, or a pharmaceutically acceptable salt, and/or hydrate, and/or cocrystal, and/or drug combination thereof) is administered as a pharmaceutical composition that includes the chemical entity and one or more pharmaceutically acceptable excipients, and optionally one or more additional therapeutic agents as described herein.
In some embodiments, the chemical entities can be administered in combination with one or more conventional pharmaceutical excipients. Pharmaceutically acceptable excipients include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-a-tocopherol polyethylene glycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens, poloxamers or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, tris, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium-chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, and wool fat. Cyclodextrins such as a-, 0, and y-cyclodextrin, or chemically modified derivatives such as hydroxyalkyl cyclodextrins, including 2- and 3- hydroxypropyl-0-cyclodextrins, or other solubilized derivatives can also be used to enhance delivery of compounds described herein. Dosage forms or compositions containing a chemical entity as described herein in the range of 0.005% to 100% with the balance made up from non-toxic excipient may be prepared. The contemplated compositions may contain 0.001%- 100% of a chemical entity provided herein, in one embodiment 0.1-95%, in another embodiment 75-85%, in a further embodiment 20-80%. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 22nd Edition (Pharmaceutical Press, London, UK. 2012).
Routes of Administration and Composition Components
In some embodiments, the chemical entities described herein or a pharmaceutical composition thereof can be administered to subject in need thereof by any accepted route of administration. Acceptable routes of administration include, but are not limited to, buccal, cutaneous, endocervical, endosinusial, endotracheal, enteral, epidural, interstitial, intra-abdominal, intra-arterial, intrabronchial, intrabursal, intracerebral, intracisternal, intracoronary, intradermal, intraductal, intraduodenal, intradural, intraepidermal, intraesophageal, intragastric, intragingival, intraileal, intralymphatic, intramedullary, intrameningeal, intramuscular, intraovarian, intraperitoneal, intraprostatic, intrapulmonary, intrasinal, intraspinal, intrasynovial, intratesticular, intrathecal, intratubular, intratumoral, intrauterine, intravascular, intravenous, nasal, nasogastric, oral, parenteral, percutaneous, peridural, rectal, respiratory (inhalation), subcutaneous, sublingual, submucosal, topical, transdermal, transmucosal, transtracheal, ureteral, urethral and vaginal. In certain embodiments, a preferred route of administration is parenteral (e.g., intratumoral).
Compositions can be formulated for parenteral administration, e.g., formulated for injection via the intravenous, intramuscular, sub-cutaneous, or even intraperitoneal routes. Typically, such compositions can be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for use to prepare solutions or suspensions upon the addition of a liquid prior to injection can also be prepared; and the preparations can also be emulsified. The preparation of such formulations will be known to those of skill in the art in light of the present disclosure.
The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil, or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that it may be easily injected. It also should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
The carrier also can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques, which yield a powder of the active ingredient, plus any additional desired ingredient from a previously sterile-filtered solution thereof.
Intratumoral injections are discussed, e.g., in Lammers, et al., “Effect of Intratumoral Injection on the Biodistribution and the Therapeutic Potential of HPMA Copolymer-Based Drug Delivery Systems” Neoplasia. 2006, 10, 788-795.
Pharmacologically acceptable excipients usable in the rectal composition as a gel, cream, enema, or rectal suppository, include, without limitation, any one or more of cocoa butter glycerides, synthetic polymers such as polyvinylpyrrolidone, PEG (like PEG ointments), glycerine, glycerinated gelatin, hydrogenated vegetable oils, poloxamers, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol Vaseline, anhydrous lanolin, shark liver oil, sodium saccharinate, menthol, sweet almond oil, sorbitol, sodium benzoate, anoxid SBN, vanilla essential oil, aerosol, parabens in phenoxyethanol, sodium methyl p-oxybenzoate, sodium propyl p- oxybenzoate, diethylamine, carbomers, carbopol, methyloxybenzoate, macrogol cetostearyl ether, cocoyl caprylocaprate, isopropyl alcohol, propylene glycol, liquid paraffin, xanthan gum, carboxy-metabisulfite, sodium edetate, sodium benzoate, potassium metabisulfite, grapefruit seed extract, methyl sulfonyl methane (MSM) , lactic acid, glycine, vitamins, such as vitamin A and E and potassium acetate. In certain embodiments, suppositories can be prepared by mixing the chemical entities described herein with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum and release the active compound. In other embodiments, compositions for rectal administration are in the form of an enema.
In other embodiments, the compounds described herein or a pharmaceutical composition thereof are suitable for local delivery to the digestive or GI tract by way of oral administration (e.g., solid or liquid dosage forms.).
Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the chemical entity is mixed with one or more pharmaceutically acceptable excipients, such as sodium citrate or dicalcium phosphate and/or: a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
In one embodiment, the compositions will take the form of a unit dosage form such as a pill or tablet and thus the composition may contain, along with a chemical entity provided herein, a diluent such as lactose, sucrose, dicalcium phosphate, or the like; a lubricant such as magnesium stearate or the like; and a binder such as starch, gum acacia, polyvinylpyrrolidine, gelatin, cellulose, cellulose derivatives or the like. In another solid dosage form, a powder, marume, solution or suspension (e.g., in propylene carbonate, vegetable oils, PEG’s, poloxamer 124 or triglycerides) is encapsulated in a capsule (gelatin or cellulose base capsule). Unit dosage forms in which one or more chemical entities provided herein or additional active agents are physically separated are also contemplated; e.g., capsules with granules (or tablets in a capsule) of each drug; two-layer tablets; two- compartment gel caps, etc. Enteric coated or delayed release oral dosage forms are also contemplated.
Other physiologically acceptable compounds include wetting agents, emulsifying agents, dispersing agents or preservatives that are particularly useful for preventing the growth or action of microorganisms. Various preservatives are well known and include, for example, phenol and ascorbic acid.
In certain embodiments the excipients are sterile and generally free of undesirable matter. These compositions can be sterilized by conventional, well-known sterilization techniques. For various oral dosage form excipients such as tablets and capsules sterility is not required. The USP/NF standard is usually sufficient.
In certain embodiments, solid oral dosage forms can further include one or more components that chemically and/or structurally predispose the composition for delivery of the chemical entity to the stomach or the lower GI; e.g., the ascending colon and/or transverse colon and/or distal colon and/or small bowel. Exemplary formulation techniques are described in, e.g., Filipski, K.J., et al., Current Topics in Medicinal Chemistry, 2013, 13, 776-802, which is incorporated herein by reference in its entirety.
Examples include upper-GI targeting techniques, e.g., Accordion Pill (Intec Pharma), floating capsules, and materials capable of adhering to mucosal walls.
Other examples include lower-GI targeting techniques. For targeting various regions in the intestinal tract, several enteric/pH-responsive coatings and excipients are available. These materials are typically polymers that are designed to dissolve or erode at specific pH ranges, selected based upon the GI region of desired drug release. These materials also function to protect acid labile drugs from gastric fluid or limit exposure in cases where the active ingredient may be irritating to the upper GI (e.g., hydroxypropyl methylcellulose phthalate series, Coateric (polyvinyl acetate phthalate), cellulose acetate phthalate, hydroxypropyl methylcellulose acetate succinate, Eudragit series (methacrylic acid-methyl methacrylate copolymers), and Marcoat). Other techniques include dosage forms that respond to local flora in the GI tract, Pressure-controlled colon delivery capsule, and Pulsincap.
Ocular compositions can include, without limitation, one or more of any of the following: viscogens (e.g., Carboxymethylcellulose, Glycerin, Polyvinylpyrrolidone, Polyethylene glycol); Stabilizers (e.g., Pluronic (triblock copolymers), Cyclodextrins); Preservatives (e.g., Benzalkonium chloride, ETDA, SofZia (boric acid, propylene glycol, sorbitol, and zinc chloride; Alcon Laboratories, Inc.), Purite (stabilized oxy chloro complex; Allergan, Inc.)).
Topical compositions can include ointments and creams. Ointments are semisolid preparations that are typically based on petrolatum or other petroleum derivatives. Creams containing the selected active agent are typically viscous liquid or semisolid emulsions, often either oil-in-water or water-in-oil. Cream bases are typically water-washable, and contain an oil phase, an emulsifier and an aqueous phase. The oil phase, also sometimes called the “internal” phase, is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol; the aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant. The emulsifier in a cream formulation is generally a nonionic, anionic, cationic or amphoteric surfactant. As with other carriers or vehicles, an ointment base should be inert, stable, nonirritating and nonsensitizing.
In any of the foregoing embodiments, pharmaceutical compositions described herein can include one or more one or more of the following: lipids, interbilayer crosslinked multilamellar vesicles, biodegradeable poly(D,L-lactic-co-glycolic acid) [PLGA]-based or poly anhydride-based nanoparticles or microparticles, and nanoporous particle-supported lipid bilayers.
Dosages
The dosages may be varied depending on the requirement of the patient, the severity of the condition being treating and the particular compound being employed. Determination of the proper dosage for a particular situation can be determined by one skilled in the medical arts. The total daily dosage may be divided and administered in portions throughout the day or by means providing continuous delivery. In some embodiments, the compounds described herein are administered at a dosage of from about 0.001 mg/Kg to about 500 mg/Kg (e.g., from about 0.001 mg/Kg to about 200 mg/Kg; from about 0.01 mg/Kg to about 200 mg/Kg; from about 0.01 mg/Kg to about 150 mg/Kg; from about 0.01 mg/Kg to about 100 mg/Kg; from about 0.01 mg/Kg to about 50 mg/Kg; from about 0.01 mg/Kg to about 10 mg/Kg; from about 0.01 mg/Kg to about 5 mg/Kg; from about 0.01 mg/Kg to about 1 mg/Kg; from about 0.01 mg/Kg to about 0.5 mg/Kg; from about 0.01 mg/Kg to about 0.1 mg/Kg; from about 0. 1 mg/Kg to about 200 mg/Kg; from about 0. 1 mg/Kg to about 150 mg/Kg; from about 0. 1 mg/Kg to about 100 mg/Kg; from about 0.1 mg/Kg to about 50 mg/Kg; from about 0. 1 mg/Kg to about 10 mg/Kg; from about 0. 1 mg/Kg to about 5 mg/Kg; from about 0. 1 mg/Kg to about 1 mg/Kg; from about 0. 1 mg/Kg to about 0.5 mg/Kg).
Regimens
The foregoing dosages can be administered on a daily basis (e.g., as a single dose or as two or more divided doses) or non-daily basis (e.g., every other day, every two days, every three days, once weekly, twice weeks, once every two weeks, once a month).
In some embodiments, the period of administration of a compound described herein is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 1 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 1 1 months, 12 months, or more. In a further embodiment, a period of during which administration is stopped is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 1 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 1 1 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 1 1 months, 12 months, or more. In an embodiment, a therapeutic compound is administered to an individual for a period of time followed by a separate period of time. In another embodiment, a therapeutic compound is administered for a first period and a second period following the first period, with administration stopped during the second period, followed by a third period where administration of the therapeutic compound is started and then a fourth period following the third period where administration is stopped. In an aspect of this embodiment, the period of administration of a therapeutic compound followed by a period where administration is stopped is repeated for a determined or undetermined period of time. In a further embodiment, a period of administration is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more. In a further embodiment, a period of during which administration is stopped is for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, or more.
Methods of Treatment
Indications
Provided herein are methods for inhibiting epidermal growth factor receptor tyrosine kinase (EGFR) and/or human epidermal growth factor receptor 2 (HER2). For example, provided herein are inhibitors of EGFR useful for treating or preventing diseases or disorders associated with dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same (i.e., an EGFR-associated disease or disorder), such as a central nervous system diseases, a pulmonary disorder, cardiovascular disease, ischemia, liver disease, a gastrointestinal disorder, a viral or bacterial infection, an inflammatory and/or autoimmune disease, or cancer (e.g., EGFR-associated cancer). In some embodiments, provided herein are inhibitors of HER2 useful for treating or preventing diseases or disorders associated with dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, such as cancer (e.g., HER2- associated cancer). In some embodiments, provided herein are inhibitors of EGFR and HER2.
An “EGFR inhibitor” as used herein includes any compound exhibiting EGFR inactivation activity (e.g., inhibiting or decreasing). In some embodiments, an EGFR inhibitor can be selective for an EGFR kinase having one or more mutations. For example, an EGFR inhibitor can bind to the adenosine triphosphate (ATP)-binding site in the tyrosine kinase domain. In some embodiments, an EGFR inhibitor is an allosteric inhibitor.
The compounds provided herein can inhibit EGFR. In some embodiments, the compounds can bind to the EGFR adenosine triphosphate (ATP)-binding site in the tyrosine kinase domain.
The ability of test compounds to act as inhibitors of EGFR may be demonstrated by assays known in the art. The activity of the compounds and compositions provided herein as EGFR inhibitors can be assayed in vitro, in vivo, or in a cell line. In vitro assays include assays that determine inhibition of the kinase and/or ATPase activity. Alternate in vitro assays quantitate the ability of the inhibitor to bind to the protein kinase and can be measured either by radio labelling the compound prior to binding, isolating the compound/kinase complex and determining the amount of radio label bound, or by running a competition experiment where new compounds are incubated with the kinase bound to known radioligands. In some cases, an EGFR inhibitor can be evaluated by its effect on the initial velocity of EGFR tyrosine kinase catalyzed peptide phosphorylation (e.g., Yun et al. Cancer Cell. 2007;l l(3):217-227). In some embodiments, the binding constant of an EGFR inhibitor can be determined using fluorescence kinetics (e.g., Yun et al. Cancer Cell. 2007;l l(3):217-227). Examples of surface plasmon resonance (SPR) binding assays include those disclosed in Li, Shiqing, et al. Cancer cell 7.4 (2005): 301-311. Additional EGFR inhibitor assays can be found, for example, in WO 2019/246541 and WO 2019/165358 both of which are incorporated by reference in their entireties).
Assays can include, for example, proliferation inhibition assays such as those that measure cell growth inhibition, such as an MTS assay or by Cell Titer Gio Luminescent Cell viability assay (Promega®). To perform such an assay, cells are seeded and grown in cell culture plates before being exposed to a test compound for varying durations. Assessment of the viability of the cells following this exposure is then performed. Data are normalized with respect to untreated cells and can be displayed graphically. Growth curves can be fitted using a nonlinear regression model with sigmoidal dose response. As another example, a Western Blot analysis can be used. In such assays cells are seeded and grown in culture plates and then treated with a test compound the following day for varying durations. Cells are washed with PBS and lysed. SDS-PAGE gels are used to separate the lysates which are transferred to nitrocellulose membranes, and probed with appropriate antibodies (e.g., phospho-EGFR(Tyrl 068)(3777), total EGFR (2232), p-Akt(Ser473) (4060), total Akt (9272), p-ERK(Thr202/Tyr204)(4370), total ERK (9102), and HSP90 (SC-7947)).
Additional assays can include, for example, assays based on ALPHALISA TECHNOLOGY® (e g., see the ALPHALISA® EGF/EGFR binding kit from Promega). Such assays use a luminescent oxygen-channeling chemistry to detect molecules of interest in, for example, buffer, cell culture media, serum, and plasma. For example, a biotinylated EGF is bound to streptavidin-coated Alpha donor beads, and EGFR-Fc is captured by antihuman IgG Fc-specific AlphaLISA acceptor beads. When EGF is bound to EGFR, donor beads and acceptor beads come into close proximity, and the excitation of the donor beads provokes the release of singlet oxygen molecules that triggers a cascade of energy transfers in the acceptor beads. This results in a sharp peak of light emission at 615 nm. Such assays can be used, for example, in competitive binding experiments.
Further examples of assays can include assays based on Sox technology (e.g., see the PHOSPHOSENS® Sox-based Homogeneous, Kinetic or Endpoint/Red Fluorescencebased Assays from ASSAYQUANT®). Such assays utilize chelation-enhanced fluorescence (CHEF) using a sulfonamido-oxine (Sox) chromophore in peptide or protein substrates to create real-time sensors of phosphorylation. See, e.g., U.S. Patent Nos. 8,586,570 and 6,906,194.
Potency of an EGFR inhibitor as provided herein can be determined by ECso value. A compound with a lower ECso value, as determined under substantially similar conditions, is a more potent inhibitor relative to a compound with a higher ECso value. In some embodiments, the substantially similar conditions comprise determining an EGFR- dependent phosphorylation level, in vitro or in vivo (e.g., in tumor cells, A431 cells, Ba/F3 cells, or 3T3 cells cells expressing a wild type EGFR, a mutant EGFR, or a fragment of any thereof).
Potency of an EGFR inhibitor as provided herein can also be determined by IC50 value. A compound with a lower IC50 value, as determined under substantially similar conditions, is a more potent inhibitor relative to a compound with a higher IC50 value. In some embodiments, the substantially similar conditions comprise determining an EGFR- dependent phosphorylation level, in vitro or in vivo (e.g., in tumor cells, A431 cells, Ba/F3 cells, or 3T3 cells expressing a wild type EGFR, a mutant EGFR, or a fragment of any thereof).
The selectivity between wild type EGFR and EGFR containing one or more mutations as described herein can also be measured using cellular proliferation assays where cell proliferation is dependent on kinase activity. For example, murine Ba/F3 cells transfected with a suitable version of wild type EGFR (such as VIII; containing a wild type EGFR kinase domain), or Ba/F3 cells transfected with L858R/T790M, Del/T790M/L718Q, L858R/T790M/L718Q, L858R/T790M/C797S, Del/T790M/C797S, L858R/T790M/I941R, exon 19 deletion/T790M, or an exon 20 insertion such as V769_D770insX, D770_N771insX, N771_P772insX, P772_H773insX, or
H773_V774insX (e.g., A767_V769dupASV, V769_D770insASV, D770_N771insNPG, D770_N771insNPY, D770_N771insSVD, D770_N771insGL, N771_H773dupNPH, N771_P772insN, N771_P772insH, N771_P772insV, P772_H773insDNP,
P772_H773insPNP, H773_V774insNPH, H773_V774insH, H773_V774insPH,
H773_V774insAH, or P772_H773insPNP) can be used. Proliferation assays are performed at a range of inhibitor concentrations (e.g., 10 pM, 3 pM, 1.1 pM, 330 nM, 110 nM, 33 nM, 11 nM, 3 nM, 1 nM) and an ECso is calculated.
An alternative method to measure effects on EGFR activity is to assay EGFR phosphorylation. Wildtype or mutant (L858R/T790M, Del/T790M, Del/T790M/L718Q, L858R/T790M/C797S, Del/T790M/C797S, L858R/T790M/I941R, or
L858R/T790M/L718Q) EGFR can be transfected into cells which do not normally express endogenous EGFR and the ability of the inhibitor (e.g., using concentrations as above) to inhibit EGFR phosphorylation can be assayed. Cells are exposed to increasing concentrations of inhibitor and stimulated with EGF. The effects on EGFR phosphorylation are assayed by Western Blotting using phospho-specific EGFR antibodies.
In some embodiments, the compounds provided herein can exhibit potent and selective inhibition of EGFR. For example, the compounds provided herein can bind to the EGFR adenosine triphosphate (ATP)-binding site in the tyrosine kinase domain. In some embodiments, the compounds provided herein can exhibit nanomolar potency against an EGFR kinase including an activating mutation or an EGFR inhibitor resistance mutation, including, for example, the resistance mutations in Table 2a and 2b (e.g., L747S, D761Y, T790M, and T854A), with minimal activity against related kinases (e.g., wild type EGFR). Inhibition of wild type EGFR can cause undesireable side effects (e.g., diarrhea and skin rashes) that can impact quality of life and compliance. In some cases, the inhibititon of wild type EGFR can lead to dose limiting toxicities. See, e.g., Morphy. J. Med. Chem. 2010, 53, 4, 1413-1437 and Peters. J. Med. Chem. 2013, 56, 22, 8955-8971.
In some embodiments, the compounds of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, can selectively target an EGFR kinase. For example, a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, can selectively target an EGFR kinase over another kinase or non-kinase target.
In some embodiments, a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I- c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, can exhibit greater inhibition of EGFR containing one or more mutations as described herein (e.g., one or more mutations as described in Table la and lb) relative to inhibition of wild type EGFR. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit at least 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or 100-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit up to 1000-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit up to 10000-fold greater inhibition of EGFR having a combination of mutations described herein relative to inhibition of wild type EGFR.
In some embodiments, a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I- c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, can exhibit from about 2- fold to about 10-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit from about 10-fold to about 100-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit from about 1 OO-fold to about 1 OOO-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit from about 1 OOO-fold to about 1 OOOO-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR.
In other embodiments, a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit greater inhibition of EGFR containing one or more mutations as described herein (e.g., one or more mutations as described in Table la and lb) relative to inhibition of wild type EGFR. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit at least 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or 100-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit up to 1000-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit up to 1 OOOO-fold greater inhibition of EGFR having a combination of mutations described herein relative to inhibition of wild type EGFR.
In other embodiments, a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit from about 2-fold to about 10-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit from about 10-fold to about 100-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit from about 100-fold to about 1000-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit from about 1000-fold to about lOOOO-fold greater inhibition of EGFR containing one or more mutations as described herein relative to inhibition of wild type EGFR.
Compounds of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or pharmaceutically acceptable salts or solvates thereof, are useful for treating diseases and disorders which can be treated with an EGFR inhibitor, such as EGFR-associated diseases and disorders, e.g., central nervous system diseases (e.g., neurodegenerative diseases), pulmonary disorders, cardiovascular disease, ischemia, liver disease, gastrointestinal disorders, viral or bacterial infections, inflammatory and/or autoimmune diseases (e.g., psoriasis and atopic dermatitis), and proliferative disorders such as cancers, including hematological cancers and solid tumors (e.g., advanced solid tumors).
A “HER2 inhibitor” as used herein includes any compound exhibiting HER2 inactivation activity (e.g., inhibiting or decreasing). In some embodiments, a HER2 inhibitor can be selective for a HER2 kinase having one or more mutations. In some embodiments, a HER2 inhibitor can bind to the HER2 adenosine triphosphate (ATP)- binding site in the tyrosine kinase domain.
The compounds provided herein can inhibit HER2. For example, the compounds can bind to the HER2 adenosine triphosphate (ATP)-binding site in the tyrosine kinase domain. In some embodiments, the compounds provided herein can inhibit wild type HER2. In some embodiments, the compounds provided herein can inhibit HER2 having one or more mutations as described herein.
The ability of test compounds to act as inhibitors of HER2 may be demonstrated by assays known in the art. The activity of the compounds or compositions provided herein as HER2 inhibitors can be assayed in vitro, in vivo, or in a cell line. In vitro assays include assays that determine inhibition of the kinase and/or ATPase activity. Alternate in vitro assays quantitate the ability of the inhibitor to bind to the protein kinase and can be measured either by radio labelling the compound prior to binding, isolating the compound/kinase complex and determining the amount of radio label bound, or by running a competition experiment where new compounds are incubated with the kinase bound to known radioligands. In some cases, a HER2 inhibitor can be evaluated by its effect on the initial velocity of HER2 tyrosine kinase catalyzed peptide phosphorylation (e.g., Yun et al. Cancer Cell. 2007;l l(3):217-227). For example, an assay that indirectly measures ADP formed from the HER2 kinase reaction can be used (see, e.g., ATP/NADH coupled assay systems and luminescent kinase assays such as ADP-GLO™ Kinase Assay from Promega). See, e.g., Hanker et al. Cancer Discov. 2017 Jun;7(6): 575-585; Robichaux et al. Nat Med. 2018 May; 24(5): 638-646; and Yun et al. Proc Natl Acad Sci U S A. 2008 Feb 12;105(6):2070-5. In some embodiments, an assay that detects substrate phosphorylation using a labeled anti-phospho-tyrosine antibody can be used (see, e.g., Rabindran et al. Cancer Res. 2004 Jun 1 ;64(11):3958-65). In some embodiments, the binding constant of a HER2 inhibitor can be determined using fluorescence kinetics (e.g., Yun et al. Cancer Cell. 2007;l l(3):217-227). Examples of SPR binding assays include those disclosed in Li, Shiqing, et al. Cancer cell 7.4 (2005): 301 -311. In some embodiments, covalent binding of a HER2 inhibitor to HER2 can be detected using mass spectrometry, see, e.g., Irie et al. Mol Cancer Ther. 2019 Apr;18(4):733-742. Additional HER2 inhibitor assays can be found, for example, in U.S. Patent No. 9,920,060, WO 2019/241715, and U.S. Publication No. 2017/0166598, each of which are incorporated by reference in their entireties.
Potency of a HER2 inhibitor as provided herein can be determined by ECso value. A compound with a lower ECso value, as determined under substantially similar conditions, is a more potent inhibitor relative to a compound with a higher ECso value. In some embodiments, the substantially similar conditions comprise determining an HER2- dependent phosphorylation level, in vitro or in vivo (e.g., in tumor cells or Ba/F3 cells expressing a wild type HER2, a mutant HER2, or a fragment of any thereof).
Potency of an HER2 inhibitor as provided herein can also be determined by IC50 value. A compound with a lower IC50 value, as determined under substantially similar conditions, is a more potent inhibitor relative to a compound with a higher IC50 value. In some embodiments, the substantially similar conditions comprise determining an HER2- dependent phosphorylation level, in vitro or in vivo (e.g., in tumor cells or Ba/F3 cells expressing a wild type HER2, a mutant HER2, or a fragment of any thereof). Assays can include, for example, proliferation inhibition assays such as those that measure cell growth inhibition, such as an MTS assay or by Cell Titer Gio Luminescent Cell viability assay (Promega®). To perform such an assay, cells are seeded and grown in cell culture plates before being exposed to a test compound for varying durations. Assessment of the viability of the cells following this exposure is then performed. Data are normalized with respect to untreated cells and can be displayed graphically. Growth curves can be fitted using a nonlinear regression model with sigmoidal dose response. As another example, a Western Blot analysis can be used. In such assays cells are seeded and grown in culture plates and then treated with a test compound the following day for varying durations. Cells are washed with PBS and lysed. SDS-PAGE gels are used to separate the lysates which are transferred to nitrocellulose membranes, and probed with appropriate antibodies (e.g., phospho-HER2(Tyrl248)(2247), phospho-EGFR-Tyrl 173 phospho- HER2-Tyr877, phospho-HER2-Tyrl221, total HER2, phospho-AKT-Thr308, phospho- AKT-Ser374, total AKT, phospho-p44/42 MAPK-Thr202/Tyr204, and p44/42 MAPK).
The selectivity between wild type HER2 and HER2 containing one or more mutations as described herein can also be measured using cellular proliferation assays where cell proliferation is dependent on kinase activity. For example, murine Ba/F3 cells transfected with a suitable version of wild type HER2, or Ba/F3 cells transfected with HER2 having one or more mutations such as S310F, S310Y, R678Q, R678W, R678P, I767M, V773M, V777L, V842I, M774AYVM, M774del insWLV, A775_G776insYVMA, A775_G776insAVMA, A775_G776insSVMA, A775_G776insVAG, A775insV G776C, A775_G776insI, G776del insVC2, G776del insW, G776del insLC, G776C V777insC, G776C V777insV, V777_G778insCG, G778_S779insCPG, or P780 Y781 insGSP can be used. Proliferation assays are performed at a range of inhibitor concentrations (e.g., 10 pM, 3 pM, 1.1 pM, 330 nM, 110 nM, 33 nM, 11 nM, 3 nM, 1 nM) and an ECso is calculated.
An alternative method to measure effects on HER2 activity is to assay HER2 phosphorylation. Wildtype or mutant (S310F, S310Y, R678Q, R678W, R678P, I767M, V773M, V777L, V842I, M774AYVM, M774del insWLV, A775_G776insYVMA, A775_G776insAVMA, A775_G776insSVMA, A775_G776insVAG, A775insV G776C, A775_G776insI, G776del insVC2, G776del insW, G776del insLC, G776C V777insC, G776C V777insV, V777_G778insCG, G778_S779insCPG, or P780 Y781 insGSP) HER2 can be transfected into cells which do not normally express endogenous HER2 and the ability of the inhibitor (e.g., using concentrations as above) to inhibit HER2 phosphorylation can be assayed. Cells are exposed to increasing concentrations of inhibitor and stimulated with EGF. The effects on HER2 phosphorylation are assayed by Western Blotting using phospho-specific HER2 antibodies.
In some embodiments, the compounds provided herein can exhibit potent and selective inhibition of HER2. For example, the compounds provided herein can bind to the HER2 adenosine triphosphate (ATP)-binding site in the tyrosine kinase domain. In some embodiments, the compounds provided herein can exhibit nanomolar potency against a HER2 kinase including an activating mutation or a HER2 inhibitor resistance mutation, including, for example, exon 20 insertions and/or the resistance mutations in Table 5 (e.g., L755S, L755P, T798I, and T798M), with minimal activity against related kinases (e.g., wild type EGFR).
In some embodiments, the compounds of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, can selectively target a HER2 kinase. For example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can selectively target a HER2 kinase over another kinase (e.g., wild type EGFR) or non-kinase target. It can be desireable to selectively target a HER2 kinase over a wild type EGFR kinase due to undesireable side effects (e.g., diarrhea and skin rashes) that can impact quality of life and compliance. See, e.g., Morphy. J. Med. Chem. 2010, 53, 4, 1413-1437 and Peters. J. Med. Chem. 2013, 56, 22, 8955-8971.
In some embodiments, a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I- c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, can exhibit greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein (e.g., one or more mutations as described in Table 3) relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit at least 2-fold, 3- fold, 5-fold, 10-fold, 25-fold, 50-fold or 100-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit up to 1000-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or nonkinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit up to lOOOO-fold greater inhibition of wild type HER2 or HER2 having a combination of mutations described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
In some embodiments, a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I- c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, can exhibit from about 2- fold to about 10-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit from about 10-fold to about 100-fold greater inhibition of wild type HER2 or containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit from about 100-fold to about 1000-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit from about 1000-fold to about 1 OOOO-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
In other embodiments, a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR inhibitor can exhibit greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein (e.g., one or more mutations as described in Table 3) relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second HER2 inhibitor can exhibit at least 2-fold, 3 -fold, 5- fold, 10-fold, 25 -fold, 50-fold or 100-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second HER2 inhibitor can exhibit up to 1 OOO-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second HER2 inhibitor can exhibit up to 1 OOOO-fold greater inhibition of wild type HER2 or HER2 having a combination of mutations described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
In other embodiments, a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, in combination with a second HER2 inhibitor can exhibit from about 2-fold to about 10-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second HER2 inhibitor can exhibit from about 10-fold to about 1 OO- fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non- kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second HER2 inhibitor can exhibit from about 100-fold to about 1000-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second HER2 inhibitor can exhibit from about 1 OOO-fold to about 1 OOOO-fold greater inhibition of wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
Compounds of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or pharmaceutically acceptable salts or solvates thereof, are useful for treating diseases and disorders which can be treated with a HER2 inhibitor, such as HER2-associated diseases and disorders, e.g., proliferative disorders such as cancers (e.g., a HER2-associated cancer), including hematological cancers and solid tumors (e.g., advanced solid tumors).
In some embodiments, the compounds provided herein can also inhibit EGFR and HER2 as described herein.
In some embodiments, the compounds provided herein can exhibit potent and selective inhibition of EGFR and HER2. In some embodiments, the compounds provided herein can exhibit nanomolar potency against an EGFR kinase having one or more mutations, including, for example, one or more of the mutations in Tables la, lb and 2a, 2b, and a HER2 kinase having one or more mutations, including, for example, the mutations in Table 3, with minimal activity against related kinases (e.g., wild type EGFR).
In some embodiments, the compounds of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, can selectively target an EGFR and a HER2 kinase. For example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can selectively target an EGFR kinase and a HER2 kinase over another kinase or non-kinase target.
In some embodiments, a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I- c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, can exhibit greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein (e.g., one or more mutations as described in Tables 3-5) relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof can exhibit at least 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or 100-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non- kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit up to 1000-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit up to 1 OOOO-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 having one or more mutations described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
In some embodiments, a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I- c), (I d) or (I-e))> or a pharmaceutically acceptable salt thereof, can exhibit from about 2- fold to about 10-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non- kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit from about 10-fold to about 100-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit from about 1 OO-fold to about 1 OOO-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can exhibit from about 1 OOO-fold to about 1 OOOO- fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
In other embodiments, a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR and/or second HER2 inhibitor can exhibit greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein (e.g., one or more mutations as described in Table 3) relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR and/or second HER2 inhibitor can exhibit at least 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or 100-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR and/or second HER2 inhibitor can exhibit up to 1000-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR and/or HER2 inhibitor can exhibit up to lOOOO-fold greater inhibition of EGFR containing one or more mutations as described herein and wild type HER2 or HER2 having a combination of mutations described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
In other embodiments, a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR and/or second HER2 inhibitor can exhibit from about 2-fold to about 10-fold greater inhibition of EGFR containing one or more mutations as described herein and HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR and/or second HER2 inhibitor can exhibit from about 10-fold to about 100-fold greater inhibition of EGFR containing one or more mutations as described herein and HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR and/or second HER2 inhibitor can exhibit from about 100-fold to about 1000-fold greater inhibition of EGFR containing one or more mutations as described herein and second HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in combination with a second EGFR and/or second HER2 inhibitor can exhibit from about 1000-fold to about 10000-fold greater inhibition of EGFR containing one or more mutations as described herein and HER2 containing one or more mutations as described herein relative to inhibition of another kinase (e.g., wild type EGFR) or non-kinase target.
Also provided herein are methods for inhibiting a BUB (budding uninhibited by benzimidazole, BUB1-3) kinase. For example, provided herein are inhibitors of BUB1 kinase useful for treating or preventing diseases or disorders associated with enhanced uncontrolled proliferative cellular processes such as, for example, cancer, inflammation, arthritis, viral diseases, cardiovascular diseases, or fungal diseases. See, for example, WO 2013/050438, WO 2013/092512, WO 2013/167698, WO 2014/147203, WO 2014/147204, WO 2014/202590, WO 2014/202588, WO 2014/202584, WO 2014/202583, WO 2015/063003, WO2015/193339, WO 2016/202755, and WO 2017/021348. In some embodiments, the disease or disorder is cancer.
A “BUB1 inhibitor” as used herein includes any compound exhibiting BUB1 inactivation activity (e.g., inhibiting or decreasing). In some embodiments, a BUB1 inhibitor can be selective for BUB1 over other kinases (e.g., wildtype EGFR).
The compounds provided herein can inhibit a Bub kinase. In some embodiments, the compounds provided herein can inhibit BUB1 kinase.
The ability of test compounds to act as inhibitors of BUB 1 may be demonstrated by assays known in the art. The activity of the compounds and compositions provided herein as BUB1 inhibitors can be assayed in vitro, in vivo, or in a cell line. In vitro assays include assays that determine inhibition of the kinase. For example, BUB1 inhibition of a compound provided herein can be determined using a time-resolved fluorescence energy transfer (TR-FRET) assay which measures phosphorylation of a synthetic peptide (e.g., Biotin-AHX-VLLPKKSFAEPG (C-terminus in amide form) by the (recombinant) catalytic domain of human BUB1 (amino acids 704-1085), expressed in Hi5 insect cells with an N-terminal His6-tag and purified by affinity- (Ni-NTA) and size exclusion chromatography. See, for example, WO 2017/021348. In addition, BUB1 activity can be determined at a high ATP concentration using a BUB1 TR-FRET high ATP kinase assay using similar methods as those described above. See, e.g. WO 2019/081486.
In some embodiments, the compounds provided herein exhibit central nervous system (CNS) penetrance. For example, such compounds can be capable of crossing the blood brain barrier (BBB) and inhibiting an EGFR and/or HER2 kinase in the brain and/or other CNS structures. In some embodiments, the compounds provided herein are capable of crossing the blood brain barrier in a therapeutically effective amount. For example, treatment of a patient with cancer (e.g., an EGFR-associated cancer or a HER2-associated cancer such as an EGFR- or HER2-associated brain or CNS cancer or an EGFR-associated or a HER2-associated cancer that has metastasized to the brain or CNS) can include administration (e.g., oral administration) of the compound to the patient.
The ability of the compounds described herein, to cross the BBB can be demonstrated by assays known in the art. Such assays include BBB models such as the transwell system, the hollow fiber (dynamic in vitro BBB) model, other microfluidic BBB systems, the BBB spheroid platform, and other cell aggregate-based BBB models. See, e.g., Cho et al. Nat Commun. 2017; 8: 15623; Bagchi et al. Drug Des Devel Ther. 2019; 13: 3591-3605; Gastfriend et al. Curr Opin Biomed Eng. 2018 Mar; 5: 6-12; and Wang et al. Biotechnol Bioeng. 2017 Jan; 114(1): 184-194. In some embodiments, the compounds described herein, are fluorescently labeled, and the fluorescent label can be detected using microscopy (e.g., confocal microscopy). In some such embodiments, the ability of the compound to penetrate the surface barrier of the model can be represented by the fluorescence intensity at a given depth below the surface. In some assays, such as a calcein- AM-based assay, the fluorescent label is non-fluorescent until it permeates live cells and is hydrolyzed by intracellular esterases to produce a fluorescent compound that is retained in the cell and can be quantified with a spectrophotometer. Non-limiting examples of fluorescent labels that can be used in the assays described herein include Cy5, rhodamine, infrared IRDye® CW-800 (LICOR #929-71012), far-red IRDye® 650 (LICOR #929- 70020), sodium fluorescein (Na-F), lucifer yellow (LY), 5 ’carboxyfluorescein, and calcein-acetoxymethylester (calcein-AM). In some embodiments, the BBB model (e.g., the tissue or cell aggregate) can be sectioned, and a compound described herein can be detected in one or more sections using mass spectrometry (e.g., MALDI-MSI analyses). In some embodiments, the ability of a compound described herein to cross the BBB through a transcellular transport system, such as receptor-mediated transport (RMT), carrier- mediated transport (CMT), or active efflux transport (AET), can be demonstrated by assays known in the art. See, e.g., Wang et al. Drug Deliv. 2019; 26(1): 551-565. In some embodiments, assays to determine if compounds can be effluxed by the P-glycoprotein (Pgp) include monolayer efflux assays in which movement of compounds through Pgp is quantified by measuring movement of digoxin, a model Pgp substrate (see, e.g., Doan et al. 2002. J Pharmacol Exp Ther. 303(3): 1029-1037). Alternative in vivo assays to identify compounds that pass through the blood-brain barriers include phage-based systems (see, e.g., Peng et al. 2019. ChemRxiv. Preprint doi.org/10.26434/chemrxiv.8242871.vl). In some embodiments, binding of the compounds described herein to brain tissue is quantified. For example, a brain tissue binding assay can be performed using equilibrium dialysis, and the fraction of a compound described herein unbound to brain tissue can be detected using LC-MS/MS (Cyprotex: Brain Tissue Binding Assay www.cyprotex.com/admepk/protein_binding/brain-tissue-binding/).
Compounds of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or pharmaceutically acceptable salts or solvates thereof, are useful for treating diseases and disorders which can be treated with an EGFR inhibitor, a HER2 inhibitor, a dual EGFR and HER2 inhibitor, and/or a BUB1 inhibitor, such as those described herein, e.g., cancer. Accordingly, provided herein is a method for treating a disease or disorder as provided herein in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments, the disease or disorder is cancer.
As used herein, terms "treat" or "treatment" refer to therapeutic or palliative measures. Beneficial or desired clinical results include, but are not limited to, alleviation, in whole or in part, of symptoms associated with a disease or disorder or condition, diminishment of the extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state (e.g., one or more symptoms of the disease), and remission (whether partial or total), whether detectable or undetectable. "Treatment" can also mean prolonging survival as compared to expected survival if not receiving treatment.
As used herein, the terms "subject," "individual," or "patient," are used interchangeably, refers to any animal, including mammals such as mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, primates, and humans. In some embodiments, the subject is a human. In some embodiments, the subject has experienced and/or exhibited at least one symptom of the disease or disorder to be treated and/or prevented.
In some embodiments, the subject has been identified or diagnosed as having a cancer with a dysregulation of an EGFR gene, an EGFR protein, or expression or activity, or level of any of the same (an EGFR-associated cancer) (e.g., as determined using a regulatory agency-approved, e.g., FDA-approved, assay or kit). In some embodiments, the subject has a tumor that is positive for a dysregulation of an EGFR gene, an EGFR protein, or expression or activity, or level of any of the same (e.g., as determined using a regulatory agency-approved assay or kit). For example, the subject has a tumor that is positive for a mutation as described in Table la and lb. The subject can be a subject with a tumor(s) that is positive for a dysregulation of an EGFR gene, an EGFR protein, or expression or activity, or level of any of the same (e.g., identified as positive using a regulatory agency-approved, e.g., FDA-approved, assay or kit). The subject can be a subject whose tumors have a dysregulation of an EGFR gene, an EGFR protein, or expression or activity, or a level of the same (e.g., where the tumor is identified as such using a regulatory agency-approved, e.g., FDA-approved, kit or assay). In some embodiments, the subject is suspected of having an EGFR-associated cancer. In some embodiments, the subject has a clinical record indicating that the subject has a tumor that has a dysregulation of an EGFR gene, an EGFR protein, or expression or activity, or level of any of the same (and optionally the clinical record indicates that the subject should be treated with any of the compositions provided herein).
In some embodiments, the subject has been identified or diagnosed as having a cancer with a dysregulation of a HER2 gene, a HER2 protein, or expression or activity, or level of any of the same (a HER2-associated cancer) (e.g., as determined using a regulatory agency-approved, e.g., FDA-approved, assay or kit). In some embodiments, the subject has a tumor that is positive for a dysregulation of a HER2 gene, a HER2 protein, or expression or activity, or level of any of the same (e.g., as determined using a regulatory agency- approved assay or kit). For example, the subject has a tumor that is positive for a mutation as described in Table 3. The subject can be a subject with a tumor(s) that is positive for a dysregulation of a HER2 gene, a HER2 protein, or expression or activity, or level of any of the same (e.g., identified as positive using a regulatory agency-approved, e.g., FDA- approved, assay or kit). The subject can be a subject whose tumors have a dysregulation of a HER2 gene, a HER2 protein, or expression or activity, or a level of the same (e.g., where the tumor is identified as such using a regulatory agency-approved, e.g., FDA-approved, kit or assay). In some embodiments, the subject is suspected of having a HER2-associated cancer. In some embodiments, the subject has a clinical record indicating that the subject has a tumor that has a dysregulation of a HER2 gene, a HER2 protein, or expression or activity, or level of any of the same (and optionally the clinical record indicates that the subject should be treated with any of the compositions provided herein).
In some embodiments, the subject is a pediatric subject.
The term “pediatric subject” as used herein refers to a subject under the age of 21 years at the time of diagnosis or treatment. The term “pediatric” can be further be divided into various subpopulations including: neonates (from birth through the first month of life); infants (1 month up to two years of age); children (two years of age up to 12 years of age); and adolescents (12 years of age through 21 years of age (up to, but not including, the twenty-second birthday)). Berhman RE, Kliegman R, Arvin AM, Nelson WE. Nelson Textbook of Pediatrics, 15th Ed. Philadelphia: W.B. Saunders Company, 1996; Rudolph AM, et al. Rudolph ’s Pediatrics, 21st Ed. New York: McGraw-Hill, 2002; and Avery MD, First LR. Pediatric Medicine, 2nd Ed. Baltimore: Williams & Wilkins; 1994. In some embodiments, a pediatric subject is from birth through the first 28 days of life, from 29 days of age to less than two years of age, from two years of age to less than 12 years of age, or 12 years of age through 21 years of age (up to, but not including, the twenty-second birthday). In some embodiments, a pediatric subject is from birth through the first 28 days of life, from 29 days of age to less than 1 year of age, from one month of age to less than four months of age, from three months of age to less than seven months of age, from six months of age to less than 1 year of age, from 1 year of age to less than 2 years of age, from 2 years of age to less than 3 years of age, from 2 years of age to less than seven years of age, from 3 years of age to less than 5 years of age, from 5 years of age to less than 10 years of age, from 6 years of age to less than 13 years of age, from 10 years of age to less than 15 years of age, or from 15 years of age to less than 22 years of age.
In certain embodiments, compounds of Formula (I) (e.g., Formula (I-a), (I-b), (I- c), (I d) or (I-e))> or pharmaceutically acceptable salts or solvates thereof, are useful for preventing diseases and disorders as defined herein (for example, autoimmune diseases, inflammatory diseases, pulmonary disorders, cardiovascular disease, ischemia, liver disease, gastrointestinal disorders, viral or bacterial infections, central nervous system diseases (e.g., neurodegenerative diseases), and cancer). The term "preventing” as used herein means to delay the onset, recurrence or spread, in whole or in part, of the disease or condition as described herein, or a symptom thereof.
The term "EGFR-associated disease or disorder" as used herein refers to diseases or disorders associated with or having a dysregulation of an EGFR gene, an EGFR kinase (also called herein an EGFR kinase protein), or the expression or activity or level of any (e.g., one or more) of the same (e.g., any of the types of dysregulation of an EGFR gene, an EGFR kinase, an EGFR kinase domain, or the expression or activity or level of any of the same described herein). Non-limiting examples of an EGFR-associated disease or disorder include, for example, cancer, a central nervous system disease, a pulmonary disorder, cardiovascular disease, ischemia, liver disease, a gastrointestinal disorder, a viral or bacterial infection, and an inflammatory and/or autoimmune disease (e.g., psoriasis, eczema, atopic dermatitis, and atherosclerosis).
In some embodiments of any of the methods or uses described herein, the inflammatory and/or autoimmune disease is selected from arthritis, systemic lupus erythematosus, atherosclerosis, and skin related disorders such as psoriasis, eczema, and atopic dermatitis. See, e.g., Wang et al. Am J Transl Res. 2019; 11(2): 520-528; Starosyla et al. World J Pharmacol. Dec 9, 2014; 3(4): 162-173; Choi et al. Biomed Res Int. 2018 May 15;2018:9439182; and Wang et al. Sci Rep. 2017; 7: 45917.
In some embodiments of any of the methods or uses described herein, the central nervous system disease is a neurodegenerative disease. In some embodiments, the central nervous system disease is selected from Alzheimer's disease, Parkinson's disease, Huntington’s disease, amyotrophic lateral sclerosis, spinal cord injury, peripheral neuropathy, brain ischemia, and a psychiatric disorder such as schizophrenia. See, e.g., Iwakura and Nawa. Front Cell Neurosci. . 2013 Feb 13;7:4; and Chen et al. Sci Rep. 2019 Feb 21;9(1):2516.
The term “EGFR-associated cancer” as used herein refers to cancers associated with or having a dysregulation of an EGFR gene, an EGFR kinase (also called herein an EGFR kinase protein), or expression or activity, or level of any of the same. Non-limiting examples of an EGFR-associated cancer are described herein.
The phrase “dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same” refers to a genetic mutation (e.g., a mutation in an EGFR gene that results in the expression of an EGFR protein that includes a deletion of at least one amino acid as compared to a wild type EGFR protein, a mutation in an EGFR gene that results in the expression of an EGFR protein with one or more point mutations as compared to a wild type EGFR protein, a mutation in an EGFR gene that results in the expression of an EGFR protein with at least one inserted amino acid as compared to a wild type EGFR protein, a gene duplication that results in an increased level of EGFR protein in a cell, or a mutation in a regulatory sequence (e.g., a promoter and/or enhancer) that results in an increased level of EGFR protein in a cell), an alternative spliced version of an EGFR mRNA that results in an EGFR protein having a deletion of at least one amino acid in the EGFR protein as compared to the wild type EGFR protein), or increased expression (e.g., increased levels) of a wild type EGFR kinase in a mammalian cell due to aberrant cell signaling and/or dysregulated autocrine/paracrine signaling (e.g., as compared to a control non-cancerous cell). As another example, a dysregulation of an EGFR gene, an EGFR protein, or expression or activity, or level of any of the same, can be a mutation in an EGFR gene that encodes an EGFR protein that is constitutively active or has increased activity as compared to a protein encoded by an EGFR gene that does not include the mutation. Non-limiting examples of EGFR kinase protein point mutations/insertions/deletions are described in Table la and lb. Additional examples of EGFR kinase protein mutations (e.g., point mutations) are EGFR inhibitor resistance mutations (e.g., EGFR inhibitor mutations). Non-limiting examples of EGFR inhibitor resistance mutations are described in Table 2a and 2b. For example, the one or more EGFR inhibitor resistance mutations can include a substitution at amino acid position 718, 747, 761, 790, 797, or 854 (e.g., L718Q, L747S, D761Y, T790M, C797S, or T854A). Such mutation and overexpression is associated with the development of a variety of cancers (Shan et al., Cell 2012, 149(4) 860-870).
In some embodiments, dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same can be caused by an activating mutation in an EGFR gene. In some embodiments, dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same can be caused by a genetic mutation that results in the expression of an EGFR kinase that has increased resistance to an EGFR inhibitor, a tyrosine kinase inhibitor (TKI), and/or a multi-kinase inhibitor (MKI), e.g., as compared to a wild type EGFR kinase (see, e.g., the amino acid substitutions in Table 2a and 2b). In some embodiments, dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same can be caused by a mutation in a nucleic acid encoding an altered EGFR protein (e.g., an EGFR protein having a mutation (e.g., a primary mutation)) that results in the expression of an altered EGFR protein that has increased resistance to inhibition by an EGFR inhibitor, a tyrosine kinase inhibitor (TKI), and/or a multi-kinase inhibitor (MKI), e.g., as compared to a wild type EGFR kinase (see, e.g., the amino acid substitutions in Table 2a and 2b). The exemplary EGFR kinase point mutations, insertions, and deletions shown in Tables la, lb and 2a, 2b can be caused by an activating mutation and/or can result in the expression of an EGFR kinase that has increased resistance to an EGFR inhibitor), tyrosine kinase inhibitor (TKI), and/or a multikinase inhibitor (MKI).
In some embodiments, the individual has two or more EGFR inhibitor resistance mutations that increase resistance of the cancer to a first EGFR inhibitor. For example, the individual can have two EGFR inhibitor resistance mutations. In some embodiments, the two mutations occur in the same EGFR protein. In some embodiments, the two mutations occur in separate EGFR proteins. In some embodiments, the individual can have three EGFR inhibitor resistance mutations. In some embodiments, the three mutations occur in the same EGFR protein. In some embodiments, the three mutations occur in separate EGFR proteins. For example, the individual has two or more EGFR inhibitor resistance mutations selected from Del 19/L718Q, Del 19/T790M, Del 19/L844V, Del 19/T790M/L718Q, Del/T790M/C797S, Del 19/T790M/L844V, L858R/L718Q, L858R/L844V, L858R/T790M, L858R/T790M/L718Q, L858R/T790M/C797S, and
L858R/T790M/I941R, or any combination thereof; e.g., any two of the aforementioned EGFR inhibitor resistance mutations.
The term “activating mutation” in reference to EGFR describes a mutation in an EGFR gene that results in the expression of an EGFR kinase that has an increased kinase activity, e.g., as compared to a wild type EGFR kinase, e.g., when assayed under identical conditions. For example, an activating mutation can be a mutation in an EGFR gene that results in the expression of an EGFR kinase that has one or more (e.g., two, three, four, five, six, seven, eight, nine, or ten) amino acid substitutions (e.g., any combination of any of the amino acid substitutions described herein) that has increased kinase activity, e.g., as compared to a wild type EGFR kinase, e.g., when assayed under identical conditions. In another example, an activating mutation can be a mutation in an EGFR gene that results in the expression of an EGFR kinase that has one or more (e.g., two, three, four, five, six, seven, eight, nine, or ten) amino acids deleted, e.g., as compared to a wild type EGFR kinase, e.g., when assayed under identical conditions. In another example, an activating mutation can be a mutation in an EGFR gene that results in the expression of an EGFR kinase that has at least one (e.g., at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 12, at least 14, at least 16, at least 18, or at least 20) amino acid inserted as compared to a wild type EGFR kinase, e.g., the exemplary wild type EGFR kinase described herein, e.g., when assayed under identical conditions. Additional examples of activating mutations are known in the art.
The term "wild type" or "wild-type" describes a nucleic acid (e.g., an EGFR gene or an EGFR mRNA) or protein (e.g., an EGFR protein) sequence that is typically found in a subject that does not have a disease or disorder related to the reference nucleic acid or protein.
The term "wild type EGFR" or "wild-type EGFR" describes an EGFR nucleic acid (e.g., an EGFR gene or an EGFR mRNA) or protein (e.g., an EGFR protein) that is found in a subject that does not have an EGFR-associated disease, e.g., an EGFR-associated cancer (and optionally also does not have an increased risk of developing an EGFR- associated disease and/or is not suspected of having an EGFR-associated disease), or is found in a cell or tissue from a subject that does not have an EGFR-associated disease, e.g., an EGFR-associated cancer (and optionally also does not have an increased risk of developing an EGFR-associated disease and/or is not suspected of having an EGFR- associated disease).
Provided herein is a method of treating cancer (e.g., an EGFR-associated cancer) in a subject in need of such treatment, the method comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I- b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof. For example, provided herein are methods for treating an EGFR- associated cancer in a subject in need of such treatment, the method comprising a) detecting a dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same in a sample from the subject; and b) administering a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof. In some embodiments, the dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same includes one or more EGFR kinase protein point mutations/insertions. Non-limiting examples of EGFR kinase protein point mutations/insertions/deletions are described in Table la and lb. In some embodiments, the EGFR kinase protein point mutations/insertions/deletions are selected from the group consisting of G719S, G719C, G719A, L747S, D761Y, T790M, T854A, L858R, L861Q, a deletion in exon 19 (e.g., L747_A750del), and an insertion in exon 20 (e.g., V769_D770insX, D770_N771insX, N771_P772insX, P772_H773insX, or
H773_V774insX). In some embodiments, the EGFR kinase protein point mutations/insertions/deletions are selected from the group consisting of L858R, deletions in exon 19 (e.g., L747_A750del), L747S, D761Y, T790M, and T854A. In some embodiments, the EGFR kinase protein insertion is an exon 20 insertion. In some embodiments, the EGFR kinase protein insertion is an exon 20 insertion selected from the group consisting of: V769_D770insX, D770_N771insX, N771_P772insX,
P772_H773insX, and H773_V774insX. For example, the EGFR kinase protein insertion is an exon 20 insertion selected from the group consisting of: A767_V769dupASV, V769_D770insASV, D770_N771insNPG, D770_N771insNPY, D770_N771insSVD, D770_N771insGL, N771_H773dupNPH, N771_P772insN, N771_P772insH,
N771_P772insV, P772_H773insDNP, P772_H773insPNP, H773_V774insNPH, H773_V774insH, H773_V774insPH, H773_V774insAH, and P772_H773insPNP; or any combination thereof; e.g., any two or more independently selected exon 20 insertions; e.g., any two independently selected exon 20 insertions (e.g., V769_D770insASV and D770_N771insSVD).
In some embodiments of any of the methods or uses described herein, the cancer (e.g., EGFR-associated cancer) is selected from a hematological cancer (e.g., acute lymphocytic cancer, Hodgkin lymphoma, non-Hodgkin lymphoma, and leukemia such as acute-myelogenous leukemia (AML), chronic-myelogenous leukemia (CML), acute- promyelocytic leukemia, and acute lymphocytic leukemia (ALL)), central or peripheral nervous system tissue cancer, an endocrine or neuroendocrine cancer including multiple neuroendocrine type I and type II tumors, Li-Fraumeni tumors, alveolar rhabdomyosarcoma, bone cancer, brain cancer, breast cancer, cancer of the anus, anal canal, or anorectum, cancer of the eye, cancer of the intrahepatic bile duct, cancer of the joints, cancer of the neck, gallbladder, or pleura, cancer of the nose, nasal cavity, or middle ear, oral cancer, oropharyngeal cancer, nasopharyngeal cancer, respiratory cancer, urogenital cancer, cancer of the vulva, colon cancer, esophageal cancer, tracheal cancer, cervical cancer, gastrointestinal carcinoid tumor, hypopharynx cancer, kidney cancer, larynx cancer, liver cancer, lung cancer, malignant mesothelioma, melanoma, multiple myeloma, nasopharynx cancer, ovarian cancer, pancreatic cancer including pancreatic islet cell cancer, peritoneum, omentum, and mesentery cancer, pharynx cancer, prostate cancer, rectal cancer, renal cancer (e.g., renal cell carcinoma (RCC)), small intestine cancer, soft tissue cancer, stomach cancer, testicular cancer, thyroid cancer, parathyroid cancer, pituitary tumors, adrenal gland tumors, ureter cancer, biliary cancer, and urinary bladder cancer. In some embodiments, the cancer is selected from the group consisting of: head and neck, ovarian, cervical, bladder and oesophageal cancers, pancreatic, gastrointestinal cancer, gastric, breast, endometrial and colorectal cancers, hepatocellular carcinoma, glioblastoma, bladder, lung cancer, e.g., non-small cell lung cancer (NSCLC), bronchioloalveolar carcinoma. In some embodiments, the cancer is pancreatic cancer, head and neck cancer, melanoma, colon cancer, renal cancer, leukemia, lung cancer, or breast cancer. In some cases, the cancer is melanoma, colon cancer, renal cancer, leukemia, or breast cancer.
In some such embodiments, the compounds provided herein are useful for treating a primary brain tumor or metastatic brain tumor. For example, the compounds can be used in the treatment of one or more of gliomas such as glioblastoma (also known as glioblastoma multiforme), astrocytomas, oligodendrogliomas, ependymomas, and mixed gliomas, meningiomas, medulloblastomas, gangliogliomas, schwannomas (neurilemmomas), and craniopharyngiomas (see, for example, Liu et al. J Exp Clin Cancer Res. 2019 May 23;38(1):219); and Ding et al. Cancer Res. 2003 Mar 1;63(5):1106-13). In some embodiments, the brain tumor is a primary brain tumor. In some embodiments, the brain tumor is a metastatic brain tumor, e.g., a metastatic brain tumor from lung cancer, melanoma, breast cancer, ovarian cancer, colorectal cancer, kidney cancer, bladder cancer, or undifferentiated carcinoma. In some embodiments, the brain tumor is a metastatic brain tumor from lung cancer (e.g., non-small cell lung cancer). In some embodiments, the compounds provided herein exhibit brain and/or central nervous system (CNS) penetrance. In some embodiments, the patient has previously been treated with another anticancer agent, e.g., another EGFR and/or HER2 inhibitor (e.g., a compound that is not a compound of Formula I) or a multi-kinase inhibitor.
In some embodiments, the cancer is a cancer of B cell origin. In some embodiments, the cancer is a lineage dependent cancer. In some embodiments, the cancer is a lineage dependent cancer where EGFR or the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, plays a role in the initiation and/or development of the cancer.
In some embodiments, the cancer is an EGFR-associated cancer. Accordingly, also provided herein is a method for treating a subject diagnosed with or identified as having an EGFR-associated cancer, e.g., any of the exemplary EGFR-associated cancers disclosed herein, comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof as defined herein.
In some embodiments, the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, includes one or more deletions (e.g., deletion of an amino acid at position 4), insertions, or point mutation(s) in an EGFR kinase. In some embodiments, dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, includes at least one deletion, insertion, or point mutation in an EGFR gene that results in the production of an EGFR kinase that has one or more of the amino acid substitutions, insertions, or deletions in Table la and lb. In some embodiments, the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, includes a deletion of one or more residues from the EGFR kinase, resulting in constitutive activity of the EGFR kinase domain.
In some embodiments, the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, includes at least one point mutation in an EGFR gene that results in the production of an EGFR kinase that has one or more amino acid substitutions, insertions, or deletions as compared to the wild type EGFR kinase (see, for example, the point mutations listed in Table la and lb). In some embodiments, dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, includes at least one point mutation in an EGFR gene that results in the production of an EGFR kinase that has one or more of the amino acid substitutions, insertions, or deletions in Table la and lb.
In some embodiments, the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, includes an insertion of one or more residues in exon 20 of the EGFR gene (e.g., any of the exon 20 insertions described in Table la and lb). Exon 20 of EGFR has two major regions, the c -helix (residues 762- 766) and the loop following the c-helix (residues 767-774). Studies suggest that for some exon 20 insertions (e.g., insertions after residue 764), a stabilized and ridged active conformation induces resistance to first generation EGFR inhibitors. In some embodiments, the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, includes an insertion of one or more residues in exon 20 selected from the group consisting of: V769_D770insX, D770_N771insX, N771_P772insX, P772_H773insX, and H773_V774insX. For example, the EGFR kinase protein insertion is an exon 20 insertion selected from the group consisting of: A767_V769dupASV, V769_D770insASV, D770_N771insNPG, D770_N771insNPY, D770_N771insSVD, D770_N771insGL, N771_H773dupNPH, N771_P772insN, N771_P772insH, N771_P772insV, P772_H773insDNP, P772_H773insPNP,
H773_V774insNPH, H773_V774insH, H773_V774insPH, H773_V774insAH, and P772_H773insPNP H773insPNP; or any combination thereof; e.g., any two 10 or more independently selected exon 20 insertions; e.g., any two independently selected exon 20 insertions (e.g., V769_D770insASV and D770_N771insSVD).
Table la. EGFR Protein Amino Acid Substitutions/Insertions/DeletionsA
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0001
Figure imgf000165_0001
Figure imgf000166_0001
Figure imgf000167_0001
Figure imgf000168_0001
A The EGFR mutations shown may be activating mutations and/or confer increased resistance of EGFR to an EGFR inhibitor and/or a multi-kinase inhibitor (MKI), e.g., as compared to a wild type EGFR
B Potentially oncogenic variant. See, e.g., Kohsaka, Shinji, et al. Science translational medicine 9.416 (2017): eaan6566.
3PCT Patent Application Publication No. WO2019/246541.
2 Grosse A, Grosse C, Rechsteiner M, Soltermann A. Diagn Pathol. 2019;14(l): 18. Published 2019 Feb 11. doi:10.1186/sl3000-019-0789-l.
3 Stewart EL, Tan SZ, Liu G, Tsao MS. Transl Lung Cancer Res. 2015;4(l):67-81. doi:10.3978/j.issn.2218- 6751.2014.11.06.
4 Pines, Gur, Wolfgang J. Kbstler, and Yosef Yarden. FEBS letters 584.12 (2010): 2699-2706.
5 Yasuda, Hiroyuki, Susumu Kobayashi, and Daniel B. Costa. The Lancet Oncology 13.1 (2012): e23-e31.
6 Kim EY, Cho EN, Park HS, et al. Cancer Biol Ther. 2016;17(3):237-245. doi:10.1080/15384047.2016.1139235.
7Shah, Riyaz, and Jason F. Lester. Clinical Lung Cancer (2019).
8 Aran, Veronica, and Jasminka Omerovic. International journal of molecular sciences 20.22 (2019): 5701. doi: 10.3390/ijms20225701.
9 Beau-Faller, Michele, et al. (2012): 10507-10507. doi: 10.1016/j.semcancer.2019.09.015.
10 Masood, Ashiq, Rama Krishna Kancha, and Janakiraman Subramanian. Seminars in oncology. WB Saunders, 2019. doi: 10.1053/j.seminoncol.2019.08.004.
11 Kohsaka, Shinji, et al. Science translational medicine 9.416 (2017): eaan6566.
12 Vyse and Huang et al. Signal Transduct Target Ther. 2019 Mar 8;4:5. doi: 10.1038/s41392-019-0038-9.
13 PCT Patent Application Publication No. WO2019/046775.
14 PCT Patent Application Publication No. WO 2018/094225.
Table lb. EGFR Protein Amino Acid Substitutions/Insertions/DeletionsA
Figure imgf000168_0002
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
A The EGFR mutations shown may be activating mutations and/or confer increased resistance of EGFR to an EGFR inhibitor and/or a multi-kinase inhibitor (MKI), e.g., as compared to a wild type EGFR
B Potentially oncogenic variant. See, e.g., Kohsaka, Shinji, et al. Science translational medicine 9.416 (2017): eaan6566.
3PCT Patent Application Publication No. WO2019/246541.
2 Grosse A, Grosse C, Rechsteiner M, Soltermann A. Diagn Pathol. 2019;14(l): 18. Published 2019 Feb 11. doi:10.1186/sl3000-019-0789-l.
3 Stewart EL, Tan SZ, Liu G, Tsao MS. Transl Lung Cancer Res. 2015;4(l):67-81. doi:10.3978/j.issn.2218- 6751.2014.11.06.
4 Pines, Gur, Wolfgang J. Kbstler, and Yosef Yarden. FEBS letters 584.12 (2010): 2699-2706.
5 Yasuda, Hiroyuki, Susumu Kobayashi, and Daniel B. Costa. The Lancet Oncology 13.1 (2012): e23-e31.
6 Kim EY, Cho EN, Park HS, et al. Cancer Biol Ther. 2016;17(3):237-245. doi:10.1080/15384047.2016.1139235.
7Shah, Riyaz, and Jason F. Lester. Clinical Lung Cancer (2019).
8 Aran, Veronica, and Jasminka Omerovic. International journal of molecular sciences 20.22 (2019): 5701. doi: 10.3390/ijms20225701.
9 Beau-Faller, Michele, et al. (2012): 10507-10507. doi: 10.1016/j.semcancer.2019.09.015.
10 Masood, Ashiq, Rama Krishna Kancha, and Janakiraman Subramanian. Seminars in oncology. WB Saunders, 2019. doi: 10.1053/j.seminoncol.2019.08.004.
11 Kohsaka, Shinji, et al. Science translational medicine 9.416 (2017): eaan6566.
12 Vyse and Huang et al. Signal Transduct Target Ther. 2019 Mar 8;4:5. doi: 10.1038/s41392-019-0038-9.
13 PCT Patent Application Publication No. WO2019/046775.
14 PCT Patent Application Publication No. WO 2018/094225.
15Mondal, Gourish, et al. Acta Neuropathol. 2020; 139(6): 1071-1088
16Udager, Aaron M., et al. Cancer Res, 2015; 75(13): 2600-2606
In some embodiments, the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, includes a splice variation in an EGFR mRNA which results in an expressed protein that is an alternatively spliced variant of EGFR having at least one residue deleted (as compared to the wild type EGFR kinase) resulting in a constitutive activity of an EGFR kinase domain.
In some embodiments, the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, includes at least one point mutation in an EGFR gene that results in the production of an EGFR kinase that has one or more amino acid substitutions or insertions or deletions in an EGFR gene that results in the production of an EGFR kinase that has one or more amino acids inserted or removed, as compared to the wild type EGFR kinase. In some cases, the resulting EGFR kinase is more resistant to inhibition (e.g., inhibition of its signaling activity) by one or more first EGFR inhibitors, as compared to a wild type EGFR kinase or an EGFR kinase not including the same mutation. Such mutations, optionally, do not decrease the sensitivity of the cancer cell or tumor having the EGFR kinase to treatment with a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof (e.g., as compared to a cancer cell or a tumor that does not include the particular EGFR inhibitor resistance mutation).
In other embodiments, the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, includes at least one point mutation in an EGFR gene that results in the production of an EGFR kinase that has one or more amino acid substitutions as compared to the wild type EGFR kinase, and which has increased resistance to a compound of Formula (I), or a pharmaceutically acceptable salt thereof, as compared to a wild type EGFR kinase or an EGFR kinase not including the same mutation. In such embodiments, an EGFR inhibitor resistance mutation can result in an EGFR kinase that has one or more of an increased Vmax, a decreased Km, and a decreased KD in the presence of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, as compared to a wild type EGFR kinase or an EGFR kinase not having the same mutation in the presence of the same compound of Formula (I), or a pharmaceutically acceptable salt thereof.
Exemplary Sequence of Mature Human EGFR Protein (UniProtKB entry P00533) (SEQ ID NO: 1)
MRPSGTAGAA LLALLAALCP ASRALEEKKV CQGTSNKLTQ LGTFEDHFLS
LQRMFNNCEV VLGNLEITYV QRNYDLSFLK TIQEVAGYVL IALNTVERIP
LENLQI IRGN MYYENSYALA VLSNYDANKT GLKELPMRNL QEILHGAVRF
SNNPALCNVE SIQWRDIVSS DFLSNMSMDF QNHLGSCQKC DPSCPNGSCW
GAGEENCQKL TKI ICAQQCS GRCRGKSPSD CCHNQCAAGC TGPRESDCLV
CRKFRDEATC KDTCPPLMLY NPTTYQMDVN PEGKYSFGAT CVKKCPRNYV
VTDHGSCVRA CGADSYEMEE DGVRKCKKCE GPCRKVCNGI GIGEFKDSLS
INATNIKHFK NCTSISGDLH ILPVAFRGDS FTHTPPLDPQ ELDILKTVKE ITGFLLIQAW PENRTDLHAF ENLEI IRGRT KQHGQFSLAV VSLNITSLGL RSLKEISDGD VI I SGNKNLC YANTINWKKL FGTSGQKTKI I SNRGENSCK ATGQVCHALC SPEGCWGPEP RDCVSCRNVS RGRECVDKCN LLEGEPREFV ENSECIQCHP ECLPQAMNIT CTGRGPDNCI QCAHYIDGPH CVKTCPAGVM GENNTLVWKY ADAGHVCHLC HPNCTYGCTG PGLEGCPTNG PKIPSIATGM VGALLLLLW ALGIGLFMRR RHIVRKRTLR RLLQERELVE PLTPSGEAPN QALLRILKET EFKKIKVLGS GAFGTVYKGL WIPEGEKVKI PVAIKELREA TSPKANKEIL DEAYVMASVD NPHVCRLLGI CLTSTVQLIT QLMPFGCLLD YVREHKDNIG SQYLLNWCVQ IAKGMNYLED RRLVHRDLAA RNVLVKTPQH VKITDFGLAK LLGAEEKEYH AEGGKVPIKW MALES ILHRI YTHQSDVWSY GVTVWELMTF GSKPYDGIPA SEISSILEKG ERLPQPPICT I DVYMIMVKC WMIDADSRPK FRELI IEFSK MARDPQRYLV IQGDERMHLP SPTDSNFYRA LMDEEDMDDV VDADEYLIPQ QGFFSSPSTS RTPLLSSLSA TSNNSTVACI DRNGLQSCPI KEDSFLQRYS SDPTGALTED SIDDTFLPVP EYINQSVPKR PAGSVQNPVY HNQPLNPAPS RDPHYQDPHS TAVGNPEYLN TVQPTCVNST FDSPAHWAQK GSHQISLDNP DYQQDFFPKE AKPNGIFKGS TAENAEYLRV
APQSSEFIGA
In some embodiments, dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, includes at least one EGFR inhibitor resistance mutation in an EGFR gene that results in the production of an EGFR kinase that has one or more of the amino acid substitutions, insertions, or deletions as described in Table 2a and 2b. In some embodiments, compounds of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)) and pharmaceutically acceptable salts and solvates thereof are useful in treating subjects that develop cancers with EGFR inhibitor resistance mutations (e.g., that result in an increased resistance to a first EGFR inhibitor, e.g., a substitution at ammo acid position 718, 747, 761, 790, 797, or 854 (e.g., L718Q, L747S, D761 Y, T790M, C797S, T854A), and/or one or more EGFR inhibitor resistance mutations listed in Table 2a and 2b) by either dosing in combination or as a subsequent or additional (e.g., followup) therapy to existing drug treatments (e.g., other inhibitors of EGFR; e.g., first and/or second EGFR inhibitors). Table 2a. EGFR Protein Amino Acid Resistance Mutations
Figure imgf000178_0001
XPCT Patent Application Publication No. WO2019/246541 2 Stewart EL, Tan SZ, Liu G, Tsao MS. Transl Lung Cancer Res. 2015;4(l):67-81. doi:10.3978/j.issn.2218- 6751.2014.11.06
3 Yasuda, Hiroyuki, Susumu Kobayashi, and Daniel B. Costa. The Lancet Oncology 13.1 (2012): e23-e31.
4 Kim EY, Cho EN, Park HS, et al. Cancer Biol Ther. 2016;17(3):237-245. doi:10.1080/15384047.2016.1139235
5Shah, Riyaz, and Jason F. Lester. Clinical Lung Cancer (2019).
6 Aran, Veronica, and Jasminka Omerovic. International journal of molecular sciences 20.22 (2019): 5701. doi: 10.3390/ijms20225701.
7 Beau-Faller, Michele, et al. (2012): 10507-10507. doi: 10.1016/j.semcancer.2019.09.015 8 Masood, Ashiq, Rama Krishna Kancha, and Janakiraman Subramanian. Seminars in oncology. WB
Saunders, 2019. doi: 10.1053/j.seminoncol.2019.08.004
Table 2b. EGFR Protein Amino Acid Resistance Mutations
Figure imgf000179_0001
Figure imgf000180_0001
3PCT Patent Application Publication No. WO2019/246541
2 Stewart EL, Tan SZ, Liu G, Tsao MS. Transl Lung Cancer Res. 2015;4(l):67-81. doi:10.3978/j.issn.2218- 6751.2014.11.06
3 Yasuda, Hiroyuki, Susumu Kobayashi, and Daniel B. Costa. The Lancet Oncology 13.1 (2012): e23-e31.
4 Kim EY, Cho EN, Park HS, et al. Cancer Biol Ther. 2016;17(3):237-245. doi:10.1080/15384047.2016.1139235
5Shah, Riyaz, and Jason F. Lester. Clinical Lung Cancer (2019).
6 Aran, Veronica, and Jasminka Omerovic. International journal of molecular sciences 20.22 (2019): 5701. doi: 10.3390/ijms20225701.
7 Beau-Faller, Michele, et al. (2012): 10507-10507. doi: 10.1016/j.semcancer.2019.09.015
8 Masood, Ashiq, Rama Krishna Kancha, and Janakiraman Subramanian. Seminars in oncology. WB Saunders, 2019. doi: 10.1053/j.seminoncol.2019.08.004
9Papadimitrakopoulou, V.A., et al. Annals of Oncology 2018; 29 Supplement 8 VIII741
In some embodiments, the EGFR Protein Amino Acid Substitutions/Insertions/Deletions include any one or more, or any two or more (e.g., any two), of the EGFR Protein Amino Acid Substitutions/Insertions/Deletions delineated in Table la, lb and/or Table 2a, 2b; e.g., any one or more, or any two or more (e.g., any two), of the following and independently selected EGFR Protein Amino Acid Substitutions/Insertions/Deletions: V769L; V769M; M766delinsMASVx2;
A767_V769dupASV; A767delinsASVDx3; A767delinsASVG; S768_V769insX; V769_D770insX; V769_D770insASV; D770delinsDN; D770delinsDNPH; D770_N771insSV; N771delinsNPH; N771_H773dup; L858R/C797S (or C797G); or Del_19 and C797S (or C797G), or any combination thereof.
As used herein, a “first inhibitor of EGFR” or “first EGFR inhibitor” is an EGFR inhibitor as defined herein, but which does not include a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof as defined herein. As used herein, a “second inhibitor of EGFR” or a “second EGFR inhibitor” is an EGFR inhibitor as defined herein, but which does not include a compound of Formula (I), or a pharmaceutically acceptable salt thereof as defined herein. When both a first and a second inhibitor of EGFR are present in a method provided herein, the first and second inhibitors of EGFR are different. In some embodiments, the first and/or second inhibitor of EGFR bind in a different location than a compound of Formula (I). For example, in some embodiments, a first and/or second inhibitor of EGFR can inhibit dimerization of EGFR, while a compound of Formula (I) can inhibit the active site. In some embodiments, a first and/or second EGFR inhibitor can be an allosteric inhibitor of EGFR, while a compound of Formula (I) can inhibit the EGFR active site.
Exemplary first and second inhibitors of EGFR are described herein. In some embodiments, a first or second inhibitor of EGFR can be selected from the group consisting of osimertinib, gefitinib, erlotinib, afatinib, lapatinib, neratinib, AZD-9291, CL-387785, CO- 1686, or WZ4002.
In some embodiments, compounds of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or pharmaceutically acceptable salts and solvates thereof are useful for treating a cancer that has been identified as having one or more EGFR inhibitor resistance mutations (that result in an increased resistance to a first or second inhibitor of EGFR, e.g., a substitution described in Table 2a and 2b including substitutions at amino acid position 747, 761, 790, 797, or 854 (e.g., L718Q, L747S, D761Y, T790M, C797S, T854A)). In some embodiments, the one or more EGFR inhibitor resistance mutations occurs in a nucleic acid sequence encoding a mutant EGFR protein (e.g., a mutant EGFR protein having any of the mutations described in Table 2a and 2b) resulting in a mutant EGFR protein that exhibits EGFR inhibitor resistance.
The epidermal growth factor receptor (EGFR) belongs to the ErbB family of receptor tyrosine kinases (RTKs) and provides critical functions in epithelial cell physiology (Schlessinger J (2014) Cold Spring Harb Perspect Biol 6, a008912). It is frequently mutated and/or overexpressed in different types of human cancers and is the target of multiple cancer therapies currently adopted in the clinical practice (Yarden Y and Pines G (2012) Nat Rev Cancer 12, 553-563).
Accordingly, provided herein are methods for treating a subject diagnosed with (or identified as having) a cancer that include administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof.
Also provided herein are methods for treating a subject identified or diagnosed as having an EGFR-associated cancer that include administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof. In some embodiments, the subject that has been identified or diagnosed as having an EGFR-associated cancer through the use of a regulatory agency- approved, e.g., FDA-approved test or assay for identifying dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, in a subject or a biopsy sample from the subject or by performing any of the non-limiting examples of assays described herein. In some embodiments, the test or assay is provided as a kit. In some embodiments, the cancer is an EGFR-associated cancer. For example, the EGFR- associated cancer can be a cancer that includes one or more EGFR inhibitor resistance mutations.
The term "regulatory agency" refers to a country's agency for the approval of the medical use of pharmaceutical agents with the country. For example, a non-limiting example of a regulatory agency is the U.S. Food and Drug Administration (FDA).
Also provided are methods for treating cancer in a subject in need thereof, the method comprising: (a) detecting an EGFR-associated cancer in the subject; and (b) administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof. Some embodiments of these methods further include administering to the subject another anti cancer agent (e.g., a second EGFR inhibitor, a second compound of Formula (I), or a pharmaceutically acceptable salt thereof, or an immunotherapy). In some embodiments, the subject was previously treated with a first EGFR inhibitor or previously treated with another anti cancer treatment, e.g., at least partial resection of the tumor or radiation therapy. In some embodiments, the subject is determined to have an EGFR-associated cancer through the use of a regulatory agency- approved, e.g., FDA-approved test or assay for identifying dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, in a subject or a biopsy sample from the subject or by performing any of the non-limiting examples of assays described herein. In some embodiments, the test or assay is provided as a kit. In some embodiments, the cancer is an EGFR-associated cancer. For example, the EGFR- associated cancer can be a cancer that includes one or more EGFR inhibitor resistance mutations.
Also provided are methods of treating a subject that include performing an assay on a sample obtained from the subject to determine whether the subject has a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, and administering (e.g., specifically or selectively administering) a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof to the subject determined to have a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same. Some embodiments of these methods further include administering to the subject another anticancer agent (e.g., a second EGFR inhibitor, a second compound of Formula (I), or a pharmaceutically acceptable salt thereof, or immunotherapy). In some embodiments of these methods, the subject was previously treated with a first EGFR inhibitor or previously treated with another anti cancer treatment, e.g., at least partial resection of a tumor or radiation therapy. In some embodiments, the subject is a subject suspected of having an EGFR-associated cancer, a subject presenting with one or more symptoms of an EGFR-associated cancer, or a subject having an elevated risk of developing an EGFR-associated cancer. In some embodiments, the assay utilizes next generation sequencing, pyrosequencing, immunohistochemistry, or break apart FISH analysis. In some embodiments, the assay is a regulatory agency-approved assay, e.g., FDA-approved kit. In some embodiments, the assay is a liquid biopsy. Additional, non-limiting assays that may be used in these methods are described herein. Additional assays are also known in the art. In some embodiments, the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same includes one or more EGFR inhibitor resistance mutations.
Also provided is a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof for use in treating an EGFR-associated cancer in a subject identified or diagnosed as having an EGFR-associated cancer through a step of performing an assay (e.g., an in vitro assay) on a sample obtained from the subject to determine whether the subject has a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, where the presence of a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, identifies that the subject has an EGFR- associated cancer. Also provided is the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating an EGFR-associated cancer in a subject identified or diagnosed as having an EGFR- associated cancer through a step of performing an assay on a sample obtained from the subject to determine whether the subject has a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same where the presence of dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, identifies that the subject has an EGFR-associated cancer. Some embodiments of any of the methods or uses described herein further include recording in the subject’s clinical record (e.g., a computer readable medium) that the subject is determined to have a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, through the performance of the assay, should be administered a compound of Formula (I), or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof. In some embodiments, the assay utilizes next generation sequencing, pyrosequencing, immunohistochemistry, or break apart FISH analysis. In some embodiments, the assay is a regulatory agency-approved assay, e.g., FDA-approved kit. In some embodiments, the assay is a liquid biopsy. In some embodiments, the dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same includes one or more EGFR inhibitor resistance mutations.
Also provided is a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, for use in the treatment of a cancer in a subject in need thereof or a subject identified or diagnosed as having an EGFR- associated cancer. Also provided is the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating a cancer in a subject identified or diagnosed as having an EGFR-associated cancer. In some embodiments, the cancer is an EGFR-associated cancer, for example, an EGFR-associated cancer having one or more EGFR inhibitor resistance mutations. In some embodiments, a subject is identified or diagnosed as having an EGFR-associated cancer through the use of a regulatory agency-approved, e.g., FDA-approved, kit for identifying dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same, in a subject or a biopsy sample from the subject. As provided herein, an EGFR-associated cancer includes those described herein and known in the art.
In some embodiments of any of the methods or uses described herein, the subject has been identified or diagnosed as having a cancer with a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same. In some embodiments of any of the methods or uses described herein, the subject has a tumor that is positive for a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same. In some embodiments of any of the methods or uses described herein, the subject can be a subject with a tumor(s) that is positive for a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same. In some embodiments of any of the methods or uses described herein, the subject can be a subject whose tumors have a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same. In some embodiments of any of the methods or uses described herein, the subject is suspected of having an EGFR-associated cancer (e.g., a cancer having one or more EGFR inhibitor resistance mutations). In some embodiments, provided herein are methods for treating an EGFR-associated cancer in a subject in need of such treatment, the method comprising a) detecting a dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same in a sample from the subject; and b) administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof. In some embodiments, the dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same includes one or more EGFR kinase protein point mutations/insertions/deletions. Non-limiting examples of EGFR kinase protein point mutations/insertions/deletions are described in Table la and lb. In some embodiments, the EGFR kinase protein point mutations/insertions/deletions are selected from the group consisting of G719S, G719C, G719A, L747S, D761Y, T790M, T854A, L858R, L861Q, a deletion in exon 19 (e.g., L747_A750del), and an insertion in exon 20. In some embodiments, the EGFR kinase protein point mutations/insertions/deletions are selected from the group consisting of L858R, deletions in exon 19 (e.g., L747_A750del), L747S, D761Y, T790M, and T854A. In some embodiments, the dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same includes one or more EGFR inhibitor resistance mutations. Non-limiting examples of EGFR inhibitor resistance mutations are described in Table 2a and 2b. In some embodiments, the EGFR inhibitor resistance mutation is a substitution at amino acid position 718, 747, 761, 790, 797, or 854 (e.g., L718Q, L747S, D761Y, T790M, C797S, and T854A). In some embodiments, the dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same includes one or more point mutations/insertions/deletions in exon 20. Non-limiting examples of EGFR exon 20 mutations are described in Tables la, lb and 2a, 2b. In some embodiments, the EGFR exon 20 mutation is an exon 20 insertion such as V769_D770insX, D770_N771insX, N771_P772insX, P772_H773insX, and
H773_V774insX. For example, the EGFR kinase protein insertion is an exon 20 insertion selected from the group consisting of: A767_V769dupASV, V769_D770insASV, D770_N771insNPG, D770_N771insNPY, D770_N771insSVD, D770_N771insGL, N771_H773dupNPH, N771_P772insN, N771_P772insH, N771_P772insV,
P772_H773insDNP, P772_H773insPNP, H773_V774insNPH, H773_V774insH, H773_V774insPH, H773_V774insAH, and P772_H773insPNP. In some embodiments, the cancer with a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same is determined using a regulatory agency-approved, e.g., FDA-approved, assay or kit. In some embodiments, the tumor that is positive for a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same is a tumor positive for one or more EGFR inhibitor resistance mutations. In some embodiments, the tumor with a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same is determined using a regulatory agency- approved, e.g., FDA-approved, assay or kit.
In some embodiments of any of the methods or uses described herein, the subject has a clinical record indicating that the subject has a tumor that has a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same (e.g., a tumor having one or more EGFR inhibitor resistance mutations). Also provided are methods of treating a subject that include administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof to a subject having a clinical record that indicates that the subject has a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same.
In some embodiments, the methods provided herein include performing an assay on a sample obtained from the subject to determine whether the subject has a dysregulation of an EGFR gene, an EGFR protein, or expression or level of any of the same. In some such embodiments, the method also includes administering to a subject determined to have a dysregulation of an EGFR gene, an EGFR protein, or expression or activity, or level of any of the same a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof. In some embodiments, the method includes determining that a subject has a dysregulation of an EGFR gene, an EGFR protein, or expression or level of any of the same via an assay performed on a sample obtained from the subject. In such embodiments, the method also includes administering to a subject a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof. In some embodiments, the dysregulation in an EGFR gene, an EGFR kinase protein, or expression or activity or level of any of the same is one or more point mutation in the EGFR gene (e.g., any of the one or more of the EGFR point mutations described herein). The one or more point mutations in an EGFR gene can result, e.g., in the translation of an EGFR protein having one or more of the following amino acid substitutions, deletions, and insertions: G719S, G719C, G719A, L747S, D761Y, T790M, T854A, L858R, L861Q, a deletion in exon 19 (e.g., L747_A750del), and an insertion in exon 20 (e.g., V769_D770insX, D770_N771insX, N771_P772insX, P772_H773insX, and H773_V774insX). The one or more mutations in an EGFR gene can result, e.g., in the translation of an EGFR protein having one or more of the following amino acid substitutions or deletions: L858R, deletions in exon 19 (e.g., L747_A750del), L747S, D761Y, T790M, and T854A. In some embodiments, the dysregulation in an EGFR gene, an EGFR kinase protein, or expression or activity or level of any of the same is one or more EGFR inhibitor resistance mutations (e.g., any combination of the one or more EGFR inhibitor resistance mutations described herein). In some embodiments, the dysregulation in an EGFR gene, an EGFR kinase protein, or expression or activity or level of any of the same is one or more EGFR exon 20 insertions (e.g., any of the exon 20 insertions described herein). In some embodiments, the EGFR kinase protein insertion is an exon 20 insertion selected from the group consisting of: V769_D770insX, D770_N771insX, N771_P772insX, P772_H773insX, and
H773_V774insX. In some embodiments, the EGFR kinase protein insertion is an exon 20 insertion selected from the group consisting of: V769_D770insX, D770_N771insX, N771_P772insX, P772_H773insX, and H773_V774insX. In some embodiments, the EGFR kinase protein insertion is an exon 20 insertion selected from the group consisting of: A767_V769dupASV, V769_D770insASV, D770_N771insNPG, D770_N771insNPY, D770_N771insSVD, D770_N771insGL, N771_H773dupNPH, N771_P772insN, N771_P772insH, N771_P772insV, P772_H773insDNP, P772_H773insPNP,
H773_V774insNPH, H773_V774insH, H773_V774insPH, H773_V774insAH, and P772_H773insPNP. Some embodiments of these methods further include administering to the subject another anti cancer agent (e.g., a second EGFR inhibitor, a second compound of Formula (I), or a pharmaceutically acceptable salt thereof, or immunotherapy).
In some embodiments of any of the methods or uses described herein, an assay used to determine whether the subject has a dysregulation of an EGFR gene, or an EGFR kinase, or expression or activity or level of any of the same, using a sample from a subject can include, for example, next generation sequencing, immunohistochemistry, fluorescence microscopy, break apart FISH analysis, Southern blotting, Western blotting, FACS analysis, Northern blotting, and PCR-based amplification (e.g., RT-PCR and quantitative real-time RT-PCR). As is well-known in the art, the assays are typically performed, e.g., with at least one labelled nucleic acid probe or at least one labelled antibody or antigenbinding fragment thereof. Assays can utilize other detection methods known in the art for detecting dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or levels of any of the same (see, e.g., the references cited herein). In some embodiments, the dysregulation of the EGFR gene, the EGFR kinase, or expression or activity or level of any of the same includes one or more EGFR inhibitor resistance mutations. In some embodiments, the sample is a biological sample or a biopsy sample (e.g., a paraffin- embedded biopsy sample) from the subject. In some embodiments, the subject is a subject suspected of having an EGFR-associated cancer, a subject having one or more symptoms of an EGFR-associated cancer, and/or a subject that has an increased risk of developing an EGFR-associated cancer).
In some embodiments, dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same can be identified using a liquid biopsy (variously referred to as a fluid biopsy or fluid phase biopsy). See, e.g., Karachialiou et al., “Real-time liquid biopsies become a reality in cancer treatment”, Ann. Transl. Med., 3(3): 36, 2016. Liquid biopsy methods can be used to detect total tumor burden and/or the dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same. Liquid biopsies can be performed on biological samples obtained relatively easily from a subject (e.g., via a simple blood draw) and are generally less invasive than traditional methods used to detect tumor burden and/or dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same. In some embodiments, liquid biopsies can be used to detect the presence of dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same at an earlier stage than traditional methods. In some embodiments, the biological sample to be used in a liquid biopsy can include, blood, plasma, urine, cerebrospinal fluid, saliva, sputum, broncho-alveolar lavage, bile, lymphatic fluid, cyst fluid, stool, ascites, and combinations thereof. In some embodiments, a liquid biopsy can be used to detect circulating tumor cells (CTCs). In some embodiments, a liquid biopsy can be used to detect cell-free DNA. In some embodiments, cell-free DNA detected using a liquid biopsy is circulating tumor DNA (ctDNA) that is derived from tumor cells. Analysis of ctDNA (e.g., using sensitive detection techniques such as, without limitation, next-generation sequencing (NGS), traditional PCR, digital PCR, or microarray analysis) can be used to identify dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same.
The term "HER2-associated disease or disorder" as used herein refers to diseases or disorders associated with or having a dysregulation of a HER2 gene, a HER2 kinase, or the expression or activity or level of any (e.g., one or more) of the same (e.g., any of the types of dysregulation of a HER2 gene, a HER2 kinase, a HER2 kinase domain, or the expression or activity or level of any of the same described herein). Non-limiting examples of a HER2-associated disease or disorder include, for example, cancer.
The term “HER2-associated cancer” as used herein refers to cancers associated with or having a dysregulation of a HER2 gene, a HER2 kinase (also called herein a HER2 protein), or expression or activity, or level of any of the same. Non-limiting examples of a HER2-associated cancer are described herein.
In some embodiments, the EGFR-associated cancer is also a HER2-associated cancer. For example, an EGFR-associated cancer can also have a dysregulation of a HER2 gene, a HER2 kinase, or the expression or activity or level of any of the same.
The phrase “dysregulation of a HER2 gene, a HER2 kinase, or the expression or activity or level of any of the same” refers to a genetic mutation (e.g., a mutation in a HER2 gene that results in the expression of a HER2 protein that includes a deletion of at least one amino acid as compared to a wild type HER2 protein, a mutation in a HER2 gene that results in the expression of a HER2 protein with one or more point mutations as compared to a wild type HER2 protein, a mutation in a HER2 gene that results in the expression of a HER2 protein with at least one inserted amino acid as compared to a wild type HER2 protein, a gene duplication that results in an increased level of HER2 protein in a cell, or a mutation in a regulatory sequence (e.g., a promoter and/or enhancer) that results in an increased level of HER2 protein in a cell), an alternative spliced version of a HER2 mRNA that results in a HER2 protein having a deletion of at least one amino acid in the HER2 protein as compared to the wild- type HER2 protein), or increased expression (e.g., increased levels) of a wild type HER2 kinase in a mammalian cell due to aberrant cell signaling and/or dysregulated autocrine/paracrine signaling (e.g., as compared to a control non-cancerous cell). As another example, a dysregulation of a H ER2 gene, a HER2 protein, or expression or activity, or level of any of the same, can be a mutation in a HER2 gene that encodes a HER2 protein that is constitutively active or has increased activity as compared to a protein encoded by a HER2 gene that does not include the mutation. Nonlimiting examples of HER2 kinase protein fusions and point mutations/insertions/deletions are described in Tables 3-5. Such mutation and overexpression is associated with the development of a variety of cancers (Moasser. Oncogene. 2007 Oct 4; 26(45): 6469-6487).
Compounds of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or pharmaceutically acceptable salts or solvates thereof, are useful for treating diseases and disorders such as HER2-associated diseases and disorders, e.g., proliferative disorders such as cancers, including hematological cancers and solid tumors (e.g., advanced solid tumors).
In some embodiments, dysregulation of a HER2 gene, a HER2 kinase, or the expression or activity or level of any of the same can be caused by an activating mutation in a HER2 gene. The exemplary HER2 kinase fusions or point mutations, insertions, and deletions shown in Tables 3-5 can be caused by an activating mutation.
The term “activating mutation” in reference to HER2 describes a mutation in a HER2 gene that results in the expression of a HER2 kinase that has an increased kinase activity, e.g., as compared to a wild type HER2 kinase, e.g., when assayed under identical conditions. For example, an activating mutation can be a mutation in a HER2 gene (that results in the expression of a HER2 kinase that has one or more (e.g., two, three, four, five, six, seven, eight, nine, or ten) amino acid substitutions (e.g., any combination of any of the amino acid substitutions described herein) that has increased kinase activity, e.g., as compared to a wild type HER2 kinase, e.g., when assayed under identical conditions. In another example, an activating mutation can be a mutation in a HER2 gene that results in the expression of a HER2 kinase that has one or more (e.g., two, three, four, five, six, seven, eight, nine, or ten) amino acids deleted, e.g., as compared to a wild type HER2 kinase, e.g., when assayed under identical conditions. In another example, an activating mutation can be a mutation in a HER2 gene that results in the expression of a HER2 kinase that has at least one (e.g., at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 12, at least 14, at least 16, at least 18, or at least 20) amino acid inserted as compared to a wild type HER2 kinase, e.g., the exemplary wild type HER2 kinase described herein, e.g., when assayed under identical conditions. Additional examples of activating mutations are known in the art.
The term "wild type HER2" or "wild-type HER2 kinase" describes a HER2nucleic acid (e.g., a HER2 gene or a HER2 mRNA) or protein (e.g., a HER2 protein) that is found in a subject that does not have a HER2-associated disease, e.g., a HER2-associated cancer (and optionally also does not have an increased risk of developing a HER2-associated disease and/or is not suspected of having a HER2-associated disease), or is found in a cell or tissue from a subject that does not have a HER2-associated disease, e.g., a HER2- associated cancer (and optionally also does not have an increased risk of developing a HER2-associated disease and/or is not suspected of having a HER2-associated disease).
Provided herein is a method of treating a HER2-associated cancer (in a subject in need of such treatment, the method comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof. For example, provided herein are methods for treating a HER2- associated cancer in a subject in need of such treatment, the method comprising a) detecting a dysregulation of a HER2 gene, a HER2 kinase, or the expression or activity or level of any of the same in a sample from the subject; and b) administering a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof. In some embodiments, the dysregulation of a HER2 gene, a HER2 kinase, or the expression or activity or level of any of the same includes one or more HER2 kinase protein point mutations/insertions. Non-limiting examples of HER2 kinase protein fusions and point mutations/insertions/deletions are described in Tables 3-5. In some embodiments, the HER2 kinase protein point mutations/insertions/deletions are selected from the group consisting of S310F, S310Y, R678Q, R678W, R678P, I767M, V773M, V777L, V842I, Y772_A775dup, A775_G776insYVMA, G776dehnsVC, G776dehnsW, V777_G778insGSP, and P780_Y781insGSP. In some embodiments, the HER2 kinase protein point mutations/insertions/deletions are exon 20 point mutations/insertions/deletions selected from the group consisting of V773M, G776C, G776V, G776S, V777L, V777M, S779T, P780L, S783P, M774AYVM, M774del insWLV, A775_G776insYVMA, A775_G776insAVMA, A775_G776insSVMA, A775_G776insVAG, A775insV G776C, A775_G776insI, G776del insVC2, G776del insW, G776del insLC, G776C V777insC, G776C V777insV, V777_G778insCG, G778_S779insCPG, and P780_Y781insGSP. In some embodiments, the HER2 kinase protein point mutations/insertions/deletions are exon 20 point mutations/insertions/deletions selected from the group consisting of Y772_A775dup, A775_G776insYVMA, G776delinsVC, G776dehnsW, V777_G778insGSP, and P780_Y781insGSP.
In some embodiments of any of the methods or uses described herein, the cancer (e.g., HER2-associated cancer) is selected from a hematological cancer (e.g., Hodgkin lymphoma, non-Hodgkin lymphoma, and leukemia such as acute-myelogenous leukemia (AML), chronic-myelogenous leukemia (CML), acute-promyelocytic leukemia, and acute lymphocytic leukemia (ALL)), alveolar rhabdomyosarcoma, central or peripheral nervous system tissue cancer, an endocrine or neuroendocrine cancer including multiple neuroendocrine type I and type II tumors, Li-Fraumeni tumors, alveolar rhabdomyosarcoma, bone cancer, brain cancer, breast cancer, cancer of the anus, anal canal, or anorectum, cancer of the eye, cancer of the intrahepatic bile duct, cancer of the joints, cancer of the neck, gallbladder, or pleura, cancer of the nose, nasal cavity, or middle ear, tracheal cancer, oral cancer, oropharyngeal cancer, nasopharyngeal cancer, respiratory cancer, urogenital cancer, cancer of the vulva, colon cancer, esophageal cancer, cervical cancer, gastrointestinal carcinoid tumor, hypopharynx cancer, kidney cancer, larynx cancer, liver cancer, lung cancer, malignant mesothelioma, melanoma, multiple myeloma, nasopharynx cancer, non-Hodgkin lymphoma, ovarian cancer, pancreatic cancer including pancreatic islet cell cancer, peritoneum, omentum, and mesentery cancer, pharynx cancer, prostate cancer, rectal cancer, renal cancer (e.g., renal cell carcinoma (RCC)), small intestine cancer, soft tissue cancer, stomach cancer, testicular cancer, thyroid cancer, parathyroid cancer, pituitary tumors, adrenal gland tumors, ureter cancer, biliary cancer, and urinary bladder cancer. In some embodiments, the cancer is selected from the group consisting of: head and neck, ovarian, cervical, bladder and oesophageal cancers, pancreatic, gastrointestinal cancer, gastric, breast, endometrial and colorectal cancers, hepatocellular carcinoma, glioblastoma, bladder, lung cancer, e.g., non-small cell lung cancer (NSCLC), bronchioloalveolar carcinoma. In some embodiments, the cancer is pancreatic cancer, head and neck cancer, melanoma, colon cancer, renal cancer, leukemia, lung cancer, or breast cancer. In some cases, the cancer is melanoma, colon cancer, renal cancer, leukemia, or breast cancer.
In some such embodiments, the compounds provided herein are useful for treating a primary brain tumor or metastatic brain tumor. For example, the compounds can be used in the treatment of one or more of gliomas such as glioblastoma (also known as glioblastoma multiforme), astrocytomas, oligodendrogliomas, ependymomas, and mixed gliomas, meningiomas, medulloblastomas, gangliogliomas, schwannomas (neurilemmomas), and craniopharyngiomas (see, for example, Liu et al. J Exp Clin Cancer Res. 2019 May 23;38(1):219); and Ding et al. Cancer Res. 2003 Mar 1;63(5):1106-13). In some embodiments, the brain tumor is a primary brain tumor. In some embodiments, the brain tumor is a metastatic brain tumor, e.g., a metastatic brain tumor from lung cancer, melanoma, breast cancer, ovarian cancer, colorectal cancer, kidney cancer, bladder cancer, or undifferentiated carcinoma. In some embodiments, the brain tumor is a metastatic brain tumor from lung cancer (e.g., non-small cell lung cancer). In some embodiments, the compounds provided herein exhibit brain and/or central nervous system (CNS) penetrance. In some embodiments, the patient has previously been treated with another anticancer agent, e.g., another EGFR and/or HER2 inhibitor (e.g., a compound that is not a compound of Formula I) or a multi-kinase inhibitor.
In some embodiments, the cancer is a cancer of B cell origin. In some embodiments, the cancer is a lineage dependent cancer. In some embodiments, the cancer is a lineage dependent cancer where HER2 or the dysregulation of an HER2 gene, an HER2 kinase, or expression or activity or level of any of the same, plays a role in the initiation and/or development of the cancer.
Also provided herein is a method for treating a subject diagnosed with or identified as having a HER2-associated cancer, e.g., any of the exemplary HER2-associated cancers disclosed herein, comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof as defined herein.
In some embodiments, the dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, includes one or more deletions (e.g., deletion of an amino acid at position 12), insertions, or point mutation(s) in a HER2 kinase. In some embodiments, the dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, includes a deletion of one or more residues from the HER2 kinase, resulting in increased signaling activity of HER2.
In some embodiments, the dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, includes at least one point mutation in a HER2 gene that results in the production of a HER2 kinase that has one or more amino acid substitutions, insertions, or deletions as compared to the wild-type HER2 kinase (see, for example, the point mutations listed in Table 3). In some embodiments, dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, includes at least one point mutation in a HER2 gene that results in the production of a HER2 kinase that has one or more of the amino acid substitutions, insertions, or deletions in Table 3.
In some embodiments, the dysregulation of an HER2 gene, an HER2 kinase, or expression or activity or level of any of the same, includes an insertion of one or more residues in exon 20 of the HER2 gene (e.g., any of the exon 20 insertions described in Table la and lb). Exon 20 of HER2 has two major regions, the c-helix (residues 770-774) and the loop following the c-helix (residues 775-783). In some embodiments, the dysregulation of an HER2 gene, an HER2 kinase, or expression or activity or level of any of the same, includes an insertion of one or more residues in exon 20 selected from the group consisting of: Y772_A775dup, A775_G776insYVMA, G776delinsVC, G776delinsW, V777_G778insGSP, and P780_Y781insGSP.
Table 3. HER2 Protein Amino Acid Substitutions/Insertions/DeletionsA
Figure imgf000195_0001
Figure imgf000196_0001
Figure imgf000197_0001
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000200_0001
Figure imgf000201_0001
A The HER2 mutations shown may be activating mutations and/or confer increased resistance of HER2 to a HER2 inhibitor and/or a multi-kinase inhibitor (MKI), e.g., as compared to a wildtype HER2.
Li et al. J Thorac Oncol. 2016 Mar;l 1(3):414-9.
2 Arcila et al. Clin Cancer Res. 2012 Sep 15; 18(18): 10.1158/1078-0432.CCR-12-0912.
3 Bose et al. Cancer Discov. 2013 Feb;3(2):224-37.
4 Hanker et al. Cancer Discov. 2017 Jun;7(6):575-585.
5 Christgen et al. Virchows Arch. 2018 Nov;473(5):577-582.
6 Si et al. Cancer Biomark. 2018;23(2): 165-171.
7Kavuri et al. Cancer Discov. 2015 Aug; 5(8): 832-841.
8Robichaux et al. Nat Med. 2018 May; 24(5): 638-646.
9Kosaka et al. Cancer Res. 2017 May 15; 77(10): 2712-2721.
10Pahuja et al. Cancer Cell. 2018 Nov 12; 34(5): 792-806.e5.
11 Ross et al. Cancer. 2018 Apr 1 ;124(7): 1358-1373.
12 Gharib et al. J Cell Physiol. 2019 Aug;234(8):13137-13144.
13 Krawczyk et al. Oncol Lett. 2013 Oct; 6(4): 1063-1067.
14 Lai et al. Eur J Cancer. 2019 Mar; 109: 28-35.
15 Sun et al. J Cell Mol Med. 2015 Dec; 19(12): 2691-2701.
16 Xu et al. Thorac Cancer. 2020 Mar;l l(3):679-685.
In some embodiments, the dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, includes a splice variation in a HER2 mRNA which results in an expressed protein that is an alternatively spliced variant of HER2 having at least one residue deleted (as compared to the wild-type HER2 kinase) resulting in a constitutive activity of a HER2 kinase domain. In some embodiments, the splice variant of HER2 is A 16HER-3 or p95HER - 2. See, e.g., Sun et al. J Cell Mol Med. 2015 Dec; 19(12): 2691 - 2701.
In some embodiments, dysregulation of an HER2 gene, an HER2 kinase, or the expression or activity or level of any of the same can be caused by a splice variation in a HER2 mRNA that results in the expression of an altered HER2 protein that has increased resistance to inhibition by an HER2 inhibitor, a tyrosine kinase inhibitor (TKI), and/or a multi-kinase inhibitor (MKI), e.g., as compared to a wild type HER2 kinase (e.g., the HER2 variants described herein). See, e.g., Rexer and Arteaga. Crit Rev Oncog. 2012; 17(1): 1- 16.
In some embodiments, the dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, includes one or more chromosome translocations or inversions resulting in HER2 gene fusions, respectively. In some embodiments, the dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, is a result of genetic translocations in which the expressed protein is a fusion protein containing residues from a non-HER2 partner protein and HER2, and include a minimum of a functional HER2 kinase domain, respectively.
Table 4. Exemplary HER2 Fusion Proteins and Cancers
Figure imgf000202_0001
1 Yu et al. J Transl Med. 2015; 13 : 116.
In some embodiments, the dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, includes at least one point mutation in a HER2 gene that results in the production of a HER2 kinase that has one or more amino acid substitutions or insertions or deletions in a HER2 gene that results in the production of a HER2 kinase that has one or more amino acids inserted or removed, as compared to the wild-type HER2 kinase.
In other embodiments, the dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, includes at least one point mutation in a HER2 gene that results in the production of a HER2 kinase that has one or more amino acid substitutions as compared to the wild-type HER2 kinase, and which has increased resistance to a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, as compared to a wild type HER2 kinase or a HER2 kinase not including the same mutation.
Exemplary Sequence of Mature Human HER2 Protein (UniProtKB entry P04626) (SEQ ID NO: 2) MELAALCRWG LLLALLPPGA ASTQVCTGTD MKLRLPASPE THLDMLRHLY QGCQWQGNL ELTYLPTNAS LSFLQDIQEV QGYVLIAHNQ VRQVPLQRLR IVRGTQLFED NYALAVLDNG DPLNNTTPVT GASPGGLREL QLRSLTEILK GGVLIQRNPQ LCYQDTILWK DIFHKNNQLA LTLIDTNRSR ACHPCSPMCK GSRCWGESSE DCQSLTRTVC AGGCARCKGP LPTDCCHEQC AAGCTGPKHS DCLACLHFNH SGICELHCPA LVTYNTDTFE SMPNPEGRYT FGASCVTACP YNYLSTDVGS CTLVCPLHNQ EVTAEDGTQR CEKCSKPCAR VCYGLGMEHL REVRAVTSAN IQEFAGCKKI FGSLAFLPES FDGDPASNTA PLQPEQLQVF ETLEEITGYL YISAWPDSLP DLSVFQNLQV IRGRILHNGA YSLTLQGLGI SWLGLRSLRE LGSGLALIHH NTHLCFVHTV PWDQLFRNPH QALLHTANRP EDECVGEGLA CHQLCARGHC WGPGPTQCVN CSQFLRGQEC VEECRVLQGL PREYVNARHC LPCHPECQPQ NGSVTCFGPE ADQCVACAHY KDPPFCVARC PSGVKPDLSY MPIWKFPDEE GACQPCPINC THSCVDLDDK GCPAEQRASP LTSI I SAWG ILLVWLGW FGILIKRRQQ KIRKYTMRRL LQETELVEPL TPSGAMPNQA QMRILKETEL RKVKVLGSGA FGTVYKGIWI PDGENVKI PV Al KVL RENTS PKANKEILDE AYVMAGVGSP YVSRLLGICL TSTVQLVTQL MPYGCLLDHV RENRGRLGSQ DLLNWCMQIA KGMSYLEDVR LVHRDLAARN VLVKS PNHVK ITDFGLARLL DIDETEYHAD GGKVPIKWMA LES ILRRRFT HQSDVWSYGV TVWELMTFGA KPYDGIPARE IPDLLEKGER LPQPPICTID
VYMIMVKCWM IDSECRPRFR ELVSEFSRMA RDPQRFWIQ NEDLGPASPL DSTFYRSLLE DDDMGDLVDA EEYLVPQQGF FCPDPAPGAG GMVHHRHRSS STRSGGGDLT LGLEPSEEEA PRSPLAPSEG AGSDVFDGDL GMGAAKGLQS LPTHDPSPLQ RYSEDPTVPL PSETDGYVAP LTCSPQPEYV NQPDVRPQPP SPREGPLPAA RPAGATLERP KTLSPGKNGV VKDVFAFGGA VENPEYLTPQ GGAAPQPHPP PAFSPAFDNL YYWDQDPPER GAPPSTFKGT PTAENPEYLG LDVPV
In some embodiments, dysregulation of an HER2 gene, an HER2 kinase, or expression or activity or level of any of the same, includes at least one HER2 inhibitor resistance mutation in an HER2 gene that results in the production of an HER2 kinase that has one or more of the amino acid substitutions, insertions, or deletions as described in Table 5. In some embodiments, compounds of Formula (I) (e.g., Formula (I-a), (I-b), (I- c), (I d) or (I-e)) and pharmaceutically acceptable salts and solvates thereof are useful in treating subjects that develop cancers with HER2 inhibitor resistance mutations (e.g., that result in an increased resistance to a first HER2 inhibitor, e.g., a substitution at amino acid position 755 or 798 (e.g., L755S, L755P, T798I, and T798M), and/or one or more HER2 inhibitor resistance mutations listed in Table 5) by either dosing in combination or as a subsequent or additional (e.g., follow-up) therapy to existing drug treatments (e.g., other inhibitors of HER2; e.g., first and/or second HER2 inhibitors).
Table 5. HER2 Protein Amino Acid Resistance Mutations
Figure imgf000204_0001
' Hanker et al. Cancer Discov. 2017 Jun;7(6):575-585.
2 Sun et al. J Cell Mol Med. 2015 Dec; 19(12): 2691-2701.
As used herein, a “first inhibitor of HER2” or “first HER2 inhibitor” is a HER2 inhibitor as defined herein, but which does not include a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof as defined herein. As used herein, a “second inhibitor of HER2” or a “second HER2 inhibitor” is a HER2 inhibitor as defined herein, but which does not include a compound of Formula (I), or a pharmaceutically acceptable salt thereof as defined herein. When both a first and a second inhibitor of HER2 are present in a method provided herein, the first and second inhibitors of HER2 are different. In some embodiments, the first and/or second inhibitor of HER2 bind in a different location than a compound of Formula (I). For example, in some embodiments, a first and/or second inhibitor of HER2 can inhibit dimerization of HER2, while a compound of Formula (I) can inhibit the active site. In some embodiments, a first and/or second inhibitor of HER2 can be an allosteric inhibitor of HER2, while a compound of Formula (I) can inhibit the HER2 active site.
Exemplary first and second inhibitors of HER2 are described herein. In some embodiments, a first or second inhibitor of HER2 can be selected from the group consisting of: trastuzumab (e.g., TRAZIMERA™, HERCEPTIN®), pertuzumab (e.g., PERJETA®), trastuzumab emtansine (T-DM1 or ado-trastuzumab emtansine, e.g., KADCYLA®), lapatinib, KU004, neratinib (e.g., NERLYNX®), dacomitinib (e.g., VIZIMPRO®), afatinib (GILOTRIF®), tucatinib (e.g., TUKYSA™), erlotinib (e.g., TARCEVA®), pyrotinib, poziotinib, CP-724714, CUDC-101, sapitinib (AZD8931), tanespimycin (17- AAG), IPI-504, PF299, pelitimb, S- 22261 1, and AEE-788.
In some embodiments, compounds of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or pharmaceutically acceptable salts and solvates thereof are useful for treating a cancer that has been identified as having one or more HERZ inhibitor resistance mutations (that result in an increased resistance to a first or second inhibitor of HERZ, e.g., a substitution described in Table 5 including substitutions at amino acid position 755 or 798 (e.g., L755S, L755P, T798I, and T798M)). In some embodiments, the one or more HERZ inhibitor resistance mutations occurs in a nucleic acid sequence encoding a mutant HERZ protein (e.g., a mutant HERZ protein having any of the mutations described in Table 3) resulting in a mutant HER2 protein that exhibits HER2 inhibitor resistance.
Like EGFR, the epidermal growth factor receptor 2 (HER2) belongs to the ErbB family of receptor tyrosine kinases (RTKs) and provides critical functions in epithelial cell physiology (Schlessinger J (2014) Cold Spring Harb Perspect Biol 6, a008912; and Moasser. Oncogene. 2007 Oct 4; 26(45): 6469-6487). It is frequently mutated and/or overexpressed in different types of human cancers and is the target of multiple cancer therapies currently adopted in the clinical practice (Moasser. Oncogene. 2007 Oct 4; 26(45): 6469-6487).
Accordingly, provided herein are methods for treating a subject identified or diagnosed as having a HERZ-associated cancer that include administering to the subject a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof. In some embodiments, the subject that has been identified or diagnosed as having a HER2-associated cancer through the use of a regulatory agency- approved, e.g., FDA-approved test or assay for identifying dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, in a subject or a biopsy sample from the subject or by performing any of the non-limiting examples of assays described herein. In some embodiments, the test or assay is provided as a kit. In some embodiments, the cancer is a HER2-associated cancer. Also provided are methods for treating cancer in a subject in need thereof, the method comprising: (a) detecting a HER2- associated cancer in the subject; and (b) administering to the subject a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof. Some embodiments of these methods further include administering to the subject another anti cancer agent (e.g., a second HER2 inhibitor, a second compound of Formula (I), or a pharmaceutically acceptable salt thereof, or an immunotherapy). In some embodiments, the subject was previously treated with a first HER2 inhibitor or previously treated with another anticancer treatment, e.g., at least partial resection of the tumor or radiation therapy. In some embodiments, the subject is determined to have a HER2-associated cancer through the use of a regulatory agency- approved, e.g., FDA-approved test or assay for identifying dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, in a subject or a biopsy sample from the subject or by performing any of the non-limiting examples of assays described herein. In some embodiments, the test or assay is provided as a kit. In some embodiments, the cancer is a HER2-associated cancer.
Also provided are methods of treating a subject that include performing an assay on a sample obtained from the subject to determine whether the subject has a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, and administering (e.g., specifically or selectively administering) a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof to the subject determined to have a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same. Some embodiments of these methods further include administering to the subject another anti cancer agent (e.g., a second HER2 inhibitor, a second compound of Formula (I), or a pharmaceutically acceptable salt thereof, or immunotherapy). In some embodiments of these methods, the subject was previously treated with a first HER2 inhibitor or previously treated with another anticancer treatment, e.g., at least partial resection of a tumor or radiation therapy. In some embodiments, the subject is a subject suspected of having a HER2-associated cancer, a subject presenting with one or more symptoms of a HER2-associated cancer, or a subject having an elevated risk of developing a HER2-associated cancer. In some embodiments, the assay utilizes next generation sequencing, pyrosequencing, immunohistochemistry, or break apart FISH analysis. In some embodiments, the assay is a regulatory agency-approved assay, e.g., FDA-approved kit. In some embodiments, the assay is a liquid biopsy. Additional, nonlimiting assays that may be used in these methods are described herein. Additional assays are also known in the art.
As used herein, a “first inhibitor of HER2” or “first HER2 inhibitor” is a HER2 inhibitor as defined herein, which does not include a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof as defined herein. As used herein, a “second inhibitor of HER2” or a “second HER2 inhibitor” is an inhibitor of HER2 as defined herein, which does not include a compound of Formula (I), or a pharmaceutically acceptable salt thereof as defined herein. When both a first and a second HER2 inhibitor are present in a method provided herein, the first and second HER2 inhibitors are different.
Also provided is a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof for use in treating a HER2-associated cancer in a subject identified or diagnosed as having a HER2-associated cancer through a step of performing an assay (e.g., an in vitro assay) on a sample obtained from the subject to determine whether the subject has a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, where the presence of a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, identifies that the subject has a HER2- associated cancer. Also provided is the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating a HER2-associated cancer in a subject identified or diagnosed as having a HER2-associated cancer through a step of performing an assay on a sample obtained from the subject to determine whether the subject has a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same where the presence of dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, identifies that the subject has a HER2-associated cancer. Some embodiments of any of the methods or uses described herein further include recording in the subject’s clinical record (e.g., a computer readable medium) that the subject is determined to have a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, through the performance of the assay, should be administered a compound of Formula (I), or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof. In some embodiments, the assay utilizes next generation sequencing, pyrosequencing, immunohistochemistry, or break apart FISH analysis. In some embodiments, the assay is a regulatory agency-approved assay, e.g., FDA-approved kit. In some embodiments, the assay is a liquid biopsy.
Also provided is a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, for use in the treatment of a cancer in a subject in need thereof or a subject identified or diagnosed as having a HER2- associated cancer. Also provided is the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating a cancer in a subject identified or diagnosed as having a HER2-associated cancer (. In some embodiments, a subject is identified or diagnosed as having a HER2-associated cancer through the use of a regulatory agency-approved, e.g., FDA-approved, kit for identifying dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, in a subject or a biopsy sample from the subject. As provided herein, a HER2- associated cancer includes those described herein and known in the art.
In some embodiments of any of the methods or uses described herein, the subject has been identified or diagnosed as having a cancer with a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same. In some embodiments of any of the methods or uses described herein, the subject has a tumor that is positive for a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same. In some embodiments of any of the methods or uses described herein, the subject can be a subject with a tumor(s) that is positive for a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same. In some embodiments of any of the methods or uses described herein, the subject can be a subject whose tumors have a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same. In some embodiments of any of the methods or uses described herein, the subject is suspected of having a HER2-associated cancer. In some embodiments, provided herein are methods for treating a HER2-associated cancer in a subject in need of such treatment, the method comprising a) detecting a dysregulation of a HER2 gene, a HER2 kinase, or the expression or activity or level of any of the same in a sample from the subject; and b) administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof. In some embodiments, the dysregulation of a HER2 gene, a HER2 kinase, or the expression or activity or level of any of the same includes one or more HER2 kinase protein point mutations/insertions/deletions. Non-limiting examples of HER2 kinase protein fusions and point mutations/insertions/deletions are described in Tables 3-5. In some embodiments, the HER2 kinase protein point mutations/insertions/deletions are selected from the group consisting of a point mutation at amino acid position 310, 678, 755, 767, 773, 777, or 842 (e.g., S310F, S310Y, R678Q, R678W, R678P, I767M, V773M, V777L, and V842I) and/or an insertion or deletion at amino acid positions 772, 775, 776, 777, and 780 (e.g., Y772_A775dup, A775_G776insYVMA, G776dehnsVC, G776dehnsW, V777_G778insGSP, and P780_Y781insGSP). In some embodiments, the HER2 kinase protein point mutation/insertion/deletion is an exon 20 point mutation/insertion/deletion. In some embodiments, the HER2 exon 20 point mutation/insertion/deletion is a point mutation at amino acid position 773, 776, 777, 779, 780, and 783 (e.g., V773M, G776C, G776V, G776S, V777L, V777M, S779T, P780L, and S783P) and/or an exon 20 insertion/deletion such as an insertion/deletion at amino acid positions 774, 775, 776, 777, 778, and 780. In some embodiments, the HER2 kinase protein insertion is an exon 20 insertion selected from the group consisting of: A775_G776insYVMA,
A775_G776insAVMA, A775_G776insSVMA, A775_G776insVAG, A775insV G776C, A775_G776insI, G776del insVC2, G776del insW, G776del insLC, G776C V777insC, G776C V777insV, V777_G778insCG, G778_S779insCPG, and P780_Y781insGSP. In some embodiments, the HER2 kinase protein mutation/insertion/deletion is an exon 20 insertion/deletion selected from the group consisting of: is Y772_A775dup, A775_G776insYVMA, G776dehnsVC, G776delinsW, V777_G778insGSP, or P780_Y781insGSP. In some embodiments, the cancer with a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same is determined using a regulatory agency-approved, e.g., FDA-approved, assay or kit. In some embodiments, the tumor that is positive for a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same is a tumor positive for one or more HER2 inhibitor resistance mutations. In some embodiments, the tumor with a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same is determined using a regulatory agency-approved, e.g., FDA-approved, assay or kit.
In some embodiments of any of the methods or uses described herein, the subject has a clinical record indicating that the subject has a tumor that has a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same. Also provided are methods of treating a subject that include administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof to a subject having a clinical record that indicates that the subject has a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same.
In some embodiments, the methods provided herein include performing an assay on a sample obtained from the subject to determine whether the subject has a dysregulation of a HER2 gene, a HER2 kinase, or expression or level of any of the same. In some such embodiments, the method also includes administering to a subject determined to have a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity, or level of any of the same a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof. In some embodiments, the method includes determining that a subject has a dysregulation of a HER2 gene, a HER2 kinase, or expression or level of any of the same via an assay performed on a sample obtained from the subject. In such embodiments, the method also includes administering to a subject a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof. In some embodiments, the dysregulation in a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same is one or more point mutation in the HER2 gene (e.g., any of the one or more of the HER2 point mutations described herein). The one or more point mutations in a HER2 gene can result, e.g., in the translation of a HER2 protein having one or more of the following amino acid substitutions: S310F, S310Y, R678Q, R678W, R678P, I767M, V773M, V777L, and V842I. The one or more point mutations in a HER2 gene can result, e.g., in the translation of a HER2 protein having one or more of the following exon 20 ammo acid substitutions: V773M, G776C, G776V, G776S, V777L, V777M, S779T, P780L, and S783P. In some embodiments, the dysregulation in a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same is one or more insertions in the HER2 gene (e.g., any of the one or more of the HER2 insertions described herein). The one or more insertions in a HER2 gene can result, e.g., in the translation of a HER2 protein having one or more of the following exon 20 insertions: M774AYVM, M774del insWLV, A775_G776insYVMA, A775_G776insAVMA, A775_G776insSVMA,
A775_G776insVAG, A775insV G776C, A775_G776insI, G776del insVC2, G776del insW, G776del insLC, G776C V777insC, G776C V777insV, V777_G778insCG, G778_S779insCPG, and P780_Y781insGSP. In some embodiments, the one or more insertions in & HER2 gene can result, e.g., in the translation of a HER2 protein having one or more of the following exon 20 insertions: Y772_A775dup, A775_G776insYVMA, G776delinsVC, G776delinsW, V777_G778insGSP, and P780_Y781insGSP. Some embodiments of these methods further include administering to the subject another anticancer agent (e.g., a second HER2 inhibitor, a second compound of Formula (I), or a pharmaceutically acceptable salt thereof, or immunotherapy).
In some embodiments of any of the methods or uses described herein, an assay used to determine whether the subject has a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same, using a sample from a subject can include, for example, next generation sequencing, immunohistochemistry, fluorescence microscopy, break apart FISH analysis, Southern blotting, Western blotting, FACS analysis, Northern blotting, and PCR-based amplification (e.g., RT-PCR and quantitative real-time RT-PCR). As is well-known in the art, the assays are typically performed, e.g., with at least one labelled nucleic acid probe or at least one labelled antibody or antigenbinding fragment thereof. Assays can utilize other detection methods known in the art for detecting dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or levels of any of the same (see, e.g., the references cited herein). In some embodiments, the sample is a biological sample or a biopsy sample (e.g., a paraffin-embedded biopsy sample) from the subject. In some embodiments, the subject is a subject suspected of having a HER2- associated cancer, a subject having one or more symptoms of a HER2-associated cancer, and/or a subject that has an increased risk of developing a HER2-associated cancer.
In some embodiments, dysregulation of a HER2 gene, a HER2 kinase, or the expression or activity or level of any of the same can be identified using a liquid biopsy (variously referred to as a fluid biopsy or fluid phase biopsy). See, e.g., Karachialiou et al., “Real-time liquid biopsies become a reality in cancer treatment”, Ann. Transl. Med., 3(3): 36, 2016. Liquid biopsy methods can be used to detect total tumor burden and/or the dysregulation of a HER2 gene, a HER2 kinasev, or the expression or activity or level of any of the same. Liquid biopsies can be performed on biological samples obtained relatively easily from a subject (e.g., via a simple blood draw) and are generally less invasive than traditional methods used to detect tumor burden and/or dysregulation of a HER2 gene, a HER2 kinase, or the expression or activity or level of any of the same. In some embodiments, liquid biopsies can be used to detect the presence of dysregulation of a HER2 gene, a HER2 kinase, or the expression or activity or level of any of the same at an earlier stage than traditional methods. In some embodiments, the biological sample to be used in a liquid biopsy can include, blood, plasma, urine, cerebrospinal fluid, saliva, sputum, broncho-alveolar lavage, bile, lymphatic fluid, cyst fluid, stool, ascites, and combinations thereof. In some embodiments, a liquid biopsy can be used to detect circulating tumor cells (CTCs). In some embodiments, a liquid biopsy can be used to detect cell-free DNA. In some embodiments, cell-free DNA detected using a liquid biopsy is circulating tumor DNA (ctDNA) that is derived from tumor cells. Analysis of ctDNA (e.g., using sensitive detection techniques such as, without limitation, next-generation sequencing (NGS), traditional PCR, digital PCR, or microarray analysis) can be used to identify dysregulation of a HER2 gene, a HER2 kinase, or the expression or activity or level of any of the same. Also provided is a method for inhibiting EGFR activity in a cell, comprising contacting the cell with a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof. Also provided is a method for inhibiting HER2 activity in a cell, comprising contacting the cell with a compound of Formula (I), or a pharmaceutically acceptable salt thereof. Further provided herein is a method for inhibiting EGFR and HER2 activity in a cell, comprising contacting the cell with a compound of Formula (I), or a pharmaceutically acceptable salt thereof. In some embodiments, the contacting is in vitro. In some embodiments, the contacting is in vivo. In some embodiments, the contacting is in vivo, wherein the method comprises administering an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof to a subject having a cell having aberrant EGFR activity and/or HER2 activity. In some embodiments, the cell is a cancer cell. In some embodiments, the cancer cell is any cancer as described herein. In some embodiments, the cancer cell is an EGFR-associated cancer cell. In some embodiments, the cancer cell is a HER2-associated cancer cell. As used herein, the term "contacting" refers to the bringing together of indicated moieties in an in vitro system or an in vivo system. For example, "contacting" an EGFR kinase with a compound provided herein includes the administration of a compound provided herein to an individual or subject, such as a human, having an EGFR kinase, as well as, for example, introducing a compound provided herein into a sample containing a cellular or purified preparation containing the EGFR kinase.
Also provided herein is a method of inhibiting cell proliferation, in vitro or in vivo, the method comprising contacting a cell with an effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof as defined herein.
Further provided herein is a method of increase cell death, in vitro or in vivo, the method comprising contacting a cell with an effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof as defined herein. Also provided herein is a method of increasing tumor cell death in a subject. The method comprises administering to the subject an effective compound of Formula (I), or a pharmaceutically acceptable salt thereof, in an amount effective to increase tumor cell death. The phrase "therapeutically effective amount" means an amount of compound that, when administered to a subject in need of such treatment, is sufficient to (i) treat an EGFR kinase-associated disease or disorder or a HER2 kinase-associated disease or disorder, (ii) attenuate, ameliorate, or eliminate one or more symptoms of the particular disease, condition, or disorder, or (iii) delay the onset of one or more symptoms of the particular disease, condition, or disorder described herein. The amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof that will correspond to such an amount will vary depending upon factors such as the particular compound, disease condition and its severity, the identity (e.g., weight) of the subject in need of treatment, but can nevertheless be routinely determined by one skilled in the art.
When employed as pharmaceuticals, the compounds of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), including pharmaceutically acceptable salts or solvates thereof, can be administered in the form of pharmaceutical compositions as described herein.
Also provided herein is a method of treating a subject having a cancer, wherein the method comprises:
(a) determining that a cancer cell in a sample obtained from a subject having a cancer and previously administered one or more doses of a first EGFR inhibitor has one or more EGFR inhibitor resistance mutations that confer increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor that was previously administered to the subject; and
(b) administering a therapeutically effective amount of a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, as a monotherapy or in conjunction with another anti cancer agent to the subject.
Further provided herein is a method of treating a subject having a cancer, wherein the method comprises:
(a) determining that a cancer cell in a sample obtained from a subject having a cancer and previously administered one or more doses of a first EGFR inhibitor does not have one or more EGFR inhibitor resistance mutations that confer increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor that was previously administered to the subject; and
(b) administering additional doses of the first EGFR inhibitor to the subject.
Combinations
In the field of medical oncology it is normal practice to use a combination of different forms of treatment to treat each subject with cancer. In medical oncology the other component(s) of such conjoint treatment or therapy in addition to compositions provided herein may be, for example, surgery, radiotherapy, and chemotherapeutic agents, such as other kinase inhibitors, signal transduction inhibitors and/or monoclonal antibodies. For example, a surgery may be open surgery or minimally invasive surgery. Compounds of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or pharmaceutically acceptable salts or solvates thereof therefore may also be useful as adjuvants to cancer treatment, that is, they can be used in combination with one or more additional therapies or therapeutic agents, for example, a chemotherapeutic agent that works by the same or by a different mechanism of action. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can be used prior to administration of an additional therapeutic agent or additional therapy. For example, a subject in need thereof can be administered one or more doses of a compound of Formula (I), or a pharmaceutically acceptable salt thereof for a period of time and then undergo at least partial resection of the tumor. In some embodiments, the treatment with one or more doses of a compound of Formula (I), or a pharmaceutically acceptable salt thereof reduces the size of the tumor (e.g., the tumor burden) prior to the at least partial resection of the tumor. In some embodiments, a subject in need thereof can be administered one or more doses of a compound of Formula (I), or a pharmaceutically acceptable salt thereof for a period of time and under one or more rounds of radiation therapy. In some embodiments, the treatment with one or more doses of a compound of Formula (I), or a pharmaceutically acceptable salt thereof reduces the size of the tumor (e.g., the tumor burden) prior to the one or more rounds of radiation therapy.
In some embodiments, a subject has a cancer (e.g., a locally advanced or metastatic tumor) that is refractory or intolerant to standard therapy (e.g., administration of a chemotherapeutic agent, such as a first EGFR inhibitor, a first HER2 inhibitor, or a multikinase inhibitor, immunotherapy, or radiation (e.g., radioactive iodine)). In some embodiments, a subject has a cancer (e.g., a locally advanced or metastatic tumor) that is refractory or intolerant to prior therapy (e.g., administration of a chemotherapeutic agent, such as a first EGFR inhibitor, a first HER2 inhibitor, or a multi-kinase inhibitor, immunotherapy, or radiation (e.g., radioactive iodine)). In some embodiments, a subject has a cancer (e.g., a locally advanced or metastatic tumor) that has no standard therapy. In some embodiments, a subject is EGFR inhibitor naive. For example, the subject is naive to treatment with a selective EGFR inhibitor. In some embodiments, a subject is not EGFR inhibitor naive. In some embodiments, a subject is HER2 inhibitor naive. For example, the subject is naive to treatment with a selective HER2 inhibitor. In some embodiments, a subject is not HER2 inhibitor naive. In some embodiments, a subject has undergone prior therapy. For example, treatment with a multi-kinase inhibitor (MKI), an EGFR tyrosine kinase inhibitor (TKI), osimertinib, gefitinib, erlotinib, afatinib, lapatinib, neratinib, AZD- 9291, CL-387785, CO-1686, or WZ4002.
In some embodiments of any the methods described herein, the compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)) (or a pharmaceutically acceptable salt thereof) is administered in combination with a therapeutically effective amount of at least one additional therapeutic agent selected from one or more additional therapies or therapeutic (e.g., chemotherapeutic) agents.
Non-limiting examples of additional therapeutic agents include: other EGFR- targeted therapeutic agents (i.e., a first or second EGFR inhibitor), other HER2-targeted therapeutic agents (i.e., a first or second HERZ inhibitor), RAS pathway targeted therapeutic agents, PARP inhibitors, other kinase inhibitors (e.g., receptor tyrosine kinase- targeted therapeutic agents (e.g., Trk inhibitors or multi-kinase inhibitors)), farnesyl transferase inhibitors, signal transduction pathway inhibitors, checkpoint inhibitors, modulators of the apoptosis pathway (e.g., obataclax); cytotoxic chemotherapeutics, angiogenesis-targeted therapies, immune-targeted agents, including immunotherapy, and radiotherapy.
In some embodiments, the other EGFR-targeted therapeutic is a multi-kinase inhibitor exhibiting EGFR inhibition activity. In some embodiments, the other EGFR- targeted therapeutic inhibitor is selective for an EGFR kinase.
Non-limiting examples of EGFR-targeted therapeutic agents (e.g., a first EGFR inhibitor or a second EGFR inhibitor) include an EGFR- selective inhibitor, a panHER inhibitor, and an anti-EGFR antibody. In some embodiments, the EGFR inhibitor is a covalent inhibitor. In some embodiments, the EGFR-targeted therapeutic agent is osimertinib (AZD9291, merelectinib, TAGRISSOTM), erlotinib (TARCEVA®), gefitinib (IRESSA®), cetuximab (ERBITUX®), necitumumab (PORTRAZZATM, IMC-11F8), neratinib (HKI-272, NERLYNX®), lapatinib (TYKERB®), panitumumab (ABX-EGF, VECTIBIX®), vandetanib (CAPRELSA®), rociletinib (CO- 1686), olmutinib (OLITATM, HM61713, BI-1482694), naquotimb (ASP8273), nazartimb (EGF816, NVS- 816), PF-06747775, icotimb (BPI-2009H), afatimb (BIBW 2992, GILOTRIF®), dacomitimb (PF-00299804, PF-804, PF-299, PF-299804), avitimb (AC0010), AC0010MA EAI045, matuzumab (EMD-7200), nimotuzumab (h-R3, BIOMAb EGFR®), zalutumab, MDX447, depatuxizumab (humanized mAb 806, ABT-806), depatuxizumab mafodotin (ABT-414), ABT-806, mAb 806, canertinib (CI-1033), shikonin, shikonin derivatives (e.g., deoxyshikonin, isobutyrylshikonin, acetylshikonin, P,P-dimethylacrylshikonin and acetylalkannin), poziotinib (NOV120101, HM781-36B), AV-412, ibrutinib, WZ4002, brigatinib (AP26113, ALUNBRIG®), pelitinib (EKB-569), tarloxotinib (TH-4000, PR610), BPI- 15086, Hemay022, ZN-e4, tesevatimb (KD019, XL647), YH25448, epitimb (HMPL-813), CK-101, MM-151, AZD3759, ZD6474, PF-06459988, varlintimb (ASLAN001, ARRY-334543), AP32788, HLX07, D-0316, AEE788, HS-10296, avitimb, GW572016, pyrotimb (SHR1258), SCT200, CPGJ602, Sym004, MAb-425, Modotuximab (TAB-H49), futuximab (992 DS), zalutumumab, KL-140, RO5083945, IMGN289, JNJ- 61186372, LY3164530, Sym013, AMG 595, BDTX-189, avatimb, Disruptin, CL-387785, EGFRBi- Armed Autologous T Cells, and EGFR CAR-T Therapy. In some embodiments, the EGFR-targeted therapeutic agent is selected from osimertinib, gefitinib, erlotinib, afatinib, lapatinib, neratinib, AZD-9291, CL-387785, CO-1686, or WZ4002.
Additional EGFR-targeted therapeutic agents (e.g., a first EGFR inhibitor or a second EGFR inhibitor) include those disclosed in WO 2019/246541; WO 2019/165385; WO 2014/176475; and US 9,029,502, each of which is incorporated by reference in its entirety. In some embodiments, the other HER2 -targeted therapeutic is a multi-kinase inhibitor exhibiting HER2 inhibition activity. In some embodiments, the other HER2- targeted therapeutic inhibitor is selective for a HER2 kinase.
Non-limiting examples of HER2-targeted therapeutic agents (e.g., a first HER2 inhibitor or a second HER2 inhibitor) include a HER2-selective inhibitor, a panHER inhibitor, and an anti-HER2 antibody. Exemplary HER2-targeted therapeutic agents include trastuzumab (e.g., TRAZIMERA™, HERCEPTIN®), pertuzumab (e.g., PERJETA®), trastuzumab emtansine (T-DM1 or ado-trastuzumab emtansine, e.g., KADCYLA®), lapatinib, KU004, neratinib (e.g., NERLYNX®), dacomitinib (e.g., VIZIMPRO®), afatimb (GILOTRIF®), tucatimb (e.g., TUKYSA™), erlotinib (e.g., TARCEVA®), pyrotimb, poziotimb, CP-724714, CUDC-101, sapitimb (AZD8931), tanespimycin (17-AAG), IPI-504, PF299, pelitimb, S- 22261 1, and AEE-788.
Additional HER2-targeted therapeutic agents (e.g., a first HERZ inhibitor or a second HERZ inhibitor) include those disclosed in WO 2019/246541; WO 2019/165385; WO 2014/176475; and US 9,029,502, each of which is incorporated by reference in its entirety.
A “RAS pathway targeted therapeutic agent” as used herein includes any compound exhibiting inactivation activity of any protein in a RAS pathway (e.g., kinase inhibition, allosteric inhibition, inhibition of dimerization, and induction of degradation). Nonlimiting examples of a protein in a RAS pathway include any one of the proteins in the RAS-RAF-MAPK pathway or PI3K/AKT pathway such as RAS (e.g., KRAS, HRAS, and NRAS), RAF, BRAF, MEK, ERK, PI3K, A KT, and mTOR. In some embodiments, a RAS pathway modulator can be selective for a protein in a RAS pathway, e.g., the RAS pathway modulator can be selective for RAS (also referred to as a RAS modulator). In some embodiments, a RAS modulator is a covalent inhibitor. In some embodiments, a RAS pathway targeted therapeutic agent is a “KRAS pathway modulator.” A KRAS pathway modulator includes any compound exhibiting inactivation activity of any protein in a KRAS pathway (e.g., kinase inhibition, allosteric inhibition, inhibition of dimerization, and induction of degradation). Non-limiting examples of a protein in a KRAS pathway include any one of the proteins in the KRAS -RAF -MAPK pathway or PI3K/AKT pathway such as KRAS, RAF, BRAF, MEK, ERK, PI3K, AKT, and mTOR. In some embodiments, a KRAS pathway modulator can be selective for a protein in a RAS pathway, e.g., the KRAS pathway modulator can be selective for KRAS (also referred to as a KRAS modulator). In some embodiments, a KRAS modulator is a covalent inhibitor. Non-limiting examples of a KRAS-targeted therapeutic agents (e.g., KRAS inhibitors) include BI 1701963, AMG 510, ARS-3248, ARS1620, AZD4785, SML-8-73-1, SML-10-70-1, VSA9, AA12, and MRTX-849.
Further non-limiting examples of RAS-targeted therapeutic agents include BRAF inhibitors, MEK inhibitors, ERK inhibitors, PI3K inhibitors, AKT inhibitors, and mTOR inhibitors. In some embodiments, the BRAF inhibitor is vemurafenib (ZELBORAF®), dabrafemb (TAFINLAR®), and encorafemb (BRAFTOVITM), BMS-908662 (XL281), sorafenib, LGX818, PLX3603, RAF265, RO5185426, GSK2118436, ARQ 736, GDC- 0879, PLX-4720, AZ304, PLX-8394, HM95573, RO5126766, LXH254, or a combination thereof.
In some embodiments, the MEK inhibitor is trametinib (MEKINIST®, GSK1120212), cobimetimb (COTELLIC®), bimmetimb (MEKTOVI®, MEK162), selumetimb (AZD6244), PD0325901, MSC1936369B, SHR7390, TAK-733, RO5126766, CS3006, WX-554, PD98059, CI1040 (PD 184352), hypothemycin, or a combination thereof.
In some embodiments, the ERK inhibitor is FRI-20 (ON-01060), VTX-l le, 25- OH-D3-3-BE (B3CD, bromoacetoxycalcidiol), FR-180204, AEZ-131 (AEZS-131), AEZS-136, AZ-13767370, BL-EI-001, LY-3214996, LTT-462, KO-947, KO-947, MK- 8353 (SCH900353), SCH772984, uhxertmib (BVD-523), CC-90003, GDC-0994 (RG- 7482), ASN007, FR148083, 5-7-Oxozeaenol, 5-iodotubercidin, GDC0994, ONC201, or a combination thereof.
In some embodiments, PI3K inhibitor is selected from buparlisib (BKM120), alpelisib (BYL719), WX-037, copanhsib (ALIQOPATM, BAY80-6946), dactohsib (NVP-BEZ235, BEZ-235), tasehsib (GDC-0032, RG7604), sonolisib (PX-866), CUDC- 907, PQR309, ZSTK474, SF1126, AZD8835, GDC-0077, ASN003, pictihsib (GDC- 0941), pilaralisib (XL147, SAR245408), gedatolisib (PF-05212384, PKI-587), serabelisib (TAK-117, MLN1117, INK 1117), BGT-226 (NVP-BGT226), PF-04691502, apitolisib (GDC-0980), omipalisib (GSK2126458, GSK458), voxtahsib (XL756, SAR245409), AMG 511, CH5132799, GSK1059615, GDC-0084 (RG7666), VS-5584 (SB2343), PKI- 402, wortmanmn, LY294002, PI-103, ngosertib, XL-765, LY2023414, SAR260301, KIN- 193 (AZD-6428), GS-9820, AMG319, GSK2636771, or a combination thereof.
In some embodiments, the AKT inhibitor is selected from miltefosine (IMPADIVO®), wortmanmn, NL-71-101, H-89, GSK690693, CCT128930, AZD5363, ipatasertib (GDC-0068, RG7440), A-674563, A-443654, AT7867, AT13148, uprosertib, afuresertib, DC 120, 2- [4-(2-aminoprop-2-yl)phenyl] -3 -phenylquinoxaline, MK-2206, edelfosine, miltefosine, perifosine, erucylphophocholine, erufosine, SRI 3668, OSU-A9, PH-316, PHT-427, PIT-1, DM-PIT-1, triciribine (Triciribine Phosphate Monohydrate), API-1, N-(4-(5-(3-acetamidophenyl)-2-(2-aminopyridin-3-yl)-3H-imidazo[4,5-b] pyridin- 3-yl)benzyl)-3-fluorobenzamide, ARQ092, BAY 1125976, 3-oxo-tirucallic acid, lactoquinomycin, boc-Phe-vinyl ketone, Perifosine (D-21266), TCN, TCN-P, GSK2141795, ONC201, or a combination thereof.
In some embodiments, the mTOR inhibitor is selected from MLN0128, AZD-2014, CC-223, AZD2014, CC-115, everohmus (RAD001), temsirolimus (CCI-779), ridaforolimus (AP-23573), sirolimus (rapamycin), or a combination thereof.
Non-limiting examples of farnesyl transferase inhibitors include lonafarnib, tipifarmb, BMS-214662, L778123, L744832, and FTI-277.
In some embodiments, a chemotherapeutic agent includes an anthracycline, cyclophosphamide, a taxane, a platinum-based agent, mitomycin, gemcitabine, eribulin (HALAVEN™), or combinations thereof.
Non-limiting examples of a taxane include paclitaxel, docetaxel, abraxane, and taxotere.
In some embodiments, the anthracycline is selected from daunorubicin, doxorubicin, epirubicin, idarubicin, and combinations thereof.
In some embodiments, the platinum-based agent is selected from carboplatin, cisplatin, oxaliplatin, nedplatin, triplatin tetranitrate, phenanthriplatin, picoplatin, satraplatin and combinations thereof
Non-limiting examples of PARP inhibitors include olaparib (LYNPARZA®), talazoparib, rucaparib, niraparib, veliparib, BGB-290 (pamiparib), CEP 9722, E7016, impanb, IMP4297, NOV1401, 2X-121, ABT-767, RBN-2397, BMN 673, KU-0059436 (AZD2281), BSI-201, PF-01367338, INO-lOOl, and JPI-289.
Non-limiting examples of immunotherapy include immune checkpoint therapies, atezolizumab (TECENTRIQ®), albumin-bound paclitaxel. Non-limiting examples of immune checkpoint therapies include inhibitors that target CTLA-4, PD-1, PD-L1, BTLA, LAG-3, A2AR, TIM-3, B7-H3, VISTA, IDO, and combinations thereof. In some embodimetnts the CTLA-4 inhibitor is ipilimumab (YERVOY®). In some embodiments, the PD-1 inhibitor is selected from pembrolizumab (KEYTRUDA®), nivolumab (OPDIVO®), cemiplimab (LIBTAYO®), or combinations thereof. In some embodiments, the PD-L1 inhibitor is selected from atezolizumab (TECENTRIQ®), avelumab (BAVENCIO®), durvalumab (IMFINZI®), or combinations thereof. In some embodiments, the LAG-3 inhibitor is IMP701 (LAG525). In some embodiments, the A2AR inhibitor is CPI-444. In some embodiments, the TIM-3 inhibitor is MBG453. In some embodiments, the B7-H3 inhibitor is enoblituzumab. In some embodiments, the VISTA inhibitor is JNJ-61610588. In some embodiments, the IDO inhibitor is indoximod. See, for example, Marin- Acevedo, et al., J Hematol Oncol. 11: 39 (2018).
In some embodiments, the additional therapy or therapeutic agent is a combination of atezolizumab and nab-paclitaxel.
Accordingly, also provided herein is a method of treating cancer, comprising administering to a subject in need thereof a pharmaceutical combination for treating cancer which comprises (a) a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, (b) an additional therapeutic agent, and (c) optionally at least one pharmaceutically acceptable carrier for simultaneous, separate or sequential use for the treatment of cancer, wherein the amounts of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, and the additional therapeutic agent are together effective in treating the cancer.
In some embodiments, the additional therapeutic agent(s) includes any one of the above listed therapies or therapeutic agents which are standards of care in cancers wherein the cancer has a dysregulation of an EGFR gene, an EGFR protein, or expression or activity, or level of any of the same.
In some embodiments, the additional therapeutic agent(s) includes any one of the above listed therapies or therapeutic agents which are standards of care in cancers wherein the cancer has a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity, or level of any of the same.
These additional therapeutic agents may be administered with one or more doses of the compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, or pharmaceutical composition thereof, as part of the same or separate dosage forms, via the same or different routes of administration, and/or on the same or different administration schedules according to standard pharmaceutical practice known to one skilled in the art.
Also provided herein is (i) a pharmaceutical combination for treating a cancer in a subject in need thereof, which comprises (a) a compound of Formula (I) (e.g., Formula (I- a), (I-b), (I-c), (I-d) or (I-e)), or a pharmaceutically acceptable salt thereof, (b) at least one additional therapeutic agent (e.g., any of the exemplary additional therapeutic agents described herein or known in the art), and (c) optionally at least one pharmaceutically acceptable carrier for simultaneous, separate or sequential use for the treatment of cancer, wherein the amounts of the compound of Formula (I), or pharmaceutically acceptable salt thereof, and of the additional therapeutic agent are together effective in treating the cancer; (ii) a pharmaceutical composition comprising such a combination; (iii) the use of such a combination for the preparation of a medicament for the treatment of cancer; and (iv) a commercial package or product comprising such a combination as a combined preparation for simultaneous, separate or sequential use; and to a method of treatment of cancer in a subject in need thereof. In some embodiments, the cancer is an EGFR-associated cancer. For example, an EGFR-associated cancer having one or more EGFR inhibitor resistance mutations. In some embodiments, the cancer is a HER2-associated cancer. For example, a HER2-associated cancer having one or more HER2 inhibitor resistance mutations.
The term "pharmaceutical combination", as used herein, refers to a pharmaceutical therapy resulting from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients. The term "fixed combination" means that a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I- d) or (I-e)), or a pharmaceutically acceptable salt thereof, and at least one additional therapeutic agent (e.g., a chemotherapeutic agent), are both administered to a subject simultaneously in the form of a single composition or dosage. The term "non-fixed combination" means that a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and at least one additional therapeutic agent (e.g., chemotherapeutic agent) are formulated as separate compositions or dosages such that they may be administered to a subject in need thereof simultaneously, concurrently or sequentially with variable intervening time limits, wherein such administration provides effective levels of the two or more compounds in the body of the subject. These also apply to cocktail therapies, e.g., the administration of three or more active ingredients
Accordingly, also provided herein is a method of treating a cancer, comprising administering to a subject in need thereof a pharmaceutical combination for treating cancer which comprises (a) a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e)), or pharmaceutically acceptable salt thereof, and (b) an additional therapeutic agent, wherein the compound of Formula (I) and the additional therapeutic agent are administered simultaneously, separately or sequentially, wherein the amounts of the compound of Formula (I), or pharmaceutically acceptable salt thereof, and the additional therapeutic agent are together effective in treating the cancer. In some embodiments, the compound of Formula (I), or pharmaceutically acceptable salt thereof, and the additional therapeutic agent are administered simultaneously as separate dosages. In some embodiments, the compound of Formula (I), or pharmaceutically acceptable salt thereof, and the additional therapeutic agent are administered as separate dosages sequentially in any order, in jointly therapeutically effective amounts, e.g., in daily or intermittently dosages. In some embodiments, the compound of Formula (I), or pharmaceutically acceptable salt thereof, and the additional therapeutic agent are administered simultaneously as a combined dosage. In some embodiments, the cancer is an EGFR-associated cancer. For example, an EGFR-associated cancer having one or more EGFR inhibitor resistance mutations. In some embodiments, the cancer is a HER2-associated cancer. For example, a HER2-associated cancer having one or more HER2 inhibitor resistance mutations.
In some embodiments, the presence of one or more EGFR inhibitor resistance mutations in a tumor causes the tumor to be more resistant to treatment with a first EGFR inhibitor. Methods useful when an EGFR inhibitor resistance mutation causes the tumor to be more resistant to treatment with a first EGFR inhibitor are described below. For example, provided herein are methods of treating a subject having a cancer that include: identifying a subject having a cancer cell that has one or more EGFR inhibitor resistance mutations; and administering to the identified subject a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof. In some embodiments, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with the first EGFR inhibitor. Also provided are methods of treating a subject identified as having a cancer cell that has one or more EGFR inhibitor resistance mutations that include administering to the subject a compound of Formula (I), or a pharmaceutically acceptable salt thereof. In some embodiments, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with the first EGFR inhibitor. In some embodiments, the one or more EGFR inhibitor resistance mutations confer increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor. In some embodiments, the one or more EGFR inhibitor resistance mutations include one or more EGFR inhibitor resistance mutations listed in Table 2a and 2b. For example, the one or more EGFR inhibitor resistance mutations can include a substitution at amino acid position 718, 747, 761, 790, 797, or 854 (e.g., L718Q, L747S, D761Y, T790M, C797S, and T854A).
For example, provided herein are methods for treating an EGFR-associated cancer in a subject in need of such treatment, the method comprising (a) detecting a dysregulation of an EGFR gene, an EGFR kinase, or the expression or activity or level of any of the same in a sample from the subject; and (b) administering to the subject a therapeutically effective amount of a first EGFR inhibitor, wherein the first EGFR inhibitor is selected from the group consisting of osimertinib, gefitinib, erlotinib, afatinib, lapatinib, neratinib, AZD- 9291, CL-387785, CO-1686, or WZ4002. In some embodiments, the methods further comprise (after (b)) (c) determining whether a cancer cell in a sample obtained from the subject has at least one EGFR inhibitor resistance mutation; and (d) administering a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof, as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has been determined to have a cancer cell that has at least one EGFR inhibitor resistance mutation; or (e) administering additional doses of the first EGFR inhibitor of step (b) to the subject if the subject has not been determined to have a cancer cell that has at least one EGFR inhibitor resistance mutation.
Methods useful when a HER2 activating mutation is present in a tumor are described herein. For example, provided herein are methods of treating a subject having a cancer that include: identifying a subject having a cancer cell that has one or more HER2 activating mutations; and administering to the identified subject a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I-e))> or a pharmaceutically acceptable salt thereof. Also provided are methods of treating a subject identified as having a cancer that has one or more HER2 activating mutations that include administering to the subject a compound of Formula (I), or a pharmaceutically acceptable salt thereof. In some embodiments, the one or more HER2 activating mutations include one or more HER2 activating mutations listed in Tables 3-5.
Methods useful when an activating mutation (e.g., HER2 activating mutation) is present in a tumor in a subject are described herein. For example, provided herein are methods of treating a subject having a cancer that include: identifying a subject having a cancer cell that has one or more HER2 activating mutations; and administering to the identified subject a compound of Formula (I) (e.g., Formula (I-a), (I-b), (I-c), (I-d) or (I- e)), or a pharmaceutically acceptable salt thereof.
Compound Preparation
The compounds disclosed herein can be prepared in a variety of ways using commercially available starting materials, compounds known in the literature, or from readily prepared intermediates, by employing standard synthetic methods and procedures either known to those skilled in the art, or in light of the teachings herein. The synthesis of the compounds disclosed herein can be achieved by generally following Scheme 1, with modification for specific desired substituents.
Standard synthetic methods and procedures for the preparation of organic molecules and functional group transformations and manipulations can be obtained from the relevant scientific literature or from standard textbooks in the field. Although not limited to any one or several sources, classic texts such as R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); Smith, M. B., March, J., March' s Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5th edition, John Wiley & Sons: New York, 2001 ; and Greene, T.W., Wuts, P.G. M., Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons: New York, 1999, are useful and recognized reference textbooks of organic synthesis known to those in the art. The following descriptions of synthetic methods are designed to illustrate, but not to limit, general procedures for the preparation of compounds of the present disclosure.
The synthetic processes disclosed herein can tolerate a wide variety of functional groups; therefore, various substituted starting materials can be used. The processes generally provide the desired final compound at or near the end of the overall process, although it may be desirable in certain instances to further convert the compound to a pharmaceutically acceptable salt thereof.
Example 1. Synthesis of 3- [(3-chloro-2-meth oxyphenyl) amino]-2-[2- methoxypyrido[3,2-d]pyrimidin-8-yl]-l,5,6,7-tetrahydroindol-4-one (Compound
143)
Figure imgf000226_0001
To a stirred solution of l,5,6,7-tetrahydroindol-4-one (1.00 g, 7.40 mmol, 1.00 equiv), bis(pinacolato)diboron (2.82 g, 11.10 mmol, 1.50 equiv) and dbbpy (0.12 g, 0.44 mmol, 0.06 equiv) in MTBE (20.00 mL) was added Ir[(COD)OMe]2Ch (0.15 g, 0.22 mmol, 0.03 equiv) in portions at room temperature under argon atmosphere. The resulting mixture was stirred for 3 h at 50 degrees C under argon atmosphere. Desired product could be detected by LCMS. The precipitated solids were collected by filtration and washed with MTBE (3 x 15 mL). The crude product was used in the next step directly without further purification. This resulted in 2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-l,5,6,7-tetrahydroindol- 4-one (1.60 g, 82.82%) as a light pink solid.
LC-MS: M+H found: 262.05.
Figure imgf000227_0001
To a stirred solution of 8-bromo-2-methoxypyrido[3,2-d] pyrimidine (100 mg, 0.417 mmol, 1.00 equiv), 2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-l,5,6,7- tetrahydroindol-4-one (130.53 mg, 0.500 mmol, 1.2 equiv) in dioxane (5 mL) and H2O (1 mL) was added Pd(PPhs)4 (48.14 mg, 0.042 mmol, 0.1 equiv) and NaiCCh (132.45 mg, 1.251 mmol, 3 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for overnight at 50 degrees C under nitrogen atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with CH2G2 / MeOH (15:1) to afford2-[2-methoxypyrido[3,2-d] pyrimidin-8-yl]-l, 5,6,7- tetrahydroindol-4-one (90 mg, 73.41%) as a yellow solid.
LC-MS: M+H found: 295.10.
Figure imgf000227_0002
To a stirred solution of 2-[2-methoxypyrido[3,2-d] pyrimidin-8-yl]-l, 5,6,7- tetrahydroindol-4-one (90.00 mg, 0.306 mmol, 1.00 equiv) in DMF (2.00 mL) was added NIS (82.56 mg, 0.367 mmol, 1.20 equiv) in portions at room temperature under air atmosphere. The resulting mixture was stirred for 3 h at room temperature under air atmosphere. Desired product could be detected by LCMS. The reaction was quenched with NaiSOdaq) at 0 degrees C. The precipitated solids were collected by filtration and washed with water (3x5 mL). The resulting solid was purified by reverse flash chromatography with the following conditions: column, silica gel; mobile phase, MeCN in water, 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 3-iodo-2-[2- methoxypyrido[3,2-d] pyrimidin-8-yl]-l,5,6,7-tetrahydroindol-4-one (120 mg, 93.39%) as a yellow solid.
LC-MS: M+H found: 420.90.
Figure imgf000228_0001
Compound 143
To a stirred solution of 3-iodo-2-[2-methoxypyrido[3,2-d] pyrimidin-8-yl]-l, 5,6,7- tetrahydroindol-4-one (100.00 mg, 0.238 mmol, 1.00 equiv) and 3-chloro-2- methoxyaniline (37.51 mg, 0.238 mmol, 1 equiv) in DMF (3 mL) was added Ephos Pd G4 (43.72 mg, 0.048 mmol, 0.2 equiv) and CS2CO3 (232.61 mg, 0.714 mmol, 3 equiv) in portions at room temperature under argon atmosphere. The resulting mixture was stirred for 3 h at 50 degrees C under argon atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under vacuum. The crude product (80 mg) was purified by Prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column, 30*150 mm, 5pm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 41% B to 71% B in 7 min; Wave Length: 254 nm; RTl(min): 6.5) to afford 3-[(3-chloro-2-methoxyphenyl) amino]-2-[2- methoxypyrido[3,2-d]pyrimidin-8-yl]-l,5,6,7-tetrahydroindol-4-one (24.6 mg, 22.82%) as a yellow solid.
LC-MS: M+H found: 450.00.
'H NMR (400 MHz, DMSO-r/6) δ 12.06 (s, 1H), 9.53 (s, 1H), 8.76 (d, J = 4.7 Hz, 1H), 7.92 (s, 1H), 7.63 (d, J= 4.7 Hz, 1H), 6.76 - 6.57 (m, 2H), 6.14 (dd, J= 7.8, 1.9 Hz, 1H), 4.20 (s, 3H), 3.85 (s, 3H), 2.97 (t, J= 6.1 Hz, 2H), 2.46 - 2.36 (m, 2H), 2.12 (q, J= 6.2 Hz, 2H). Example 2. Synthesis of 3- [(3-chloro-2-meth oxyphenyl) amino]-2-(6,7-dimethoxy-l,5- naphthyridin-4-yl)-l,5,6,7-tetrahydroindol-4-one (Compound 144)
Figure imgf000229_0001
To a stirred solution of 8-bromo-2, 3 -dimethoxy- 1,5-naphthyridine (150.00 mg, 0.56 mmol, 1.00 equiv) and 2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH,5H,6H,7H- pyrrolo[3,2-c] pyridin-4-one (175.33 mg, 0.67 mmol, 1.20 equiv) in dioxane (5.00 mb) and H2O (1.00 mL) were added Pd(PPhs)4 (64.41 mg, 0.06 mmol, 0.10 equiv) and NaiCCh (177.24 mg, 1.67 mmol, 3.00 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 4 h at 50 degrees C under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by silica gel column chromatography, eluted with CH2G2 / MeOH (15: 1) to afford 2-(6,7-dimethoxy-l,5- naphthyridin-4-yl)-l ,5,6,7-tetrahydroindol-4-one (140 mg, 77.67%) as a light yellow solid. LC-MS: M+H found: 324.05.
Figure imgf000229_0002
To a stirred solution of 2-(6,7-dimethoxy-l,5-naphthyridin-4-yl)-l,5,6,7-tetrahydroindol- 4-one (140 mg, 0.433 mmol, 1.00 equiv) inDMF (3 mL) was added NIS (116.89 mg, 0.520 mmol, 1.2 equiv) in portions at room temperature under air atmosphere. The resulting mixture was stirred for overnight at room temperature under air atmosphere. Desired product could be detected by LCMS. The reaction was quenched by the addition of Na2SCh (aq.) (3 mL) at 0 degrees C. The precipitated solids were collected by filtration and washed with water (3x5 mL). The resulting solid was purified by silica gel column chromatography, eluted with CH2G2 / MeOH (10: 1) to afford 2-(6, 7-dimethoxy-l, 5- naphthyridin-4-yl)-3-iodo-l,5,6,7-tetrahydroindol-4-one (120 mg, 61.69%) as a yellow solid.
LC-MS: M+H found: 449.95.
Figure imgf000230_0001
Compound 144
To a stirred solution of 2-(6, 7-dimethoxy-l, 5-naphthyridin-4-yl)-3-iodo-l, 5,6,7- tetrahydroindol-4-one (120.00 mg, 0.267 mmol, 1.00 equiv) and 3-chloro-2- methoxyaniline (42.10 mg, 0.267 mmol, 1 equiv) in DMF (3 mL) were added Ephos Pd G4 (49.07 mg, 0.053 mmol, 0.2 equiv) and CS2CO3 (174.06 mg, 0.534 mmol, 2 equiv) in portions at room temperature under argon atmosphere. The resulting mixture was stirred for 3 h at 50 degrees C under argon atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under vacuum. The residue was purified by reverse flash chromatography with the following conditions: column, silica gel; mobile phase, MeCN in water, 10% to 50% gradient in 10 min; detector, UV 254 nm. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge Prep Cl 8 OBD Column, 30*100 mm, 5pm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 38% B to 63% B in 9 min; Wave Length: 254/220 nm; RTl(min): 8.85) to afford 3-[(3-chloro-2-methoxyphenyl) amino]-2- (6, 7-dimethoxy-l, 5-naphthyridin-4-yl)-l, 5, 6, 7-tetrahydroindol-4-one (23.6 mg, 22.03%) as a yellow solid.
LC-MS: M+H found: 479.00.
1H NMR (400 MHz, DMSO-r/6) δ 12.03 (s, 1H), 8.56 (d, J = 4.9 Hz, 1H), 7.70 (d, J= 14.3 Hz, 2H), 7.42 (d, J= 4.9 Hz, 1H), 6.69 - 6.60 (m, 2H), 6.15 (dd, J= 6.7, 3.0 Hz, 1H), 4.18 (s, 3H), 4.00 (s, 3H), 3.80 (s, 3H), 2.94 (t, J= 6.1 Hz, 2H), 2.45 - 2.36 (m, 2H), 2.11 (p, J = 6.3 Hz, 2H).
Example 3. Synthesis of 3-[(3-chloro-2-methoxyphenyl)amino]-2-[3-(2-methoxy-2- methylpropoxy)pyridin-4-yl]-l,5,6,7-tetrahydroindol-4-one (compound 161)
Figure imgf000231_0001
To a stirred solution of 3-chloro-2-methoxyaniline (800 mg, 5.076 mmol, 1 equiv) inDCM (8 mL) were added NaHCO3 (2132.13 mg, 25.380 mmol, 5 equiv) and H2O (8 mL) in portions at 0 degrees C under N2 atmosphere. To the above mixture was added thiophosgene (758.69 mg, 6.599 mmol, 1.3 equiv) dropwise over 5 minutes at 0 degrees C. The resulting mixture was stirred for additional 16 hours at RT. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE (100%) to afford 1 -chi oro-3 -isothiocyanato-2- methoxybenzene (900 mg, 88.81%) as a off-white oil.
Figure imgf000231_0002
To a stirred solution of l-chloro-3-isothiocyanato-2-methoxybenzene (500 mg, 2.504 mmol, 1 equiv) and 1,3 cyclohexanedione (336.97 mg, 3.005 mmol, 1.2 equiv) in MeCN (5 mL) was added DBU (571.90 mg, 3.756 mmol, 1.5 equiv) dropwise at 0 degrees C under N2 atmosphere. The resulting mixture was stirred for 16 hours at RT under N2 atmosphere. The resulting mixture was concentrated under vacuum to afford N-(3-chloro-2- methoxyphenyl)-2-hydroxy-6-oxocyclohex-l-ene-l -carbothioamide (700 mg, 89.65%) as a light yellow solid. LC-MS: M+H found: 209.10.
Figure imgf000232_0001
To a stirred solution of N-(3-chloro-2-methoxyphenyl)-2-hydroxy-6-oxocyclohex-l-ene- 1 -carbothioamide (500.00 mg, 1.604 mmol, 1.00 equiv) in DMA (5.00 mL) was added 1- [3-(2-methoxy-2-methylpropoxy)pyridin-4-yl]methanamine (404.66 mg, 1.924 mmol, 1.20 equiv) in portions at N2 atmosphere. The resulting mixture was stirred for 2 hours at 120 degrees C. The aqueous layer was extracted with EA and H2O (2x1 10 mL). The residue was purified by Prep-TLC (DCM:MeOH =20:1) to afford N-(3-chloro-2- methoxyphenyl)-2-([[3-(2-methoxy-2-methylpropoxy)pyridin-4-yl]methyl]amino)-6- oxocyclohex-l-ene-1 -carbothioamide (300 mg, 37.11%) as a light yellow solid.
LC-MS: M+H found: 504.1.
Figure imgf000232_0002
To a stirred solution of N-(3-chloro-2-methoxyphenyl)-2-([[3-(2-methoxy-2- methylpropoxy)pyridin-4-yl]methyl]amino)-6-oxocyclohex-l-ene-l -carbothioamide (288.00 mg, 0.571 mmol, 1.00 equiv) in MeOH (10.00 mL) was added H2O2 (97.18 mg, 2.855 mmol, 5.00 equiv) dropwise at room temperature under N2 atmosphere. The resulting mixture was stirred for 18 hours at 50 degrees C. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep -HPLC with the following conditions (Column: YMC-Actus Triart Cl 8 ExRS, 30*150 mm, 5pm; Mobile Phase A: Water(10MMOL/L NH4HCO3+0.1%NH3.H2O), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 28% B to 61% B in 7 min; Wave Length: 254/220 nm; RTl(min): 6.83) to afford 3-[(3-chloro-2-methoxyphenyl)amino]-2-[3-(2-methoxy-2- methylpropoxy)pyridin-4-yl]-l,5,6,7-tetrahydroindol-4-one (48.7 mg, 18.14%) as a off- white solid.
LC-MS: (M+H)+ found 470.1.
1H NMR (400 MHz, DMSO-d6) δ 11.29 (s, 1H), 8.45 (s, 1H), 8.06 (d, J = 5.0 Hz, 1H), 7.47 (s, 1H), 7.29 (d, J = 5.0 Hz, 1H), 6.74 - 6.65 (m, 2H), 6.14 (dd, J = 6.1, 3.5 Hz, 1H), 4.18 (s, 2H), 3.89 (s, 3H), 2.85 (t, J = 6.1 Hz, 2H), 2.36 (t, J = 6.3 Hz, 2H), 2.08 (t, J = 6.0 Hz, 2H), 1.27 (s, 6H).
Example 4. Synthesis of 3-[(3-chloro-2-methoxyphenyl)amino]-2-[3-[(2S)-oxolan-2- ylmethoxy]pyridin-4-yl]-l,5,6,7-tetrahydroindol-4-one (compound 162)
Figure imgf000233_0001
To a stirred solution of (2S)-oxolan-2-ylmethanol (1.62 g, 0.016 mmol, 1.10 equiv) inDMF was added NaH (0.69 g, 0.029 mmol, 2.00 equiv) in portions at 0 degrees C under nitrogen atmosphere. The resulting mixture was stirred for 0.5 h at room temperature under nitrogen atmosphere. To the above mixture was added 3-chloropyridine-4-carbonitrile (2.00 g, 14.435 mmol, 1.00 equiv) at 0 degrees C. The resulting mixture was stirred for additional 2 h at room temperature. The reaction was quenched by the addition of Water/Ice at room temperature. The resulting mixture was extracted with CH2C12 (4 x 20mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2C12 / MeOH (10:1) to afford 3-[(2S)-oxolan-2- ylmethoxy]pyridine-4-carbonitrile (1 g, 33.92%) as a yellow oil.
Figure imgf000234_0001
To a stirred solution of 3-[[(2S)-l-methylpyrrolidin-2-yl]methoxy]pyridine-4-carbonitrile (0.80 g, 3.682 mmol, 1.00 equiv) in NH3(g) in MeOH (10.00 mL, 1 equiv) was added Raney Ni (0.63 g, 7.364 mmol, 2.00 equiv) at room temperature under hydrogen atmosphere. The resulting mixture was stirred for overnight at room temperature under hydrogen atmosphere. The resulting mixture was filtered, and the filter cake was washed with MeOH. The filtrate was concentrated under reduced pressure. This resulted in l-(3- [[(2S)-l-methylpyrrolidin-2-yl]methoxy]pyridin-4-yl)methanamine (600 mg, 73.63%) as a yellow oil.
Figure imgf000234_0002
Into a 8 mL vial were added N-(3-chloro-2-methoxyphenyl)-2-hydroxy-6-oxocyclohex-l- ene-1 -carbothioamide (200.00 mg, 0.641 mmol, 1.00 equiv) and l-[3-[(2S)-oxolan-2- ylmethoxy]pyridin-4-yl]methanamine (146.95 mg, 0.706 mmol, 1.10 equiv) in DMA. The resulting mixture was stirred for 2 h at 120 degrees C under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was extracted with CH2C12 and concentrated under reduced pressure. The residue was purified by Prep-TLC (CH2C12 / MeOH 10: 1) to afford N-(3-chloro-2-methoxyphenyl)-6-oxo-2- [([3-[(2S)-oxolan-2-ylmethoxy]pyridin-4-yl]methyl)amino]cyclohex-l-ene-l- carbothioamide (150 mg, 46.58%) as a yellow solid.
Figure imgf000235_0001
To a stirred solution of N-(3-chloro-2-methoxyphenyl)-6-oxo-2-[([3-[(2S)-oxolan-2- ylmethoxy]pyridin-4-yl]methyl)amino]cyclohex-l-ene-l -carbothioamide (150.00 mg, 0.299 mmol, 1.00 equiv) and TFA (34.07 mg, 0.299 mmol, 1.00 equiv) in MeOH was added H2O2(30%) (15.24 mg, 0.448 mmol, 1.50 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 80 degrees C under nitrogen atmosphere. The resulting mixture was extracted with CH2C12 (3 x lOmL). The combined organic layers were concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xselect CSH OBD Column 30* 150mm 5pm; Mobile Phase A:Water(0.1%FA), Mobile Phase B:ACN; Flow rate:60 mL/min; Gradient: 25% B to 40% B in 8 min; Wave Length: 254 nm; RTl(min):6) to 3 - [(3 -chloro-2-methoxyphenyl)amino] -2- [3 - [(2S)-oxolan-2-ylmethoxy]pyridin-4-yl] - l,5,6,7-tetrahydroindol-4-one (25.2 mg) as a yellow solid.
Example 5. Synthesis of 3-[(3-chloro-2-methoxyphenyl)amino]-2-(3-[[(2S)-l- methylpyrrolidin-2-yl] methoxy] pyridin-4-yl)- 1,5, 6, 7-tetrahydroindol-4-one (compound 160)
Figure imgf000235_0002
To a stirred solution of [(2S)-l-methylpyrrolidin-2-yl]methanol (1.83 g, 0.016 mmol, 1.10 equiv) in DMF was added NaH (0.69 g, 0.029 mmol, 2.00 equiv) in portions at 0 degrees C under nitrogen atmosphere. The resulting mixture was stirred for 0.5 h at room temperature under nitrogen atmosphere. To the above mixture was added 3-chloropyridine- 4-carbonitrile (2.00 g, 14.435 mmol, 1.00 equiv) at 0 degrees C. The resulting mixture was stirred for additional 2 h at room temperature. The reaction was quenched by the addition of Water/Ice at room temperature. The resulting mixture was extracted with CH2C12 (4 x 20mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2C12 / MeOH (10:1) to afford 3-[[(2S)-l-methylpyrrolidin-2-yl]methoxy]pyridine-4-carbonitrile (1 g, 31.88%) as a yellow oil.
Figure imgf000236_0001
To a stirred solution of 3-[(2S)-oxolan-2-ylmethoxy]pyridine-4-carbonitrile (0.80 g, 3.917 mmol, 1.00 equiv) in NH3(g) in MeOH (10.00 mL, 1 equiv) was added Raney Ni (0.67 g, 7.834 mmol, 2 equiv) at room temperature under hydrogen atmosphere. The resulting mixture was stirred for overnight at room temperature under hydrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with MeOH. The filtrate was concentrated under reduced pressure. This resulted in l-[3-[(2S)-oxolan-2- ylmethoxy]pyridin-4-yl]methanamine (0.6 g, 73.55%) as a yellow oil.
Figure imgf000236_0002
Into a 8 mL vial were added N-(3-chloro-2-methoxyphenyl)-2-hydroxy-6-oxocyclohex-l- ene-1 -carbothioamide (200.00 mg, 0.641 mmol, 1.00 equiv) and l-(3-[[(2S)-l- methylpyrrolidin-2-yl]methoxy]pyridin-4-yl)methanamine (156.16 mg, 0.705 mmol, 1.10 equiv) in DMA. The resulting mixture was stirred for 2 h at 120 degrees C under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was extracted with CH2C12 and concentrated under reduced pressure. The residue was purified by Prep-TLC (CH2C12 / MeOH 10:1) to afford N-(3-chloro-2- methoxyphenyl)-2-[[(3-[[(2S)-l-methylpyrrolidin-2-yl]methoxy]pyridin-4- yl)methyl] amino] -6-oxocyclohex-l-ene-l -carbothioamide (150 mg, 45.40%) as a yellow solid.
Example 6. Synthesis of 3-[(3-chloro-2-methoxyphenyl)amino]-2-[3-(2- methoxyethoxy)pyridin -4-yl]-l,5,6,7-tetrahydroindol-4-one (compound 163)
Figure imgf000237_0001
A solution of l-[3-(2-methoxyethoxy)pyridin-4-yl]methanamine (400.00 mg, 2.195 mmol, 1.00 equiv) and N-(3-chloro-2-methoxyphenyl)-2-hydroxy-6-oxocyclohex-l-ene-l- carbothioamide (319.00 mg, 1.023 mmol, 0.74 equiv) in DMA (1.50 mL) was stirred at 120 °C under N2 atmosphere for 3h. The mixture was allowed to cool down to RT. The resulting mixture was extracted with EA (150ml) for three times. The combined organic layers were washed with brine(lOOml) for 2 times, dried over anhydrous NaiSO-i. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (DCM/MeOH=15: l) to afford N-(3-chloro-2-methoxyphenyl)-2-([[3-(2- methoxyethoxy)pyridin-4-yl]methyl]amino)-6-oxocyclohex-l-ene-l -carbothioamide (254 mg, 23.64%) as a yellow solid.
LC-MS: (M+H)+ found: 476.03.
Figure imgf000238_0001
A solution of N-(3-chloro-2-methoxyphenyl)-2-([[3-(2-methoxyethoxy)pyridin-4- yl] methyl] amino)-6-oxocyclohex-l-ene-l -carbothioamide (200.00 mg, 0.423 mmol, lequiv) in CH3OH (2.50 mL) was added H2O2 (0.25 mb) in portions at 50 °C under N2 atmosphere for 15h. The mixture was allowed to cool down to RT. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: YMC- Actus Triart Cl 8 ExRS, 30*150 mm, 5pm; MobilePhase A: Water(10MMOL/LNH4HCO3+0.1%NH3.H2O), Mobile PhaseB: ACN; Flow rate: 60 mL/min; Gradient: 38% B to 71% B in 7 min; Wave Length: 254/220 nm; RTl(min): 6.82) to afford 3-[(3-chloro-2-methoxyphenyl)amino]-2-[3-(2- methoxyethoxy)pyridin-4-yl]-l,5,6,7-tetrahydroindol-4-one (25.4 mg, 13.64%) as a light yellow solid .
LC-MS: (M+H)+ found 442.01.
1H NMR (400 MHz, DMSO-d6) δ 11.23 (s, 1H), 8.43 (s, 1H), 8.08 (d, J = 5.0 Hz, 1H), 7.47 (s, 1H), 7.29 (d, J = 5.0 Hz, 1H), 6.70 - 6.63 (m, 2H), 6.15 - 6.09 (m, 1H), 4.35 - 4.29 (m, 2H), 3.88 (s, 3H), 3.78 - 3.72 (m, 2H), 3.38 (s, 3H), 2.87 (t, J = 6.1 Hz, 2H), 2.36 (dd, J = 7.1, 5.5 Hz, 2H), 2.08 (dd, J = 7.0, 5.2 Hz, 2H). Example 7. Synthesis of 3-[(3-chloro-2-methoxyphenyl)amino]-2-(3-{2-[l- (difluoromethyl)cyclopropyl]ethynyl}pyridin-4-yl)-l,5,6,7-tetrahydroindol-4-one (compound 170)
Figure imgf000239_0001
To a stirred mixture of 2-(3-bromopyridin-4-yl)-3-[(3-chloro-2-methoxyphenyl)amino]- l,5,6,7-tetrahydroindol-4-one (100 mg, 0.22 mmol, 1.00 equiv) and 1 -(difluoromethyl)- 1- ethynylcyclopropane (248 mg, 0.45 mmol, 2.0 equiv)in DMF (2 mL) was added Pd(dppf)C12CH2C12 (91 mg, 0.11 mmol, 0.5 equiv) and Cui (21 mg, 0.11 mmol, 0.5 equiv) and DIEA (145 mg, 1.12 mmol, 5.0 equiv) in portions at room temperature under argon atmosphere. The resulting mixture was stirred for 4h at 65 degrees C under argon atmosphere. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water, 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 3-[(3-chloro-2-methoxyphenyl)amino]-2- (3-{2-[l-(difluoromethyl)cyclopropyl]ethynyl}pyridin-4-yl)-l,5,6,7-tetrahydroindol-4- one (58.7 mg, 51.69%) as a white solid.
LC-MS: M+H found: 482.0
'H NMR (400 MHz, DMSO) δ 1.1-1.38 (m, 4H), 2.07 (p, J= 6.2 Hz, 2H), 2.36 (t, J= 6.2 Hz, 2H), 2.84 (t, J = 6.1 Hz, 2H), 3.83 (s, 3H), 5.79 (t, J = 55.7 Hz, 1H), 6.01-6.13 (m, 1H), 6.56-6.68 (m, 2H), 7.26-7.4 (m, 2H), 8.40 (d, J= 5.3 Hz, 1H), 8.58 (s, 1H), 11.62 (s, 1H). Example 8. Synthesis of 3-[(3-chloro-2-methoxyphenyl)amino]-2-(3-{[(2R)-l-(prop-2- enoyl)azetidin-2-yl]methoxy}pyridin-4-yl)-l,5,6,7-tetrahydroindol-4-one (Compound 164)
Figure imgf000240_0001
To a stirred mixture of 1,5,6, 7-tetrahydroindol-4-one (500 mg, 3.69 mmol, 1.00 equiv) and bis(pinacolato)diboron (469 mg, 1.85 mmol, 0.50 equiv) in MTBE (6.00 mL) were added ( 1 , 5-cyclooctadiene)(methoxy)iridium(I) dimerdi-m-methoxobis( 1 , 5 -cyclooctadiene) diiridium (I) (73 mg, 0.11 mmol, 0.03 equiv) and 4,4'-Di-tert-butyl-2,2'-bipyridine (59 mg, 0.22 mmol, 0.06 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for Ih at 50 degrees C under argon atmosphere. The reaction was monitored by LCMS. LCMS showed the reaction was completed. The resulting mixture was concentrated under reduced pressure. The residue was washed with MTBE (3x10 mL). To afford 2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-l,5,6,7-tetrahydroindol-4-one (910 mg, 94.21%) as an off-white solid.
LC-MS: (M+H)+ found 262.00.
Figure imgf000240_0002
To a stirred solution of tert-butyl (2R)-2-(hydroxymethyl) azetidine- 1 -carboxylate (200 mg, 1.06 mmol, 1.00 equiv) and 4-bromopyri din-3 -ol (278 mg, 1.60 mmol, 1.50 equiv) in THF (10.00 mL) were addedPPhs (420 mg, 1.60 mmol, 1.50 equiv) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for 0.5 h at 0 °C under nitrogen atmosphere. To the above mixture was added DEAD (279 mg, 1.60 mmol, 1.50 equiv) dropwise at 0 °C. The resulting mixture was stirred for additional 0.5 h at 0 °C. The reaction was monitored by LCMS. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with EA/PE (30%) to afford tert-butyl (2R)-2-{[(4-bromopyridin-3-yl)oxy]methyl}azetidine-l -carboxylate (380 mg, 93.29%) as a colorless oil.
LC-MS: (M+H)+ found 345.1.
Figure imgf000241_0001
To a stirred mixture of tert-butyl (2R)-2-{[(4-bromopyridin-3-yl)oxy]methyl}azetidine-l- carboxylate (360 mg, 1.05 mmol, 1.00 equiv) and 2-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-l,5,6,7-tetrahydroindol-4-one (328 mg, 1.26 mmol, 1.20 equiv) in 1,4- dioxane (5.00 mb) and H2O (1.00 mL) were added NaiCCh (333 mg, 3.15 mmol, 3.00 equiv) and 2nd Generation XPhos Precatalyst (83 mg, 0.11 mmol, 0.10 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for Ih at 50 °C under argon atmosphere. The reaction was monitored by LCMS. LCMS showed the reaction was completed. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2G2 / MeOH (10:1) to afford tert-butyl (2R)-2-({[4-(4-oxo-l,5,6,7-tetrahydroindol-2-yl)pyridin- 3 -yl] oxy} methyl)azetidine-l -carboxylate (280 mg, 67.16%) as a yellow oil.
LC-MS: (M+H)+ found 398.2.
Figure imgf000241_0002
To a stirred solution of tert-butyl (2R)-2-({[4-(4-oxo-l,5,6,7-tetrahydroindol-2-yl)pyridin- 3 -yl] oxy} methyl)azetidine-l -carboxylate (270 mg, 0.68 mmol, 1.00 equiv) in DMF (3.00 mb) was added N-iodosuccinimide (168 mg, 0.75 mmol, 1.10 equiv) in portions at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. LCMS showed the reaction was completed. The reaction was quenched by the addition of Na2SO3(sat.) (3 mL) at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2G2 / MeOH (10: 1) to afford tert-butyl (2R)-2-({[4-(3-iodo-4-oxo-l,5,6,7-tetrahydroindol-2-yl)pyridin- 3 -yl] oxy} methyl)azetidine-l -carboxylate (110 mg, 30.94%) as a yellow solid.
LC-MS: (M+H)+ found 524.20.
Figure imgf000242_0001
To a stirred solution of tert-butyl (2R)-2-({[4-(3-iodo-4-oxo-l,5,6,7-tetrahydroindol-2- yl)pyridin-3-yl]oxy}methyl)azetidine-l-carboxylate (200 mg, 0.38 mmol, 1.00 equiv) in DMF (3.00 mL) was added CS2CO3 (249 mg, 0.76 mmol, 2.00 equiv) and EPhos Pd G4 (35 mg, 0.04 mmol, 0.10 equiv) at room temperature under nitrogen atmosphere. To the above mixture was added 3-chloro-2-methoxyaniline (60 mg, 0.38 mmol, 1.00 equiv) at room temperature. The resulting mixture was stirred for additional 2h at 50 °C. The reaction was monitored by LCMS. LCMS showed the reaction was completed. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2CI2 / MeOH (10: 1) to afford the crude product. The crude product was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 0% to 100% gradient in 45 min; detector, UV 254 nm. To afford tert-butyl (2R)-2-{[(4-{3-[(3-chloro- 2-methoxyphenyl)amino]-4-oxo-l,5,6,7-tetrahydroindol-2-yl}pyridin-3- yl)oxy] methyl} azetidine- 1 -carboxylate (75 mg, 35.49%) as a yellow solid.
LC-MS: (M+H)+ found 553.1.
Figure imgf000243_0001
To a stirred solution of tert-butyl (2R)-2-{[(4-{3-[(3-chloro-2-methoxyphenyl)amino]-4- oxo- 1 , 5 ,6,7-tetrahydroindol-2-yl } pyridin-3 -yl)oxy]methyl } azetidine- 1 -carboxylate (75 mg, 0.14 mmol, 1.00 equiv) in TFA (1.00 mL) was added DCM (1.00 mL) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for Ih at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. LCMS showed the reaction was completed. The resulting mixture was concentrated under reduced pressure. The crude product was used in the next step directly without further purification.
LC-MS: (M+H)+ found 453.00.
Figure imgf000243_0002
To a stirred solution of 2-{3-[(2R)-azetidin-2-ylmethoxy]pyridin-4-yl}-3-[(3-chloro-2- methoxyphenyl)amino]-l,5,6,7-tetrahydroindol-4-one (70 mg, 0.15 mmol, 1.00 equiv) in THF (1.00 mL) and NaHCO3(sat.) (1.00 mL) was added acryloyl chloride (16 mg, 0.18 mmol, 1.15 equiv) at O°C under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 0°C under nitrogen atmosphere. The reaction was monitored by LCMS. LCMS showed the reaction was completed. The resulting mixture was extracted with EtOAc (3 x 15mL), dried over anhydrous NaiSO-i. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: YMC- Actus Triart Cl 8 ExRS, 30*150 mm, 5pm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 37% B to 58% B in 9 min; Wave Length: 254/220 nm; RTl(min): 9.67) to afford 3-[(3- chloro-2-methoxyphenyl)amino] -2-(3 - { [(2R)- 1 -(prop-2-enoyl)azetidin-2- yl]methoxy}pyridin-4-yl)-l,5,6,7-tetrahydroindol-4-one (20.2 mg, 25.70%) as an off- white solid.
LC-MS: (M+H)+ found 506.90.
’H NMR (400 MHz, DMSO4) δ 11.53 (s, 1H), 8.37 (s, 1H), 7.99 (d, 1H), 7.29 (d, 2H), 6.61 (d, 2H), 6.32-6.20 (m, 1H), 6.16 - 6.07 (m, 2H), 5.64 (s, 1H), 4.80 (d, 1H), 4.55-4.38 (m, 2H), 4.10 (s, 2H), 3.90(s, 3H), 2.94-2.89 (m, 2H), 2.46-2.42(m, 1H), 2.38-2.32 (m, 2H), 2.20 - 1.99 (m, 3H).
Bioactivity
EXAMPLE A. Inhibitor activity on EGFR-dependent cell growth
Cell lines are generated by transducing Ba/F3 cells with retroviruses containing vectors with EGFR WT, EGFR L858R, EGFR exon 19del, EGFR L858R/C797S, EGFR exon 20 NPGIns D770_N771, EGFR exon 20 ASV Ins V769_D770, EGFR exon 20 SVD Ins D770_N771, or EGFR exon 20 FQEA Ins A763_V764 genes and a puromycin selection marker. Transduced cells are selected with puromycin for 7 days and are then be transferred into culture media without Interleukin 3 (IL3). EGFR WT cells are maintained with supplemental EGF. Surviving cells are confirmed to express EGFR by Western blot and maintained as a pool. The IC50 date are included in Table 6. Study Design
1 Cell seeding
1.1 Cells are harvested from flask into cell culture medium and the cell number counted.
1.2 Cells are diluted with culture medium to the desired density and 40 pL of cell suspension is added into each well of 384- well cell culture plate and the seeding density is 800 (FQEA, exon 19del), 600 (WT, NPG, L858R/C797S), or 400 (ASV, SVD, L858R) cells/well.
2 Compound preparation and treatment
2.1 Test compounds are dissolved to 10 mM in a DMSO stock solution. 45 pL of stock solution is transferred to a 384 polypropylene plate (pp-plate). Perform 3 -fold, 10-point dilution via transferring 15 pL compound into 30 pL DMSO using a TECAN (EV0200) liquid handler.
2.2 Spin plates at room temperature at 1,000 RPM for 1 minute.
2.3 Transfer 120 nL of diluted compound from compound source plate into the cell plate.
2.4 After compound treatment for 72 hours, perform CTG detection for compound treatment plates as described in "Detection" section.
3 Detection
3.1 Plates are removed from incubators and equilibrated at room temperature for 15 minutes.
3.2 Thaw the CellTiter Gio (CTG) reagents and allow to equilibrate to room temperature before the experiment.
3.3 Add 40 pL of CellTiter-Glo reagent into each well (at 1: 1 to culture medium). Then place the plates at room temperature for 30 min followed by reading on EnVision.
4 Data analysis
4.1 Inhibition activity is calculated following the formula below:
%Inhibition = 100 x (LumHC - LumSample) / (LumHC -LumLC) where HC is obtained from cells treated with 0.1% DMSO only; and LC is obtained from culture medium only. 4.22. Calculate the IC50 by fitting the Curve using Xlfit (v5.3.1.3), equation 201:
Y = Bottom + (Top - Bottom)/(1 + 10A((LogICso - X)*HillSlope))
The IC50 date are included in Table 6.
Table 6. IC50 Data for EGFR Inhibitor Activity on EGFR Proliferation (CTG)1
Figure imgf000246_0001
1 CTG means Cell Titer Gio which is a measure of proliferation;
“+++” indicates that IC50 < 100 nM;
“++” indicates that 100 nM <= IC50 < 1000 nM;
“+” indicates that IC50 >= 1000 nM;
“NA” indicates that the IC50 data is not available for this compound.
EXAMPLE B. Inhibitor Activity on EGFR phosphorylation (pEGFR)
EGFR mutant Ba/F3 cells were generated by transduction with retrovirus containing vectors expressing EGFR L858R, EGFR exon 19del, EGFR L858R/C797S, EGFR exon 20 NPGIns D770 N771, EGFR exon 20 ASV Ins V769 D770, or EGFR exon 20 SVD Ins D770_N771 genes along with a puromycin selection marker. Transduced cells are selected with puromycin for 7 days and are then be transferred into culture media without Interleukin 3 (IL3). Surviving cells are confirmed to express EGFR by Western blot and maintained as a pool. CUTO14 cells were obtained from Dr. Robert C. Doebele at the University of Colorado. Study Design
1 Cell seeding
1.1 Cells are harvested from flask into cell culture medium and the cell number counted.
1.2 Cells are diluted with culture medium to the desired density and 40 pL of cell suspension is added into each well of 384- well cell culture plate and the seeding density is 50K cells/well (Ba/F3) or 12.5K cells/well (CUTO14).
2 Compound preparation and treatment
2.1 Test compounds are dissolved to 10 mM in a DMSO stock solution. 45 pL of stock solution is transferred to a 384 polypropylene plate (pp-plate). Perform 3 -fold, 10-point dilution via transferring 15 pL compound into 30 pL DMSO using a TECAN (EV0200) liquid handler.
2.2 Spin plates at room temperature at 1,000 RPM for 1 minute.
2.3 Transfer 5 nL of diluted compound from compound source plate into the cell plate.
2.4 After compound treatment for 2 hours, perform pEGFR detection by AlphaLISA for compound treatment plates as described in "Detection" section.
3 Detection by pEGFR AlphaLISA (Perkin-Elmer)
3.1 Plates are removed from incubators and equilibrated at room temperature for 10 minutes, and media was removed
3.2 10 pL of lysis buffer is added and plates shaken at 600 rpm for 1 hr.
3.3 Prepare acceptor mix just before use and dispense 5 pL of acceptor mix to all the wells. Shake 350 rpm for Ihr in the dark
3.4 Prepare donor mix under low light conditions prior to use. Dispense 5 pL of donor mix to all the wells. Mix well on the shaker, seal and wrap in aluminum foil and incubate 1.5 hrs at room temperature in the dark
3.5 Transfer 18.5 pL mixture to OptiPlate 384, and read using an Envision.
The pEGFR IC50 date were included in Table 7. Table 7. IC50 Data for EGFR Inhibitor Activity on EGFR phosphorylation (pEGFR)1
Figure imgf000248_0001
1 “+++” indicates that IC50 < 100 nM;
“++” indicates that 100 nM <= IC50 < 1000 nM;
“+” indicates that IC50 >= 1000 nM;
“NA” indicates that the IC50 data is not available for this compound.

Claims

WHAT IS CLAIMED IS:
1. A compound of Formula (I):
Figure imgf000249_0001
Formula (I) or a pharmaceutically acceptable salt thereof, wherein:
Ring C is selected from the group consisting of:
Figure imgf000249_0002
• bicyclic heteroaryl including 7-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with X1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc;
• bicyclic C5-10 cycloalkyl or C5-10 cycloalkenyl, each of which is optionally substituted with X1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc;
• heterocyclyl or heterocycloalkenyl including from 5-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with X1 and further optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc; and
• C 10 or C 14 aryl optionally substituted with X1 and further optionally substituted with from 1-4 Rc; R6 . / I — -
X1 is selected from the group consisting of (a) -(X2)m-L1-R5; and (b) « 2
; wherein: m is 0 or 1 ;
X2 is selected from the group consisting of:
Figure imgf000250_0001
• -C2-6 alkenyl ene optionally substituted with from 1-3 Ra;
• -C(=O)O-*, -C(=O)N(RN)-*, or -S(O)I-2N(RN)-*;
• -OC(=O)-*, -N(RN)C(=O)-*, or -N(RN)S(O)I-2-*; and
• -OC(=O)N(RN)-*, -N(RN)C(=O)O-*, -N(RN)C(=O)N(RN)-*, or - N(RN)S(O)I-2N(RN)-*, wherein the asterisk represents point of attachment to L1;
L1 and L2 is selected from the group consisting of: a bond and C1-10 alkylene optionally substituted with from 1-6 Ra;
R5 is selected from the group consisting of:
• H;
• halo;
• -OH;
• -NReRf;
• -C1-6 alkoxy or -S(0)o-2(C1-6 alkyl), each optionally substituted with from 1-6 Ra;
• -Rg;
• -L5-Rg;
• -Rg2-Rw or -Rg2-RY; and
• -L5-Rg2-Rw or -L5-Rg2-RY; R6 is selected from the group consisting of:
• H;
• halo;
• -OH;
• -NReRf;
• -Rg;
• -R"
• -L6-Rg;
• -Rg2-Rw or -Rg2-RY;
• -L6-Rg2-Rw or -L6-Rg2-RY; and
• -C1-6 alkoxy or -S(0)o-2(C1-6 alkyl), each optionally substituted with from 1-6 Ra; provided that: when L1 is a bond, then R5 is selected from the group consisting of: H, -Rg, -Rg2- Rw, and -Rg2-RY; and
X1 is other than H, -OH, or NH2;
L5 is selected from the group consisting of: -O-, -S(0)o-2, -NH-, and -N(Rd)-;
Rw is -Lw-W, wherein Lw is C(=O), S(O)i-2, OC(=O)*, NHC(=O)*, NRdC(=O)*, NHS(O)I-2*, or NRdS(0)i-2*, wherein the asterisk represents point of attachment to W, and
W is selected from the group consisting of:
• C2-6 alkenyl; C2-6 alkynyl; or C3-10 allenyl, each of which is optionally substituted with from 1-3 Ra and further optionally substituted with Rg, wherein W is attached to Lw via an sp2 or sp hybridized carbon atom, thereby providing an a, P- unsaturated system; and
• bicyclo [x.y.O] cycloalkyl optionally substituted with from 1 -2 Rc, wherein x is 1 or 2; and y is an integer from 1 to 6; RY is selected from the group consisting of: -Rg and -(Lg)g-Rg; each of Rla, Rlb, R2a, R2b, R3a, and R3b is independently selected from the group consisting of:
• H;
• halo;
• -OH;
• -C(O)OH or -C(O)NH2;
• -CN;
• -Rb;
• -Lb-Rb;
• -C1-6 alkoxy or -C1-6 thioalkoxy, each optionally substituted with from 1-6 Ra;
• NReRf;
• Rg; and
. -(Lg)g-Rg;
. -(Lg)g-Rw;
• -(Lg)g-Rg2-Rw; or any two of variables Rla, Rlb, R2a, R2b, R3a, and R3b, together with the Ring B ring atoms to which each is attached, form a fused saturated or unsaturated ring of 3-12 ring atoms;
• wherein from 0-2 of the ring atoms are each an independently selected heteroatom , wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated or unsaturated ring of 3-12 ring atoms is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rw;
Ring A is Rg;
R4 is selected from the group consisting of: H and Rd; each R7 is an independently selected Rc; n is 0, 1, 2, or 3; each occurrence of Ra is independently selected from the group consisting of: - OH; -halo; -NReRf; C1-4 alkoxy; C1-4 haloalkoxy; -C(=O)O(C1-4 alkyl); -C(=O)(C1-4 alkyl); -C(=O)OH; -CONR’R”; -S(O)I-2NR’R”; -S(O)I-2(CM alkyl); and cyano; each occurrence of Rb is independently C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each of which is optionally substituted with from 1-6 Ra; each occurrence of Lb is independently C(=O); S(O)i-2; C(=O)O*; C(=O)NH*; C(=O)NRd*; S(O)I-2NH*; or S(O)i-2N(Rd)*, wherein the asterisk represents point of attachment to Rb; each occurrence of Rc is independently selected from the group consisting of: halo; cyano; C1-10 alkyl which is optionally substituted with from 1-6 independently selected Ra; C2-6 alkenyl; C2-6 alkynyl; C1-4 alkoxy optionally substituted with C1-4 alkoxy or CM haloalkoxy;C1-4 haloalkoxy; -S(O)I-2(CM alkyl); -S(O)(=NH)(CM alkyl); -NReRf; -OH; -S(O)I-2NR’R”; -CM thioalkoxy; -NO2; -C(=0)(Ci-io alkyl); -C(=O)O(CM alkyl); - C(=O)OH; -C(=O)NR’R”; and -SF5; each occurrence of Rd is independently selected from the group consisting of: CM alkyl optionally substituted with from 1-3 independently selected Ra; -C(O)(CM alkyl); - C(O)O(Ci-4 alkyl); -CONR’R”; -S(O)I-2NR’R”; - S(O)I-2(CM alkyl); -OH; and CM alkoxy; each occurrence of Re and Rf is independently selected from the group consisting of: H; C1-4 alkyl optionally substituted with from 1-3 substituents each independently selected from the group consisting of NR’R”, -OH, C1-4 alkoxy, C1- h4aloalkoxy, and halo; -C(O)(CM alkyl); -C(O)O(CM alkyl); -CONR’R”; -S(O)I-2NR’R”; - S(O)I-2(CM alkyl); -OH; and C1-4 alkoxy; each occurrence of Rg is independently selected from the group consisting of: • C3-8 cycloalkyl or C3-8 cycloalkenyl, each of which is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc;
• heterocyclyl or heterocycloalkenyl including from 3-8 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc;
• heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc; and
• Ce-io aryl optionally substituted with from 1-4 Rc; each occurrence of Lg is independently selected from the group consisting of: -O-, -NH-, -NRd, -S(0)o-2, C(O), and C1-3 alkylene optionally substituted with from 1-3 Ra; each g is independently 1, 2, or 3; each Rg2 is a divalent Rg group; each occurrence of R’ and R” is independently selected from the group consisting of: H; -OH; and C1-4 alkyl;
Figure imgf000254_0001
2. The compound of claim 1, wherein Ring C is x1 (R7ln .
3. The compound of claim 2, wherein X1 is -(X2) 111" L1-R5.
4. The compound of claim 2 or 3, wherein X1 is -O-L1-Rg2-Rw.
R6
5. The compound of claim 2, wherein X1 is
Figure imgf000254_0002
6. The compound of any one of claims 2 or 5, wherein R6 is -Rg2-Rw.
7. The compound of any one of claims 2 or 5-6, wherein -R6 is
Figure imgf000255_0001
, wherein Ring D is heterocyclylene including from 3-10 ring atoms, wherein from 0-2 ring atoms (in addition to the ring nitrogen atom bonded to Rw) are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclylene is optionally substituted with from 1-3 substituents each independently selected from the group consisting of: oxo and -Rc; optionally wherein -R6 is a monocyclic heterocyclylene ring including from 3-10 ring atoms
Figure imgf000255_0002
R6 is a bicyclic heterocyclylene ring including from 3-10 ring atoms as defined above with
Figure imgf000255_0003
8. The compound of any one of claims 2 or 5-7, wherein
Figure imgf000256_0001
optionally substituted with from 1-2 Rc, wherein xl and x2 are each independently 0, 1, or 2.
9. The compound of claim 8, wherein xl = 0, and x2 = 0; or xl = 0, and x2 = 1 ; or xl = 0, and x2 = 2.
10. The compound of any one of claims 5-9, wherein R6 is selected from the
Figure imgf000256_0002
11. The compound of any one of claims 4 or 6-10, wherein -Rw is -Lw-W; and Lw is C(=O) NHC(=O)*, NRdC(=O)* (e.g., NMeC(=O)*), or NHS(O)I-2* wherein the asterisk represents point of attachment to W.
12. The compound of claim 11, wherein W is C2-6 alkenyl or C2-6 optionally substituted with from 1-3 Ra and further optionally substituted with Rg, wherein W is attached to Lw via an sp2 or sp hybridized carbon atom.
13. The compound of any one of claims 11 or 12, wherein W is C2-4 alkenyl (e.g., CH=CH2) or C2-4 alkynyl (e.g.,
Figure imgf000256_0003
optionally substituted with from 1-3 Ra and further optionally substituted with Rg, wherein W is attached to Lw via an sp2 or sp hybridized carbon atom.
14. The compound of claim 11, wherein -Lw-W is -C(=O)CH=CH2; - C(=O)NHCH=CH2; C(=O)CH=CHCH2NReRf (e g., C(=O)CH=CHCH2N(HMe),
Figure imgf000257_0001
15. The compound of claim 1, wherein Ring C is bicyclic heteroaryl including 7-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with X1 and further optionally substituted with from 1-4 RcA, wherein each RcA is an independently selected Rc.
16. The compound of claims 1 or 15, wherein Ring C is bicyclic heteroaryl including 9-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with X1 and further optionally substituted with from 1 -4 RcA, wherein each RcA is an independently selected Rc.
17. The compound of any one of claims 1 or 15-16, wherein Ring C is bicyclic heteroaryl including 9-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 RcA, wherein each RcA is an independently selected Rc.
18. The compound of any one of claims 15-17, wherein Ring C is connected to
Figure imgf000258_0001
. via a 6-membered ring present in Ring C.
19. The compound of any one of claims 1 or 15-18, wherein Ring C is (RcA)n
HO
( D ) ; Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA; n is 0, 1, or 2; and each RcA is an independently selected
Rc
20. The compound of claim 19, wherein Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
21. The compound of any one of claims 1 or 15-20, wherein Ring C is selected from the group consisting of:
Figure imgf000258_0002
Figure imgf000259_0001
each further optionally substituted with RcA, wherein each RcA is an independently selected Rc.
22. The compound of any one of claims 1 or 15-21, wherein Ring C is selected
Figure imgf000259_0002
optionally substituted with RcA, wherein each RcA is an independently selected Rc.
23. The compound of any one of claims 1, or 15-22, wherein Ring C is
Figure imgf000259_0003
25. The compound of any one of claims 1 or 15-22, wherein Ring C is selected from the group consisting of:
Figure imgf000260_0001
each occurrence of RcA is independently selected from the group consisting of: halo; NReRf; C1-4 alkoxy; C1-4 haloalkoxy; C1-3 alkyl; C1-3 alkyl substituted with from 1-3 independently selected halo; C1-3 alkyl substituted with C1-4 alkoxy; and C1-4 alkoxy substituted with C1-4 alkoxy; such as wherein each occurrence of RcA is independently selected from the group consisting of: C1-4 alkoxy; C1-4 haloalkoxy; C1-3 alkyl; and C1-3 alkyl substituted with from
1-3 independently selected halo.
26. The compound of claim 19, wherein Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
27. The compound of any one of claims 1, 15-19 or 26, wherein Ring C is
Figure imgf000260_0002
substituted with RcA, wherein each RcA is an independently selected Rc.
28. The compound of any one of claims 1 or 15-19, wherein Ring C is
Figure imgf000261_0001
Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA; n2 is 0 or 1; and each RcA is an independently selected Rc.
29. The compound of claim 28, wherein Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
30. The compound of any one of claims 1 or 28-29, wherein Ring C is selected from the group consisting of:
Figure imgf000261_0002
Figure imgf000261_0003
each further optionally substituted with RcA, wherein each
RCA is an independently selected Rc.
31. The compound of claim 28, wherein Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
32. The compound of any one of claims 28 or 31, wherein Ring C is selected from the group consisting of:
Figure imgf000262_0001
Figure imgf000262_0002
each further optionally substituted with RcA, wherein each
RCA is an independently selected Rc.
33. The compound of any one of claims 1 or 15-19, wherein Ring C is selected from the group consisting of:
Figure imgf000262_0003
Figure imgf000262_0004
each further optionally substituted with RcA, wherein each RcA is an independently selected Rc.
34. The compound of claims 15 or 16, wherein Ring C is connected to
Figure imgf000262_0005
via a 5-membered ring present in Ring C.
35. The compound of any one of claims 1, 15-16 or 34, wherein Ring C is
Figure imgf000262_0006
Figure imgf000263_0001
Figure imgf000263_0002
, each further optionally substituted with RcA, wherein each RcA is an independently selected Rc.
36. The compound of any one of claims 1 or 15-16, wherein Ring C is bicyclic heteroaryl including 9-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is substituted with X1 and further optionally substituted with from 1-4 RCA, wherein each RcA is an independently selected Rc.
37. The compound of any one of claims 1, 15-16 or 36, wherein Ring C is
Figure imgf000263_0003
Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA; n is 0, 1, or 2; and each RcA is an independently selected Rc
38. The compound of claim 37, wherein Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
39. The compound of any one of claims 1, 15-16 or 36-38, wherein Ring C is selected from the group consisting of:
Figure imgf000264_0001
, and
Figure imgf000264_0002
, each further optionally substituted with RcA, wherein each RcA is an independently selected Rc.
40. The compound compound of any one of claims 1, 15-16, or 39, wherein
Ring C is selected from the group consisting of:
Figure imgf000264_0003
,
Figure imgf000264_0004
each of which is further optionally substituted with from 1-2
RCA, wherein each RcA is an independently selected Rc.
41. The compound of any one of claims 1, 15-16, or 40, wherein Ring C is
Figure imgf000264_0005
42. The compound of any one of claims 1, 15-16, or 40, wherein Ring C is
Figure imgf000265_0001
43. The compound of any one of claims 1, 15-16, or 40, wherein Ring C is
Figure imgf000265_0002
wherein RcA is an independently selected Rc.
44. The compound of claim 37, wherein Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
45. The compound of any one of claims 37 or 44, wherein Ring C is selected from the group consisting of:
Figure imgf000265_0003
and
Figure imgf000265_0004
each further optionally substituted with RcA, wherein each RcA is an independently selected Rc.
46. The compound of any one of claims 1,15-16 or 36, wherein Ring C is
Figure imgf000266_0001
Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA; n2 is 0 or 1; and each RcA is an independently selected Rc.
47. The compound of claim 46, wherein Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
48. The compound of any one of claims 46-47, wherein Ring C is selected from the group consisting of:
Figure imgf000266_0002
substituted with RcA, wherein each RcA is an independently selected Rc.
49. The compound of claim 46, wherein Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
50. The compound of any one of claims 1 -49, wherein m is 1.
51. The compound of any one of claims 1-50, wherein X2 is selected from the group consisting of: -O-, -N(RN)-, and -S(0)o-2.
52. The compound of any one of claims 1-51, wherein X2 is -N(RN)-.
53. The compound of any one of claims 1-51, wherein X2 is -N(H)-.
54. The compound of any one of claims 1-51, wherein X2 is -O-.
55. The compound of any one of claims 1-50, wherein X2 is selected from the group consisting of: -OC(=O)-*, -N(RN)C(=O)-*, and -N(RN)S(O)I-2-*.
56. The compound of any one of claims 1-50, or 55, wherein X2 is - N(RN)C(=O)-*.
57. The compound of any one of claims 1-50, or 55-56, wherein X2 is - N(H)C(=O)-*.
58. The compound of any one of claims 1-50, or 55, wherein X2 is - N(RN)S(O)2-*, such as -NHS(O)2-.
59. The compound of any one of claims 1-50, wherein X2 is selected from the group consisting of: -OC(=O)N(RN)-*, -N(RN)C(=O)O-*, -N(RN)C(=O)N(RN)-*, and - N(RN)S(O)I-2N(RN)-*.
60. The compound of any one of claims 1-50 or 59, wherein X2 is - N(RN)C(=O)O-*, such as -N(H)C(=O)O-*.
61. The compound of any one of claims 1-50 or 59, wherein X2 is - N(RN)C(=O)N(RN)-*, such as -N(H)C(=O)N(H)-*.
62. The compound of any one of claims 1-50 or 59, wherein X2 is -C(=O)O-*, -C(=O)N(RN)-*, or -S(O)I-2N(RN)-*.
63. The compound of any one of claims 1-50 or 62, wherein X2 is - C(=O)N(RN)-*, such as -C(=O)N(H)-*.
64. The compound of any one of claims 1-50, wherein X2 is
Figure imgf000268_0001
65. The compound of any one of claims 1-50, wherein X2 is C2-6 alkenylene optionally substituted with from 1-3 Ra.
66. The compound of any one of claims 1-50 or 65, wherein X2 is
Figure imgf000268_0002
67. The compound of any one of claims 1 -49, wherein m is 0.
68. The compound of any one of claims 1-67, wherein L1 is a bond.
69. The compound of any one of claims 1-67, wherein L1 is C1-10 alkylene optionally substituted with from 1-6 Ra.
70. The compound of any one of claims 1-67 or 69, wherein L1 is C1-3 alkylene optionally substituted with from 1-6 Ra, such as wherein L1 is unsubstituted C1-3 alkylene.
71. The compound of any one of claims 1-67 or 69-70, wherein L1 is -CH2-, - CH2CH2-, -CH2CF2-, or-CH(Me)-, such as wherein L1 is -CH2-, -CH2CH2-, or-CH(Me)-
72. The compound of any one of claims 1-67 or 69, wherein L1 is branched C3-
6 alkylene optionally substituted with from 1-6 Ra, such as wherein L1 is
Figure imgf000268_0003
Figure imgf000268_0004
, wherein aa is the point of attachment to R5.
73. The compound of any one of claims 1-67 or 69-72, wherein R5 is -C1-6 alkoxy or -S(0)o-2(C1-6 alkyl), each optionally substituted with from 1-6 Ra.
74. The compound of any one of claims 1-67 or 69-73, wherein R5 is -C1-6 alkoxy optionally substituted with from 1-6 Ra.
75. The compound of any one of claims 1-67 or 69-74, wherein R5 is -C1-3 alkoxy, such as methoxy.
76. The compound of any one of claims 1-72, wherein R5 is H or halo, such as wherein R5 is H or -F.
77. The compound of any one of claims 1-72 or 76, wherein R5 is H.
78. The compound of any one of claims 1-72, wherein R5 is -OH or -NReRf.
79. The compound of any one of claims 1-72 or 78, wherein R5 is -OH.
80. The compound of any one of claims 1 -72, wherein R5 is -Rg.
81. The compound of any one of claims 1-72 or 80, wherein R5 is selected from the group consisting of:
• heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1 -4 Rc; and
• Ce-io aryl optionally substituted with from 1-4 Rc.
82. The compound of any one of claims 1-72 or 80-81, wherein R5 is Ce-io aryl optionally substituted with from 1-4 Rc.
83. The compound of any one of claims 1-72 or 80-82, wherein R5 is phenyl optionally substituted with from 1-4 Rc.
84. The compound of any one of claims 1-72 or 80-83, wherein R5 is phenyl optionally substituted with from 1-2 independently selected halo, such as -F.
85. The compound of any one of claims 1-72 or 80-81, wherein R5 is heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc.
86. The compound of any one of claims 1-72, 80-81, or 85, wherein R5 is heteroaryl including from 5-6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc.
87. The compound of any one of claims 1-72, 80-81, or 85-86, wherein R5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(Rd), and wherein the heteroaryl is optionally substituted with from 1 -4 Rc, such as wherein
Figure imgf000270_0001
Figure imgf000270_0002
88. The compound of any one of claims 1-72, 80-81, or 85-86, wherein R5 is heteroaryl including 5 ring atoms, wherein from 1-4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc, such as wherein
Figure imgf000270_0003
89. The compound of any one of claims 1 -72 or 80, wherein R5 is selected from the group consisting of:
• C3-10 cycloalkyl or C3-10 cycloalkenyl, each of which is optionally substituted with from 1 -4 substituents independently selected from the group consisting of oxo and Rc; and • heterocyclyl or heterocycloalkenyl including from 3-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
90. The compound of any one of claims 1-72, 80, or 89, wherein R5 is C3-10 cycloalkyl or C3-10 cycloalkenyl, each of which is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
91. The compound of any one of claims 1-72, 80, or 89-90, wherein R5 is C3-10 cycloalkyl, such as C3-6 cycloalkyl, optionally substituted with from 1-4 Rc, such as wherein R5 is cyclopropyl.
92. The compound of any one of claims 1-72, 80, or 89, wherein R5 is heterocyclyl or heterocycloalkenyl including from 3-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with from 1 -4 substituents independently selected from the group consisting of oxo and Rc.
93. The compound of any one of claims 1-72, 80, 89 or 92, wherein R5 is heterocyclyl including from 4-8, such as 4-6, ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc, such as
Figure imgf000271_0001
94. The compound of any one of claims 1 -72, wherein R5 is selected from the group consisting of: -Rg2-Rw and -Rg2-RY.
95. The compound of any one of claims 1 -72 or 94, wherein R5 is -Rg2-RY.
96. The compound of claims 94 or 95, wherein the -Rg2 group present in R5 is Ce-io arylene optionally substituted with from 1 -4 Rc.
97. The compound of any one of claims 94-96, wherein the -Rg2 group present in R5 is phenylene optionally substituted with from 1-4 Rc.
98. The compound of any one of claims 94-96, wherein the -Rg2 group present in R5 is 1,3-phenylene or 1 ,4-phenylene, each optionally substituted with from 1-4 Rc, such
Figure imgf000272_0001
wherein bb is the point of attachment to RY.
99. The compound of any one of claims 94-95, wherein the RY group present in R5 is -Rg.
100. The compound of any one of claims 94-95 or 99, wherein the RY group present in R5 is heterocyclyl or heterocycloalkenyl including from 3-10 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl or heterocycloalkenyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
101. The compound of any one of claims 94-95 or 99-100, wherein the RY group present in R5 is heterocyclyl including from 4-8, such as 4-6, ring atoms, wherein from 1- 3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc, such as wherein
Figure imgf000272_0002
102. The compound of any one of claims 1-72, wherein R5 is -L5-Rg.
103. The compound of any one of claims 1-72, wherein R5 is -O-Rg.
104. The compound of any one of claims 1-72 or 102-103, wherein R5 is -O-(Ce- io aryl) wherein the Ce-io aryl is optionally substituted with from 1-4 Rc.
105. The compound of any one of claims 1-72 or 102-104, wherein R5 is -O- phenyl wherein the phenyl is optionally substituted with from 1-2 Rc, such as wherein R5
Figure imgf000273_0001
106. The compound of any one of claims 1-49, wherein X1 is -(X2) 111" L'-R5 wherein:
• m is 0 or 1;
• X2 is -N(RN)- or -O-;
• L1 is a bond or C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is -Rg.
107. The compound of any one of claims 1-49, wherein X1 is -X2-L1-R5, wherein:
Figure imgf000273_0002
• L1 is a bond or C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is -Rg.
108. The compound of any one of claims 1-49, wherein X1 is -X2-L1-R5, wherein:
Figure imgf000273_0003
L1 is a bond or C1-6 alkylene optionally substituted with from 1-3 Ra; and R5 is - R"
109. The compound of any one of claims 106-108, wherein R5 is phenyl optionally substituted with from 1-4 Rc, such as wherein R5 is phenyl optionally substituted with from 1-2 independently selected halo, such as -F.
110. The compound of any one of claims 106-108, wherein R5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(Rd), and wherein the heteroaryl is optionally substituted with from 1-4 Rc, such as wherein
Figure imgf000274_0001
Figure imgf000274_0002
111. The compound of any one of claims 106-108, wherein R5 is heteroaryl including 5 ring atoms, wherein from 1-4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc, such as wherein R5 is
Figure imgf000274_0003
112. The compound of any one of claims 106-108, wherein R5 is C3-10 cycloalkyl, such as C3-6 cycloalkyl, optionally substituted with from 1-4 Rc, such as wherein R5 is cyclopropyl.
113. The compound of any one of claims 106-108, wherein R5 is heterocyclyl including from 4-8, such as 4-6, ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc, such as wherein R5 is
Figure imgf000275_0001
114. The compound of any one of claims 1-49, wherein X1 is -(X2) 111" L'-R5 wherein:
• m is 0 or 1;
• X2 is -N(RN)- or -O-;
• L1 is a bond or C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is -Rg2-RY
115. The compound of claim 114, wherein the -Rg2 group present in R5 is 1,3- phenylene or 1 ,4-phenylene, each optionally substituted with from 1-4 Rc, such as wherein
Figure imgf000275_0002
116. The compound of claims 114 or 115, wherein the RY group present in R5 is
-Rg.
117. The compound of any one of claims 114-116, wherein the RY group present in R5 is heterocyclyl including from 4-8, such as 4-6, ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1- 4 substituents independently selected from the group consisting of oxo and Rc, such as wherein
Figure imgf000275_0003
118. The compound of any one of claims 1-49, wherein X1 is -X2-L1-R5, wherein:
• X2 is -N(RN)-, -O-, -N(RN)C(=O)-*, -N(RN)S(O)2-, -N(RN)C(=O)O-*, or -N(RN)C(=O)N(RN)-*;
• L1 is C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is H, halo, C1-6 alkoxy optionally substituted with from 1-3 Ra, or -OH.
119. The compound of claim 118, wherein R5 is H.
120. The compound of claim 118, wherein R5 is halo, such as -F.
121. The compound of claim 118, wherein R5 is C1-6 alkoxy optionally substituted with from 1-3 Ra, such as wherein R5 is C1-3 alkoxy such as methoxy.
122. The compound of claim 118, wherein R5 is -OH.
123. The compound of any one of claims 106-122, wherein m is 0.
124. The compound of any one of claims 106-122, wherein m is 1.
125. The compound of any one of claims 106 or 114, wherein X2 is -N(RN)-, such as N(H).
126. The compound of any one of claims 106 or 114, wherein X2 is -O-.
127. The compound of any one of claims 107 or 118, wherein X2 is - N(RN)C(=O)-*, such as -N(H)C(=O)-*.
128. The compound of any one of claims 107 or 118, wherein X2 is -N(RN)S(O)2- , such as -N(H)S(O)2-*.
129. The compound of any one of claims 107 or 118, wherein X2 is - N(RN)C(=O)O-*, or -N(RN)C(=O)N(RN)-*, such as -N(H)C(=O)O-* or - N(H)C(=O)N(H)-*.
130. The compound of any one of claims 108 or 123, wherein X2 is
Figure imgf000277_0001
131. The compound of any one of claims 108 or 123, wherein X2 is
Figure imgf000277_0002
132. The compound of any one of claims 108-131, wherein L1 is a bond.
133. The compound of any one of claims 108-131, wherein L1 is C1-3 alkylene, such as -CH2-, -CH2CH2-, or -CH(Me)-.
134. The compound of any one of claims 108-131, wherein L1 is branched C3-6 alkylene, such as
Figure imgf000277_0003
, wherein aa is the point of attachment to R5.
135. The compound of any one of claims 1-92, wherein X1 is L'-R5, wherein L1 is C1-6 alkylene optionally substituted with from 1-3 Ra; and R5 is -L5-Rg.
136. The compound of claim 135, wherein R5 is -O-Rg.
137. The compound of claims 135 or 136, wherein R5 is -O-(phenyl), wherein the phenyl is optionally substituted with from 1-2 Rc.
138. The compound of any one of claims 135-137, wherein L1 is C1-3 alkylene, such as -CH2-, -CH2CH2-, or -CH(Me)-.
139. The compound of any one of claims 2-138, wherein each occurrence of RcA is independently selected from the group consisting of: halo; cyano; C1-10 alkyl which is optionally substituted with from 1-6 independently selected Ra; C1-4 alkoxy optionally substituted with C1-4 alkoxy or C1-4 haloalkoxy; C1-4 haloalkoxy; -S(O)i-2(C1-4 alkyl); - NReRf; -OH; -S(O)I-2NR’R”; -CM thioalkoxy; -C(=0)(Ci-io alkyl); -C(=O)O(CM alkyl); -C(=O)OH; and -C(=O)NR’R”
140. The compound of any one of claims 2-139, wherein one occurrence of RcA is -NReRf.
141. The compound of any one of claims 2-140, wherein one occurrence of RcA is -NH2.
142. The compound of any one of claims 2-140, wherein one occurrence of RcA is -NH(CI-6 alkyl), wherein the C1-6 alkyl is optionally substituted with from 1-3 substituents each independently selected from the group consisting of NR’R”, -OH, C1-6 alkoxy, C1-6 haloalkoxy, and halo.
143. The compound of any one of claims 2-140 or 142, wherein one occurrence of RCA IS -NHMe, -NHCH2CF3, -NHCH2CH2OH, or -NHiPr.
144. The compound of any one of claims 2-140, wherein one occurrence of RcA is -NHC(=O)CI-4 alkyl, such as NHC(=O)CH3; or wherein one occurrence of RcA is N(Ci- 3 alkyl)2 such as NMe2.
145. The compound of any one of claims 2-139, wherein one occurrence of RcA is Ci-4 alkoxy optionally substituted with C1-4 alkoxy or C1-4 haloalkoxy, such as wherein one occurrence of RcA is OMe or OCH2CH2OMe.
146. The compound of any one of claims 2-139, wherein one occurrence of RcA isC1-4 haloalkoxy, such as -OCH2CF3.
147. The compound of any one of claims 2-139, wherein one occurrence of RcA isC1-4 thioalkoxy, such as -SCH3.
148. The compound of any one of claims 2-139, wherein one occurrence of RcA is C1-6 alkyl, such as methyl; or wherein one occurrence of RcA is C1-6 alkyl substituted with from 1-6 independently selected halo, such as -CF3.
149. The compound of any one of claims 2-139, wherein one occurrence of RcA is Ci -6 alkyl substituted with Ra, such as C1-6 alkyl substituted with C1-3 alkoxy or
C(=O)NR’R”, such as wherein one occurrence of RcA is
Figure imgf000279_0001
, or
Figure imgf000279_0002
150. The compound of any one of claims 2-139, wherein one occurrence of RcA is halo, such as -F.
151. The compound of any one of claims 2-139, wherein one occurrence of RcA is -OH.
152. The compound of any one of claims 2-139, wherein one occurrence of RcA is C(=O)NR’R”, such as C(=O)NHMe.
153. The compound of any one of claims 1-151, wherein Rlaand Rlb are both H.
154. The compound of any one of claims 1-151, wherein R2a and R2b are both H.
155. The compound of any one of claims 1-151, wherein from 1-2 of R2a and R2b is an independently selected substituent that is other than H.
156. The compound of any one of claims 1-151 or 155, wherein one of R2a and R2b, such as R2a, is a substituent that is other than H.
157. The compound of any one of claims 1-151 or 155-156, wherein one of R2a and R2b, such as R2a, is Rb.
158. The compound of any one of claims 1-151 or 155-157 wherein one of R2a and R2b, such as R2a, is C1-6 alkyl, which is optionally substituted with from 1-6 Ra.
159. The compound of any one of claims 1-151 or 155-157, wherein one of R2a and R2b, such as R2a, is C1-3 alkyl, such as methyl or ethyl.
160. The compound of any one of claims 156-159, wherein the other of R2a and R2b, such as R2b, is H.
161. The compound of any one of claims 1-160, wherein R3a and R3b are both H.
162. The compound of any one of claims 1-160, wherein from 1-2 of R3a and R3b is an independently selected substituent that is other than H.
163. The compound of any one of claims 1-160 or 162, wherein one of R3a and R3b, such as R3a, is a substituent that is other than H.
164. The compound of any one of claims 1-160 or 162-163, wherein one of R3a and R3b, such as R3a, is Rb.
165. The compound of any one of claims 1-160 or 162-163, wherein one of R3a and R3b, such as R3a, is C1-6 alkyl which is optionally substituted with from 1-6 Ra.
166. The compound of any one of claims 1-160 or 162-164, wherein one of R3a and R3b, such as R3a, is C1-3 alkyl, such as methyl or ethyl.
167. The compound of any one of claims 1-160 or 162-164, wherein one of R3a and R3b, such as R3a, is C1-3 alkyl substituted with from 1-3 independently selected halo.
168. The compound of any one of claims 1-160, 162-165, or 167, wherein one of R3a and R3b, such as R3a, is -CH2F, -CHF2, -CF3, -CH2CHF2, or -CH2CH2F.
169. The compound of any one of claims 1-160 or 162-165, wherein one of R3a and R3b, such as R3a, is C1-3 alkyl substituted with C a1l-k4oxy, C h1a-4loalkoxy, or NReRf.
170. The compound of any one of claims 1-160, 162-165, or 169, wherein one of R3a and R3b, such as R3a, is -CH2OMe, -CH2CH2OMe, -CH(Me)CH2OMe, - CH2CH(Me)OMe, -CH2OEt, -CH2CH2OCHF2 -CH2NReRf (e g., -CH2N(CF3)Me), or - CH2CH2NReRf (e g., -CH2CH2NMe2).
171. The compound of any one of claims 1-160, 162-165 or 169-170, wherein one of R3a and R3b, such as R3a, is C1-3 alkyl substituted with C al1k-4oxy.
172. The compound of any one of claims 1-160, 162-165, or 169-171, wherein one of R3a and R3b, such as R3a, is -CH2OMe, -CH2CH2OMe, -CH(Me)CH2OMe, - CH2CH(Me)OMe, or -CH2OEt, such as -CH2OMe; such as -CH2CH2OMe; optionally the other one of R3a and R3b, such as R3b is H.
173. The compound of any one of claims 1-160 or 162-163, wherein one of R3a and R3b, such as R3a, is Rg or -(Lg)g-Rg.
174. The compound of any one of claims 1-160, 162-163, or 173, wherein one of R3a and R3b, such as R3a, is selected from the group consisting of: heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc; and
C3-6 cycloalkyl optionally substituted with from 1-4 Rc.
175. The compound of any one of claims 1-160, 162-163, or 173-174, wherein one of R3a and R3b, such as R3a, is selected from the group consisting of: cyclopropyl, cyclobutyl, oxetanyl, and azetidinyl, each of which is optionally substituted with from 1-2 substituents independently selected from the group consisting of: C1-3 alkyl and halo, wherein the ring nitrogen of the azetidinyl is optionally substituted with Rd.
176. The compound of any one of claims 1-160, 162-163, or 173-174, wherein one of R3a and R3b, such as R3a, is -(C1-3 alkylene)-Rg or -(C1-3 alkylene)-O-Rg, and optionally the Rg group of R3a or R3b is:
C3-6 cycloalkyl optionally substituted with from 1-4 Rc, or heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
177. The compound of any one of claims 1-160, 162-163, or 173, or 176, wherein one of R3a and R3b, such as R3a, is -CH2-Rg, -CH2CH2Rg, or -CH2-O-Rg, wherein the Rg group of R3a or R3b is:
C3-6 cycloalkyl optionally substituted with from 1-4 Rc, or heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
178. The compound of any one of claims 1-160, 162-163, or 173-174, or 176-
177, wherein one of R3a and R3b, such as R3a, is -CH2-Rg, -CH2CH2Rg, or -CH2-O-Rg, wherein the Rg group of R3a or R3b is selected from the group consisting of: cyclopropyl, cyclobutyl, oxetanyl, 1 ,4-dioxanyl, and azetidinyl, each of which is optionally substituted with from 1-2 substituents independently selected from the group consisting of: C1-3 alkyl and halo, wherein the ring nitrogen of the azetidinyl is optionally substituted with Rd.
179. The compound of any one of claims 1-160, 162-163, or 173-174, or 176-
178, wherein one of R3a and R3b, such as R3a, is selected from the group consisting of:
Figure imgf000283_0001
180. The compound of any one of claims 1-160, 162-163, wherein one of R3a and R3b, such as R3a, is-(Lg)g-Rw.
181. The compound any one of claim 1-160, 162-163 or 180, wherein one of R3a and R3b, such as R3a, is -(C1-3 alkylene)-Rw ; optionally one of R3a and R3b, such as R3a, is -CH2-RW, or -CH2CH2-RW
182. The compound of claim 1-160, 162- 163 or 180- 181 , wherein the Rw group of R3a or R3b is: C(=O)-CH=CH2, or -NHC(=O)-CH=CH2.
183. The compound of any claims of 1-160, 162-163 or 180-182, wherein one of
Figure imgf000283_0002
184. The compound of any one of claims 1-160, or 162-163, wherein one of R3a and R3b, such as R3a, is (Lg)g-Rg2-Rw
185. The compound of any one of claims 1-160, 162-163, 180 or 184, wherein one of R3a and R3b, such as R3a, is -(C1-3 alkylene)-Rg2-Rw, and optionally one of R3a and R3b, such as R3a, is -CH2-Rg2-Rw, or -CH2CH2-Rg2-Rw
186. The compound of any one of claims 1-160, 162-163, 180 or 184, wherein the Rg2 group
Figure imgf000284_0001
Figure imgf000284_0002
, wherein the waveline represents the point of attachment to Lg (e.g., -CH2- or - CH2CH2-) and the asterisk represents the point of attachment to Rw; and wherein the Rw group of R3a or R3b is -C(=O)-CH=CH2, or -NHC(=O)-CH=CH2.
187. The compound of any one of claims 1-160, 162-163, or 180-182, wherein one of R3a and R3b, such as R3a, is -CH2-Rg2-Rw, and wherein the Rg2 group of R3a or R3b
Figure imgf000284_0003
wherein the waveline represents the point of attachment to Lg (e. g. , -CH2- or -CH2CH2-) and the asterisk represents the point of attachment to Rw; and wherein the Rw group of R3a or R3b is - C(=O)-CH=CH2, or -NHC(=O)-CH=CH2.
188. The compound of any one of claims 1-160, 162-163, or 180-183, wherein
Figure imgf000285_0001
189. The compound of any one of claims 1-160 or 163-188, wherein the other of R3a and R3b is -H.
190. The compound of any one of claims 1-160 or 163-188, wherein the other of R3a and R3b is C1-3 alkyl, such as methyl; or wherein the other of R3a and R3b is halo, such as -F.
191. The compound of any one of claims 1-160, wherein R3a and R3b, together with the Ring B ring atom to which each is attached, form a fused saturated or unsaturated ring of 3-12 ring atoms;
• wherein from 0-2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated or unsaturated ring of 3-12 ring atoms is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rw.
192. The compound of any one of claims 1-160 or 191, wherein R3a and R3b, together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-8 ring atoms; • wherein from 0-2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated ring of 4-8 ring atoms is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rw.
193. The compound of any one of claims 1-160 or 191-193, whereinR3a andR3b, together with the Ring B ring atom to which each is attached, form:
Figure imgf000286_0001
, which is optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc, wherein: pl and p2 are independently 0, 1, or 2;
Rz is H, Rd, C(=0)-W, or S(O)2W; and cc represents the point of attachment to C(R2aR2b).
194. The compound of any one of claims 1-160 or 191-193, whereinR3a andR3b, together with the Ring B ring atom to which each is attached, form
Figure imgf000286_0002
, wherein Rz is H, Rd, C(=0)-W, or S(O)2W; and cc represents the point of attachment to C(R2aR2b).
195. The compound of any one of claims 1-160 or 191-193, whereinR3a andR3b, together with the Ring B ring atom to which each is attached, form a fused ring selected
Figure imgf000286_0003
Figure imgf000287_0001
W; and cc represents the point of attachment to C(R2aR2b).
196. The compound of any one of claims 194-195, wherein Rz is H.
197. The compound of any one of claims 194-195, wherein Rz is Rd.
198. The compound of any one of claims 194-195 or 197, wherein Rz is C1-6 alkyl optionally substituted with from 1-3 independently selected Ra.
199. The compound of any one of claims 194-195, wherein Rz is C(=O)-W or S(O)2W.
200. The compound of any one of claims 194-195 or 199, wherein W is C2-4 alkenyl.
201. The compound of any one of claims 194-195 or 199-200, wherein Rz is C(=O)-CH2=CH2.
202. The compound of any one of claims 1-160, wherein R3a and R3b together with the Ring B ring atom to which each is attached form a fused C3-6 cycloalkyl, wherein the fused C3-6 cycloalkyl is optionally substituted with from 1-2 Rc.
203. The compound of any one of claims 1-160 or 202, wherein R3a and R3b together with the Ring B ring atom to which each is attached form a fused cyclopropyl or cyclobutyl.
204. The compound of any one of claims 1-160, wherein R3a and R3b together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-6 ring atoms; wherein from 1-2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and wherein the fused saturated ring of 4-6 ring atoms is optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc.
205. The compound of any one of claims 1-160 or 204, wherein R3a and R3b, together with the Ring B ring atom to which each is attached, form
Figure imgf000288_0001
Figure imgf000288_0002
206. The compound of any one of claims 1-153, wherein one of R2a and R2b (such as R2a) and one of R3a and R3b (such as R3a) taken together with the Ring B ring atoms to which each is attached, form a fused saturated or unsaturated ring of 3-12 ring atoms; wherein from 0-2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and wherein the fused saturated or unsaturated ring of 3-12 ring atoms is optionally substituted with from 1 -4 substituents independently selected from the group consisting of oxo and Rc.
207. The compound of any one of claims 1-153 or 206, wherein one of R2a and R2b (such as R2a) and one of R3a and R3b (such as R3a) taken together with the Ring B ring atoms to which each is attached, form a fused saturated ring of 3-8 ring atoms; wherein from 0-2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and wherein the fused saturated ring of 3-8 ring atoms is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
208. The compound of any one of claims 1-156 or 206-207, wherein one of R2a and R2b (such as R2a) and one of R3a and R3b (such as R3a) taken together with the Ring B ring atoms to which each is attached, form a fused C3-6 cycloalkyl which is optionally substituted with from 1-2 Rc.
209. The compound of any one of claims 1-156 or 206-208, wherein one of R2a and R2b (such as R2a) and one of R3a and R3b (such as R3a) taken together with the Ring B ring atoms to which each is attached, form a fused cyclopropyl.
210. The compound of any one of claims 1-156 or 206-208, wherein one of R2a and R2b (such as R2a) and one of R3a and R3b (such as R3a) taken together with the Ring B ring atoms to which each is attached, form a fused cyclobutyl.
211. The compound of any one of claims 206-210, wherein the other of R2a and R2b and the other of R3a and R3b are each H.
212. The compound of any one of claims 1-153 or 211, wherein the other one of R3a and R3b is R" or -(Lg)g-Rg.
213. The compound of any one of claims 1-153, 211 or 212, wherein the other one of R3a and R3b is -(Lg)g-Rg.
214. The compound of any one of claims 1-153, or 211-213, wherein the other one of R3a and R3b is -(C1-3 alkylene)-Rg or -(C1-3 alkylene)-O-Rg, and optionally the Rg group of R3a or R3b is:
C3-6 cycloalkyl optionally substituted with from 1-4 Rc, or heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
215. The compound of any one of claims 1-153, or 211-214, wherein the other one of R3a and R3b, such as R3a, is -CH2-Rg, -CH2CH2Rg, or -CH2-O-Rg, wherein the Rg group of R3a or R3b is:
C3-6 cycloalkyl optionally substituted with from 1-4 Rc, or heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
216. The compound of any one of claims 1-153, or 211-215, wherein the other one of R3a and R3b, such as R3a, is -CH2-Rg, -CH2CH2Rg, or -CH2-O-Rg, wherein the Rg group of R3a or R3b is selected from the group consisting of: cyclopropyl, cyclobutyl, oxetanyl, 1 ,4-dioxanyl, and azetidinyl, each of which is optionally substituted with from 1-2 substituents independently selected from the group consisting of: C1-3 alkyl and halo, wherein the ring nitrogen of the azetidinyl is optionally substituted with Rd.
217. The compound of any one of claims 1-156, or 211-216, wherein the other one of R3a and R3b, such as R3a, is selected from the group consisting of:
Figure imgf000290_0001
Figure imgf000291_0001
218. The compound of any one of claims 1-153, wherein Rla, Rlb, R2a, and R2b are each H; one of R3a and R3b, such as R3a, is C1-3 alkyl optionally substituted with from 1-3 Ra; and the other of R3a and R3b is H, optionally each Ra substituent present in R3a or R3b is independently selected from the group consisting of: halo, C1-4 alkoxy, and CM haloalkoxy.
219. The compound of any one of claims 1-196, wherein Rla, Rlb, R2a, and R2b are each H; one of R3a and R3b, such as R3a, is C1-3 alkyl optionally substituted with from C1-4 alkoxy; optionally one of R3a and R3b, such as R3a, is -CTTCTT-OMe; and the other of R3a and R3b is H.
220. The compound of any one of claims 1-153, wherein Rla, Rlb, R2a, and R2b are each H; and R3a and R3b are independently selected C1-3 alkyl.
221. The compound of any one of claims 1-153, wherein Rla, Rlb, R2a, and R2b are each H; one of R3a and R3b, such as R3a, is -Rg, -(C1-3 alkylene)-Rg, or -(C1-3 alkylene)- O-Rg, optionally wherein the Rg group of R3a or R3b is:
C3-6 cycloalkyl optionally substituted with from 1-4 Rc, or heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc; and the other of R3a and R3b is H.
222. The compound of any one of claims 1-153, wherein Rla, Rlb, R2a, and R2b are each H; and R3a and R3b taken together with the Ring B ring carbon atom to which each is attached form a fused C3-6 (such as C3 or C4) cycloalkyl, wherein the fused cycloalkyl ring is optionally substituted with from 1 -2 Rc.
223. The compound of any one of claims 1-153, wherein Rla, Rlb, R2a, and R2b are each H; and R3a and R3b together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-6 ring atoms;
• wherein from 1-2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated ring of 4-6 ring atoms is optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc.
224. The compound of any one of claims 1-153, wherein Rla, Rlb is H; one of R2a and R2b (such as R2a) and one of R3a and R3b (such as R3a) taken together with the Ring B ring atoms to which each is attached, form a fused C3-6 (such as C3 or C4) cycloalkyl which is optionally substituted with from 1-2 Rc; and the other of R2a and R2b and the other of R3a and R3b are each H.
225. The compound of any one of claims 1-153, wherein Rla, Rlb, R2a, R2b, R3a, and R3b are each H.
226. The compound of any one of claims 1-225, wherein R4 is H.
227. The compound of any one of claims 1 -226, wherein Ring A is
Figure imgf000293_0001
, wherein each RcB is an independently selected Rc; and ml is 0, 1, 2, 3, or 4.
228. The compound of claim 227, wherein ml is 1, 2, or 3.
229. The compound of claims 227 or 228, wherein ml is 1 or 2, such as 2.
RCB
230. The compound of any one of claims 1 -229, wherein Ring A is
Figure imgf000293_0002
Figure imgf000293_0003
wherein each RcB is an independently selected Rc.
231. The compound of any one of claims 1 or 227-230, wherein Ring A is selected from the group consisting of:
Figure imgf000293_0004
Figure imgf000293_0005
independently selected Rc.
232. The compound of any one of claims 227-231, wherein each RcB is independently selected from the group consisting of: -halo, such as -Cl and -F; -CN; CM alkoxy;C1-4 haloalkoxy; C1-4 alkyl; and C1-4 alkyl substituted with from 1-6 independently selected halo. 233. The compound of any one of claims 1 -231 , wherein Ring A is
Figure imgf000294_0001
, wherein RcB1 is Rc; and RcB2 is H or Rc, optionally wherein RcB1 and RcB2 are each independently selected from the group consisting of: -halo, such as -Cl and -F; -CN; CM alkoxy;C1-4 haloalkoxy; C1-4 alkyl; and C1-4 alkyl substituted with from 1-6 independently selected halo.
234. The compound of claim 233, wherein RcB1 is halo, such as -F or -Cl, such as -F.
235. The compound of claim 234, wherein RcB1 is C1-4 alkyl or C1-4 alkyl substituted with from 1-6 independently selected halo, such as wherein RcB1 is methyl, - CHF2, or -CF3.
236. The compound of any one of claims 233-235, wherein RcB2 is selected from the group consisting of: halo; -CN; C1-4 alkoxy; C1-4 haloalkoxy; C1-4 alkyl; and C1- a4lkyl substituted with from 1-6 independently selected halo.
237. The compound of any one of claims 233-235, wherein RcB2 is C1-4 alkoxy orC1-4 haloalkoxy.
238. The compound of any one of claims 233-237, wherein RcB2 is selected from the group consisting of cyano; C1-4 alkyl; and C1-4 alkyl substituted with from 1-6 independently selected halo, such as wherein RcB2 is cyano, methyl, ethyl, -CHF2, -CF3, or -CH2CHF2.
239. The compound of any one of claims 1-231, or 233-238, wherein Ring A is
Figure imgf000294_0002
240. The compound of any one of claims 1 -231 , wherein Ring A is heteroaryl including from 5-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc.
241. The compound of any one of claims 1-231 or 240, wherein Ring A is bicyclic heteroaryl including from 9-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc.
242. The compound of any one of claims 1-231 or 240-241, wherein Ring A is selected from the group consisting of:
Figure imgf000295_0001
Figure imgf000295_0002
each of which is further optionally substituted with Rc.
243. The compound of claim 1, wherein the compound is a compound of Formula (I-a):
Figure imgf000295_0003
or a pharmaceutically acceptable salt thereof, wherein: each RCA is an independently selected Rc; n is 0, 1, or 2; and
Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
244. The compound of claim 243, wherein Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
245. The compound of claims 243 or 244, wherein
Figure imgf000296_0001
the group consisting of:
Figure imgf000296_0002
Figure imgf000296_0003
each further optionally substituted with RcA, wherein each
RCA is an independently selected Rc.
246. The compound of claims 243 or 244, wherein
Figure imgf000296_0004
Figure imgf000296_0005
Figure imgf000297_0001
each further optionally substituted with RcA, wherein each RcA is an independently selected Rc.
247. The compound of any one of claims 243-246, wherein
Figure imgf000297_0002
, wherein RcA is an independently selected Rc. pound of any one of claims 243-246, wherein
Figure imgf000297_0004
Figure imgf000297_0003
wherein each RcA is an independently selected Rc.
249. The compound of any one of claims 243-246, wherein
Figure imgf000298_0001
selected from the group consisting of:
Figure imgf000298_0002
wherein: each occurrence of RcA is independently selected from the group consisting of: halo; NReRf; C1-4 alkoxy; C1- h4aloalkoxy; C1-3 alkyl; C1-3 alkyl substituted with from 1-3 independently selected halo; C1-3 alkyl substituted with C1-4 alkoxy; and C1-4 alkoxy substituted with C1-4 alkoxy; such as wherein each occurrence of RcA is independently selected from the group consisting of: C1-4 alkoxy; C1-4 haloalkoxy; C1-3 alkyl; and C1-3 alkyl substituted with from 1-3 independently selected halo.
250. The compound of claim 243, wherein Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
251. The compound of claims 243 or 250, wherein
Figure imgf000298_0003
the group consisting of:
Figure imgf000298_0004
Figure imgf000299_0001
is an independently selected Rc.
252. The compound of claim 1, wherein the compound is a compound of
Formula (I-b):
Figure imgf000299_0002
Formula (I-b) or a pharmaceutically acceptable salt thereof, wherein: each RCA is an independently selected Rc; n is 0 or 1 ; and
Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
253. The compound of claim 252, wherein Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
254. The compound of claims 252 or 253, wherein
Figure imgf000300_0001
selected from
Figure imgf000300_0002
RCA is an independently selected Rc.
255. The compound of claim 252, wherein Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
256. The compound of claims 252 or 255, wherein
Figure imgf000300_0003
the group consisting of:
Figure imgf000300_0004
Figure imgf000300_0005
each further optionally substituted with RcA, wherein each RcA is an independently selected Rc.
257. The compound of claim 1, wherein the compound is a compound of Formula (I-c):
Figure imgf000301_0001
or a pharmaceutically acceptable salt thereof; wherein n is 0, 1, or 2; each RCA is an independently selected Rc; and
Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
258. The compound of claim 257, wherein Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
259. The compound of claims 257 or 258, wherein
Figure imgf000302_0001
selected from the group consisting of:
Figure imgf000302_0002
each further optionally substituted with RcA, wherein each RcA is an independently selected Rc.
Figure imgf000302_0003
wherein each RcA is an independently selected Rc.
Figure imgf000303_0001
262. The compound of claim 261, wherein Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
Figure imgf000303_0002
, each further optionally substituted with RcA, wherein each RcA is an independently selected Rc.
264. The compound of claim 1, wherein the compound is a compound of Formula (I-d):
Figure imgf000304_0001
Formula (I-d) or a pharmaceutically acceptable salt thereof; wherein n is 0 or 1 ; each RCA is an independently selected Rc; and
Ring D is a partially unsaturated or aromatic ring including from 5-6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1 -2 RcA.
265. The compound of claim 264, wherein Ring D is a partially unsaturated or aromatic ring including 6 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA.
266. The compound of claims 264 or 265, wherein
Figure imgf000305_0001
selected from
Figure imgf000305_0002
each further optionally substituted with RcA, wherein each RcA is an independently selected Rc. 267. The compound of claim 264, wherein Ring D is a partially unsaturated or aromatic ring including 5 ring atoms, wherein from 0-2 of the ring atoms are heteroatoms each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, wherein Ring D is optionally substituted with from 1-2 RcA. 268. The compound of claim 1, wherein the compound of Formula (I) is a compound of Formula (I-e)
Figure imgf000305_0003
Formula (I-e), or a pharmaceutically acceptable salt thereof, wherein Ring D is heterocyclylene including from 3-10 ring atoms, wherein from 0-2 ring atoms (in addition to the ring nitrogen atom bonded to Rw) are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclylene is optionally substituted with from 1-3 substituents each independently selected from the group consisting of: oxo and -Rc.
269. The compound of claim 268, wherein Rw is -Lw-W; and Lw is C(=O).
270. The compound of claims 268 or 269, wherein W is C2-6 alkenyl or C2-6 alkynyl optionally substituted with from 1-3 Ra and further optionally substituted with Rg, wherein W is attached to Lw via an sp2 or sp hybridized carbon atom.
271. The compound of any one of claims 268-270, wherein W is CH=CH2,
Figure imgf000306_0001
272. The compound of any one of claims 268-271, wherein Ring
Figure imgf000306_0002
which is optionally substituted with from 1-2 Rc, wherein xl and x2 are each independently 0, 1, or 2.
273. The compound of claim 272, wherein xl is 0.
274. The compound of any one of claims 268-273, wherein Ring D is selected
Figure imgf000306_0003
275. The compound of any one of claims 268-274, wherein n is 0.
276. The compound of any one of claims 268-274, wherein n is 1 or 2, such as wherein n is i.
277. The compound of any one of claims 268-274 or 276, wherein
Figure imgf000307_0001
278. The compound of any one of claims 268-274 or 276, wherein R7 is NReRf, such as NH2, NH(CI-3 alkyl), or N(CI-3 alkyl)2, such as wherein R7 is NH2.
279. The compound of any one of claims 243-278, wherein each occurrence of RCA is independently selected from the group consisting of: halo; cyano; Ci-io alkyl which is optionally substituted with from 1-6 independently selected Ra; C1-4 alkoxy optionally substituted with C1-4 alkoxy or C1-4 haloalkoxy; C1-4 haloalkoxy; -S(O)i-2(C1-4 alkyl); - NReRf; -OH; -S(O)I-2NR’R”; -Ci-4 thioalkoxy; -C(=0)(Ci-io alkyl); -C(=O)O(Ci-4 alkyl); -C(=O)OH; and -C(=O)NR’R”
280. The compound of any one of claims 243-279, wherein one occurrence of RCA IS -NReRf
281. The compound of any one of claims 243-280, wherein one occurrence of RCA IS -NH2.
282. The compound of any one of claims 243-281, wherein one occurrence of RCA is -NH(CI-6 alkyl), wherein the C1-6 alkyl is optionally substituted with from 1-3 substituents each independently selected from the group consisting of NR’R”, -OH, C1-6 alkoxy, C1-6 haloalkoxy, and halo, such as wherein one occurrence of RcA is -NHMe, - NHCH2CF3, -NHCH2CH2OH, or -NHiPr.
283. The compound of any one of claims 243-282, wherein one occurrence of RCA is -NHC(=0)CI-4 alkyl, such as NHC(=0)CH3; or wherein one occurrence of RcA is N(CI-3 alky 1)2 such as NMe2.
284. The compound of any one of claims 243-278, wherein one occurrence of RCA is C1-4 alkoxy optionally substituted with C1-4 alkoxy orC1-4 haloalkoxy, such as wherein one occurrence of RcA is OMe or OCTFCTFOMe; or wherein one occurrence of RCA is C1-4 haloalkoxy, such as -OCH2CF3.
285. The compound of any one of claims 243-278, wherein one occurrence of RCA is C1-4 thioalkoxy, such as -SCH3.
286. The compound of any one of claims 243-278, wherein one occurrence of RCA is C1-6 alkyl, such as methyl; or wherein one occurrence of RcA is C1-6 alkyl substituted with from 1-6 independently selected halo, such as -CF3.
287. The compound of any one of claims 243-278, wherein one occurrence of
RCA is C1-6 alkyl substituted with Ra, such as C1-6 alkyl substituted with C1-3 alkoxy or
C(=O)NR’R”, such as wherein one occurrence of RcA is
Figure imgf000308_0001
, or
Figure imgf000308_0002
288. The compound of any one of claims 243-278, wherein one occurrence of RCA is halo, such as -F.
289. The compound of any one of claims 243-278, wherein one occurrence of RCA IS -OH.
290. The compound of any one of claims 243-278, wherein one occurrence of RCA IS C(=O)NR’R”, such as C(=O)NHMe.
291. The compound of any one of claims 257-278, wherein X1 is -(X2)m-L1-R5, wherein:
• m is 0 or 1;
• X2 is -N(RN)- or -O-;
• L1 is a bond or C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is - R"
292. The compound of any one of claims 257-278, wherein X1 is -X^lAR5, wherein:
Figure imgf000309_0001
• L1 is a bond or C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is - R"
293. The compound of any one of claims 257-278, wherein X1 is -X^lAR5, wherein:
Figure imgf000309_0002
• L1 is a bond or C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is - R"
294. The compound of any one of claims 279-293, wherein R5 is phenyl optionally substituted with from 1-4 Rc, such as wherein R5 is phenyl optionally substituted with from 1-2 independently selected halo, such as -F.
295. The compound of any one of claims 279-293, wherein R5 is heteroaryl including 6 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), and N(Rd), and wherein the heteroaryl is optionally substituted with from 1-4 Rc, such as wherein R5 is
Figure imgf000310_0001
Figure imgf000310_0002
296. The compound of any one of claims 279-293, wherein R5 is heteroaryl including 5 ring atoms, wherein from 1-4, such as 2-4, ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc, such as wherein R5 is
Figure imgf000310_0003
297. The compound of any one of claims 279-293, wherein R5 is C3-10 cycloalkyl, such as C3-6 cycloalkyl, optionally substituted with from 1-4 Rc, such as wherein R5 is cyclopropyl.
298. The compound of any one of claims 279-293, wherein R5 is heterocyclyl including from 4-8, such as 4-6, ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc, such as wherein R5 is
Figure imgf000310_0004
299. The compound of any one of claims 257-278, wherein X1 is -(X2)m-L1-R5, wherein:
• m is 0 or 1; • X2 is -N(RN)- or -O-;
• L1 is a bond or C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is R"2-Rv
300. The compound of claim 299, wherein the -Rg2 group present in R5 is 1,3- phenylene or 1 ,4-phenylene, each optionally substituted with from 1-4 Rc, such as wherein
Figure imgf000311_0001
wherein bb is the point of attachment to RY.
301. The compound of claims 299 or 300, wherein the RY group present in R5 is
— Rg.
302. The compound of any one of claims 299-301, wherein the RY group present in R5 is heterocyclyl including from 4-8, such as 4-6, ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1- 4 substituents independently selected from the group consisting of oxo and Rc, such as wherein
Figure imgf000311_0002
303. The compound of any one of claims 257-278, wherein X1 is -X^lAR5, wherein:
• X2 is -N(RN)-, -O-, -N(RN)C(=O)-*, -N(RN)S(O)2-, -N(RN)C(=O)O-*, or -N(RN)C(=O)N(RN)-*;
• L1 is C1-6 alkylene optionally substituted with from 1-3 Ra; and
• R5 is H, halo, C1-6 alkoxy optionally substituted with from 1-3 Ra, or -OH.
304. The compound of claim 303, wherein R5 is H.
305. The compound of claim 303, wherein R5 is halo, such as -F.
306. The compound of claim 303, wherein R5 is C1-6 alkoxy optionally substituted with from 1-3 Ra, such as wherein R5 is C1-3 alkoxy such as methoxy.
307. The compound of claim 303, wherein R5 is -OH.
308. The compound of any one of claims 257 or 294-302, wherein m is 0.
309. The compound of any one of claims 257 or 294-302, wherein m is 1.
310. The compound of any one of claims 257, 294-302 or 309, wherein X2 is - N(RN)-, such as N(H).
311. The compound of any one of claims 257, 294-302, or 309, wherein X2 is - O-.
312. The compound of any one of claims 257, 294-302, or 309, wherein X2 is - N(RN)C(=O)-*, such as -N(H)C(=O)-*.
313. The compound of any one of claims 257, 294-302, or 309, wherein X2 is - N(RN)S(O)2-, such as -N(H)S(O)2-*.
314. The compound of any one of claims 257, 294-302, or 309, wherein X2 is - N(RN)C(=O)O-*, or -N(RN)C(=O)N(RN)-*, such as -N(H)C(=O)O-* or - N(H)C(=O)N(H)-*.
315. The compound of any one of claims 294-302, wherein X2 is
Figure imgf000312_0001
316. The compound of any one of claims 294-302, wherein X2 is
Figure imgf000313_0001
.
317. The compound of any one of claims 257-278, wherein L1 is a bond.
318. The compound of any one of claims 257-278, wherein L1 is C1-3 alkylene, such as -CH2-, -CH2CH2-, or -CH(Me)-.
319. The compound of any one of claims 257-278, wherein L1 is branched C3-6 alkylene, such as
Figure imgf000313_0002
wherein aa is the point of attachment to R5.
320. The compound of any one of claims 257-278, wherein X1 is L'-R5, wherein L1 is C1-6 alkylene optionally substituted with from 1-3 Ra; and R5 is -L5-Rg.
321. The compound of claim 320, wherein R5 is -O-Rg.
322. The compound of claims 320-321, wherein R5 is -O-(phenyl), wherein the phenyl is optionally substituted with from 1-2 Rc.
323. The compound of any one of claims 320-322, wherein L1 is C1-3 alkylene, such as -CH2-, -CH2CH2-, or -CH(Me)-.
324. The compound of any one of claims 243-323, wherein Rla, Rlb is H.
325. The compound of any one of claims 243-324, wherein R2a and R2b are both H.
326. The compound of any one of claims 243-325, wherein R2a is a substituent that is other than H.
327. The compound of any one of claims 243-324 or 326, wherein R2a is C1-6 alkyl which is optionally substituted with from 1-6 Ra, such as wherein R2a is C1-3 alkyl, such as methyl or ethyl.
328. The compound of claims 326 or 327, wherein R2b is H.
329. The compound of any one of claims 243-328, wherein R3a and R3b are both H.
330. The compound of any one of claims 243-328, wherein R3a is a substituent that is other than H.
331. The compound of any one of claims 243-328 or 330, wherein R3a is C1-6 alkyl which is optionally substituted with from 1-6 Ra, such as wherein R3a is C1-3 alkyl, such as methyl or ethyl.
332. The compound of any one of claims 243-308 or 333, wherein R3a is C1-3 alkyl substituted with from 1-3 independently selected halo, such as wherein R3a is -CH2F, -CHF2, -CF3, -CH2CHF2, or -CH2CH2F.
333. The compound of any one of claims 243-308 or 330, wherein R3a is C1-3 alkyl substituted with C1-4 alkoxy, C1-4 haloalkoxy, or NReRf, such as wherein R3a is - CH2OMe, -CH2CH2OMe, -CH(Me)CH2OMe, -CH2CH(Me)OMe, -CH2OEt, -CH2NReRf (e g., -CH2N(CF3)Me), or -CH2CH2NReRf (e g., -CH2CH2NMe2).
334. The compound of any one of claims 243-328 or 330, wherein R3a is selected from the group consisting of: heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc; and
C3-6 cycloalkyl optionally substituted with from 1-4 Rc.
335. The compound of any one of claims 243-328 or 330, wherein R3a is -(C1-3 alkylene)-Rg or -(C1-3 alkylene)-O-Rg, and optionally the Rg group of R3a is:
C3-6 cycloalkyl optionally substituted with from 1-4 Rc, or heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc.
336. The compound of claims 243-328, 330 or 335, wherein R3a is -CH2-Rg, or -CH2CH2R", wherein Rg is 1 ,4-dioxanyl.
337. The compound of any one of claims 243-328 or 330, wherein R3a is-(Lg)g- Rw.
338. The compound of any one of claims 243-328, 330 or 337, wherein R3a is- CH2CH2-RW, wherein the Rw group is C(=O)-CH=CH2, or -NHC(=O)-CH=CH2.
339. The compound of any one of claims 243-328, 330, or 337-338, wherein R3a
Figure imgf000315_0001
340. The compound of any one of claims 243-328 or 330, wherein R3a is -(Lg)g-
Rg2-Rw
341. The compound of any one of claims 243-304, 330 or 340 , wherein R3a is -
CH2-Rg2-Rw, wherein the Rg2 group
Figure imgf000315_0002
Figure imgf000315_0003
and the asterisk represents the point of attachment to Rw; and optionally the Rw group is C(=O)-CH=CH2.
342. The compound of any one of claims 243-328, 330 or 340-341, wherein R3a
Figure imgf000316_0001
343. The compound of claims 243-328 or 340-342, wherein R3b is H.
344. The compound of any one of claims 243-328 or 340-342, wherein R3b is Ci- 3 alkyl, such as methyl.
345. The compound of any one of claims 243-328, wherein R3a and R3b, together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-8 ring atoms;
• wherein from 0-2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated ring of 4-8 ring atoms is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo, Rc, and Rw.
346. The compound of any one of claims 243-328 or 345, wherein R3a and R3b, together with the Ring B ring atom to which each is attached, form:
Figure imgf000316_0002
is optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc, wherein: pl and p2 are independently 0, 1, or 2;
Rz is H, Rd, C(=0)-W, or S(O)2W; and cc represents the point of attachment to C(R2aR2b). 347. The compound of any one of claims 243-328 or 345-346, wherein R3a and
R3b, together with the Ring B ring atom to which each is attached, form
Figure imgf000317_0001
Figure imgf000317_0002
rZ , wherein Rz is H, Rd, C(=O)-W, or S(O)2W; and cc represents the point of attachment to C(R2aR2b). 348. The compound of any one of claims 243-328 or 345-346, wherein R3a and
R3b, together with the Ring B ring atom to which each is attached, form a fused ring selected from the group consisting of:
Figure imgf000317_0003
Figure imgf000317_0004
S(0)2W; and cc represents the point of attachment to C(R2aR2b).
349. The compound any one of claims 346-348, wherein Rz is H.
350. The compound of any one of claims 346-348, wherein Rz is C1-6 alkyl optionally substituted with from 1-3 independently selected Ra.
351. The compound of any one of claims 346-348, wherein Rz is C(=0)-W or S(O)2W, optionally wherein W is C2-4 alkenyl.
352. The compound of any one of claims 243-328, wherein R3a and R3b together with the Ring B ring atom to which each is attached form a fused C3-6 cycloalkyl, wherein the fused C3-6 cycloalkyl is optionally substituted with from 1-2 Rc.
353. The compound of any one of claims 243-328, wherein R3a and R3b together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-6 ring atoms;
• wherein from 1 -2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated ring of 4-6 ring atoms is optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc
354. The compound of any one of claims 243-324, wherein R2a and R3a taken together with the Ring B ring atoms to which each is attached, form a fused C3-6 (e.g., C3 or C4) cycloalkyl which is optionally substituted with from 1-2 Rc.
355. The compound of any one of claims 243-325, wherein R2b and R3b are each H.
356. The compound of any one of claims 243-325, wherein Rla, Rlb, R2a, and R2b are each H; R3a is C1-3 alkyl optionally substituted with from 1-3 Ra; and R3b is H, optionally each Ra substituent present in R3a is independently selected from the group consisting of: halo, C1-4 alkoxy, and C1-4 haloalkoxy.
357. The compound of any one of claims 243-325 or 356, wherein Rla, Rlb, R2a, and R2b are each H; and R3a and R3b are independently selected C1-3 alkyl.
358. The compound of any one of claims 243-325, wherein Rla, Rlb, R2a, and R2b are each H; R3a, is -Rg, -(C1-3 alkylene)-Rg, or -(C1-3 alkylene)-O-Rg, optionally wherein the Rg group of R3a is:
C3-6 cycloalkyl optionally substituted with from 1-4 Rc, or heterocyclyl including from 4-6 ring atoms, wherein from 1-3 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heterocyclyl is optionally substituted with from 1-4 substituents independently selected from the group consisting of oxo and Rc; and
R3b is H.
359. The compound of any one of claims 243-325, wherein Rla, Rlb, R2a, and R2b are each H; and R3a and R3b taken together with the Ring B ring carbon atom to which each is attached form a fused C3-6 (such as C3 or C4) cycloalkyl, wherein the fused cycloalkyl ring is optionally substituted with from 1 -2 Rc.
360. The compound of any one of claims 243-325, wherein Rla, Rlb, R2a, and R2b are each H; and R3a and R3b together with the Ring B ring atom to which each is attached, form a fused saturated ring of 4-6 ring atoms;
• wherein from 1 -2 of the ring atoms are each an independently selected heteroatom, wherein each of the independently selected heteroatoms is selected from the group consisting of N, NH, N(Rd), O, and S(0)o-2; and
• wherein the fused saturated ring of 4-6 ring atoms is optionally substituted with from 1-2 substituents independently selected from the group consisting of oxo and Rc
361. The compound of any one of claims 243-324, wherein Rla, Rlb is H; R2a and R3a taken together with the Ring B ring atoms to which each is attached, form a fused C3-6 (e.g., C3 or C4) cycloalkyl which is optionally substituted with from 1-2 Rc; and R2b and R3b are each H.
362. The compound of any one of claims 243-324, wherein Rla, Rlb is H; and R2b is H; and R3b is -(Lg)g-Rg.
363. The compound of any one of claims 243-328 or 362, wherein Rla, Rlb is H;
Figure imgf000320_0001
364. The compound of any one of claims 243-324, wherein Rla, Rlb, R2a, R2b, R3a, and R3b are each H.
365. The compound of any one of claims 243-364, wherein R4 is H.
366. The compound of any one of claims 243-365, wherein Ring A is
Figure imgf000320_0002
, wherein each RcB is an independently selected Rc; and ml is 0, 1, 2, 3, or 4.
367. The compound of claim 366, wherein ml is 1, 2, or 3, such as 1 or 2.
368. The compound of any one of claims 243-367, wherein Ring A is
Figure imgf000320_0003
369. The compound of any one of claims 243-368, wherein Ring A is
Figure imgf000321_0001
Figure imgf000321_0002
370. The compound of any one of claims 243-368, wherein Ring A is selected
Figure imgf000321_0003
independently selected Rc. 371. The compound of any one of claims 243-370, wherein each RcB is independently selected from the group consisting of: -halo, such as -Cl and -F; -CN; CM alkoxy;C1-4 haloalkoxy; C1-4 alkyl; and C1-4 alkyl substituted with from 1-6 independently selected halo. 372. The compound of any one of claims 243-365, wherein Ring A is bicyclic heteroaryl including from 9-10 ring atoms, wherein from 1-4 ring atoms are heteroatoms, each independently selected from the group consisting of N, N(H), N(Rd), O, and S(0)o-2, and wherein the heteroaryl is optionally substituted with from 1-4 Rc, such as wherein:
Ring A is selected from the group consisting of:
Figure imgf000321_0004
Figure imgf000322_0001
optionally substituted with Rc.
373. The compound of any one of claims 1-372, wherein the
Figure imgf000322_0002
moiety i
Figure imgf000322_0003
374. The compound of any one of claims 1-372, wherein the
Figure imgf000322_0004
moiety i
Figure imgf000322_0005
375. The compound of claim 1 , wherein the compound is selected from the group consisting of the compounds delineated in Table Cl, or a pharmaceutically acceptable salt thereof.
376. A pharmaceutical composition comprising a compound of any one of claims 1-375, or a pharmaceutically acceptable salt thereof, and pharmaceutically acceptable diluent or carrier.
377. A method for treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of any one of claims 1-375, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to 376.
378. A method for treating cancer in a subject in need thereof, the method comprising (a) determining that the cancer is associated with a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same; and (b) administering to the subject a therapeutically effective amount of a compound of any one of claims 1-375, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to 376.
379. A method of treating an EGFR-associated cancer in a subject, the method comprising administering to a subject identified or diagnosed as having an EGFR- associated cancer a therapeutically effective amount of a compound of any one of claims 1-375 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 376.
380. A method of treating an EGFR-associated cancer in a subject, the method comprising:
(a) determining that the cancer in the subject is an EGFR-associated cancer; and
(b) administering to the subject a therapeutically effective amount of a compound of any one of claims 1-375 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 376.
381. A method of treating a subject, the method comprising administering a therapeutically effective amount of a compound of any one of claims 1-375 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 376, to a subject having a clinical record that indicates that the subject has a dysregulation of an EGFR gene, an EGFR kinase, or expression or activity or level of any of the same.
382. The method of any one of claims 378 or 380, wherein the step of determining that the cancer in the subject is an EGFR-associated cancer includes performing an assay to detect dysregulation in an EGFR gene, an EGFR kinase protein, or expression or activity or level of any of the same in a sample from the subject.
383. The method of claim 382, further comprising obtaining a sample from the subject.
384. The method of claim 383, wherein the sample is a biopsy sample.
385. The method of any one of claims 382-384, wherein the assay is selected from the group consisting of sequencing, immunohistochemistry, enzyme-linked immunosorbent assay, and fluorescence in situ hybridization (FISH).
386. The method of claim 385, wherein the FISH is break apart FISH analysis.
387. The method of claim 385, wherein the sequencing is pyrosequencing or next generation sequencing.
388. The method of any one of claims 378, 381, and 382, wherein the dysregulation in an EGFR gene, an EGFR kinase protein, or expression or activity or level of any of the same is one or more point mutations in the EGFR gene.
389. The method of claim 388, wherein the one or more point mutations in an EGFR gene results in the translation of an EGFR protein having one or more amino acid substitutions at one or more of the following amino acid positions exemplified in Table la and lb.
390. The method of claim 389, wherein the one or more point mutations is selected from the mutations in Table la and lb (e.g., L858R, G719S, G719C, G719A, L861Q, a deletion in exon 19 and/or an insertion in exon 20).
391. The method of claim 389, wherein the one or more point mutations is an EGFR inhibitor resistance mutation (e.g., L718Q, L747S, D761Y, T790M, C797S, T854A).
392. The method of claim 389, wherein the one or more point mutations in an EGFR gene include a deletion in exon 19 of a human EGFR gene.
393. The method of claim 389, wherein the one or more mutations is an EGFR insertion in exon 20 of a human EGFR gene.
394. The method of claim 391, wherein the insertion in exon 20 of a human EGFR gene is selected from: V769_D770insX, D770_N771insX, N771_P772insX, P772_H773insX, and H773_V774insX.
395. The method of claims 391 or 392, wherein the insertion in exon 20 of a human EGFR gene is selected from: Y772_A775dup, A775_G776insYVMA, G776delinsVC, G776delinsW, V777_G778insGSP, and P780_Y781insGSP.
396. The method of any one of claims 389, 390 and 392-393, wherein the EGFR- associated cancer is selected from the group consisting of: oral cancer, oropharyngeal cancer, nasopharyngeal cancer, respiratory cancer, urogenital cancer, gastrointestinal cancer, central or peripheral nervous system tissue cancer, an endocrine or neuroendocrine cancer, a hematopoietic cancer, glioma, sarcoma, carcinoma, lymphoma, melanoma, fibroma, meningioma, brain cancer, oropharyngeal cancer, nasopharyngeal cancer, renal cancer, biliary cancer, pheochromocytomaLi-Fraumeni tumor, thyroid cancer, parathyroid cancer, pituitary tumors, adrenal gland tumors, osteogenic sarcoma tumors, breast cancer, lung cancer, head and neck cancer, prostate cancer, esophageal cancer, tracheal cancer, liver cancer, bladder cancer, stomach cancer, pancreatic cancer, ovarian cancer, uterine cancer, cervical cancer, testicular cancer, colon cancer, rectal cancer and skin cancer.
397. The method of any one of claims 376 and 377-396, wherein the EGFR- associated cancer is selected from the group consisting of: lung cancer, pancreatic cancer, head and neck cancer, melanoma, colon cancer, renal cancer, leukemia, glioblastoma, or breast cancer.
398. The method of claim 396 or 397, wherein the lung cancer is non-small cell lung cancer.
399. The method of any one of claims 377-398, wherein the cancer is a HER2- associated cancer.
400. The method of claim 399, wherein the HER2-associated cancer is associated with a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same.
401. The method of any one of claims 399 and 400, wherein determining that the cancer in the subject is a HER2-associated cancer includes performing an assay to detect dysregulation in a HER2 gene, a HER2 kinase protein, or expression or activity or level of any of the same in a sample from the subject.
402. The method of claim 401, further comprising obtaining a sample from the subject.
403. The method of claim 402, wherein the sample is a biopsy sample.
404. The method of any one of claims 401-403, wherein the assay is selected from the group consisting of sequencing, immunohistochemistry, enzyme-linked immunosorbent assay, and fluorescence in situ hybridization (FISH).
405. The method of claim 404, wherein the sequencing is pyrosequencing or next generation sequencing.
406. The method of any one of claims 400-405, wherein the dysregulation in a HER2 gene, a HER2 kinase protein, or expression or activity or level of any of the same is one or more point mutations in the HER2 gene.
407. The method of claim 406, wherein the one or more point mutations in a HER2 gene results in the translation of a HER2 protein having one or more amino acid substitutions at one or more of the following amino acid positions exemplified in Table 3.
408. The method of claim 407, wherein the one or more point mutations is selected from the mutations in Table 3 (e.g., S310F, S310Y, R678Q, R678W, R678P, I767M, V773M, V777L, and V842I).
409. The method of any one of claims 377-408, wherein the cancer is selected from the group consisting of: non-small cell lung cancer, pancreatic cancer, and colorectal cancer.
410. The method of any one of claims 377-409, further comprising administering an additional therapy or therapeutic agent to the subject.
411. The method of claim 410, wherein the additional therapy or therapeutic agent is selected from radiotherapy, cytotoxic chemotherapeutics, kinase targeted- therapeutics, apoptosis modulators, signal transduction inhibitors, immune-targeted therapies, and angiogenesis-targeted therapies.
412. The method of claim 411, wherein said additional therapeutic agent is selected from one or more kinase targeted therapeutics.
413. The method of claim 412, wherein said additional therapeutic agent is a tyrosine kinase inhibitor.
414. The method of claim 413, wherein said additional therapeutic agent is a second EGFR inhibitor.
415. The method of claim 410, wherein said additional therapeutic agent is selected from osimertinib, gefitinib, erlotinib, afatinib, lapatinib, neratinib, AZD-9291, CL-387785, CO-1686, WZ4002, and combinations thereof.
416. The method of claim 410, wherein said additional therapeutic agent is a second compound of any one of claims 1-375 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 376.
417. The method of claim 410, wherein said additional therapeutic agent is a HER2 inhibitor.
418. The method of claim 417, wherein the HER2 inhibitor is selected from trastuzumab, pertuzumab, trastuzumab emtansine, lapatinib, KU004, neratinib, dacomitinib, afatinib, tucatinib, erlotinib, pyrotinib, poziotinib, CP-724714, CUDC-101, sapitinib (AZD8931), tanespimycin (17-AAG), IPI-504, PF299, pelitinib, S- 22261 1, and AEE-788.
419. The method of any one of claims 417-418, wherein the compound of any one of claims 1-375 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 376, and the additional therapeutic agent are administered simultaneously as separate dosages.
420. The method of any one of claims 417-418, wherein the compound of any one of claims 1-375 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 376, and the additional therapeutic agent are administered as separate dosages sequentially in any order.
421. A method of treating a subject having a cancer, wherein the method comprises:
(a) administering one or more doses of a first EGFR inhibitor to the subject for a period of time;
(b) after (a), determining whether a cancer cell in a sample obtained from the subject has at least one EGFR inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor of step (a); and
(c) administering a compound of any one of claims 1 -375 or a pharmaceutically acceptable salt thereof as a monotherapy or in conjunction with another anti cancer agent to the subject if the subject has been determined to have a cancer cell that has at least one EGFR inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor of step (a); or
(d) administering additional doses of the first EGFR inhibitor of step (a) to the subject if the subject has not been determined to have a cancer cell that has at least one EGFR inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor of step (a).
422. The method of claim 421, wherein the anticancer agent in step (c) is a second EGFR inhibitor, an immunotherapy, a HER2 inhibitor, or a combination thereof.
423. The method of claim 421 , wherein the anti cancer agent in step (c) is the first EGFR inhibitor administered in step (a).
424. The method of claim 421, wherein the subject is administered additional doses of the first inhibitor of EGFR of step (a), and the method further comprises (e) administering another anti cancer agent to the subject.
425. The method of claim 424, wherein the anticancer agent of step (e) is a second EGFR inhibitor, an immunotherapy, or a combination thereof.
426. The method of claim 424, wherein the anticancer agent of step (e) is a compound of any one of claims 1-375 or a pharmaceutically acceptable salt thereof.
427. The method of any one of claims 421-426, wherein the EGFR inhibitor resistance mutation is a substitution at amino acid position 718, 747, 761, 790, 797, or 854 (e.g., L718Q, L747S, D761Y, T790M, C797S, T854A).
428. A method of treating an EGFR-associated cancer in a subject, the method comprising administering to a subject identified or diagnosed as having an EGFR- associated cancer that has one or more EGFR inhibitor resistance mutations a therapeutically effective amount of a compound of any one of claims 1-375 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 376.
429. A method of treating an EGFR-associated cancer in a subject, the method comprising:
(a) determining that the cancer in the subject has one or more EGFR inhibitor resistance mutations; and
(b) administering to the subject a therapeutically effective amount of a compound of any one of claims 1-375 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 376.
430. A method of treating a subject having a cancer, wherein the method comprises:
(a) determining whether a cancer cell in a sample obtained from a subject having a cancer and previously administered one or more doses of a first EGFR inhibitor has one or more EGFR inhibitor resistance mutations that confer increased resistance to a cancer cell or tumor to treatment with the first EGFR inhibitor that was previously administered to the subject; and
(b) administering a compound of any one of claims 1 -375 or a pharmaceutically acceptable salt thereof as a monotherapy or in conjunction with another anti cancer agent to the subject if the subject has been determined to have a cancer cell that has at least one EGFR inhibitor resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first modulator of EGFR that was previously administered to the subject; or
(c) administering additional doses of the first modulator of EGFR to the subj ect if the subject has not been determined to have a cancer cell that has at least one EGFR modulator resistance mutation that confers increased resistance to a cancer cell or tumor to treatment with the first modulator of EGFR previously administered to the subject.
431. The method of claim 430, wherein the anticancer agent of step (b) is a second EGFR innhibitor, an immunotherapy, a HER2 inhibitor, or a combination thereof.
432. The method of claim 430, wherein the anticancer agent of step (b) is the first EGFR inhibitor previously administered to the subject.
433. The method of claim 430, wherein the subject is administered additional doses of the first EGFR inhibitor previously administered to the subject, and the method further comprises (d) administering another anticancer agent to the subject.
434. The method of claim 433, wherein the anti cancer agent of step (d) is a second EGFR inhibitor, an immunotherapy, or a combination thereof.
435. The method of claim 433, wherein the anti cancer agent of step (d) is a compound of any one of claims 1-375 or a pharmaceutically acceptable salt thereof.
436. The method of claim 435, wherein the second EGFR inhibitor is selected from osimertinib, gefitinib, erlotinib, afatinib, lapatinib, neratinib, AZD-9291, CL-387785, CO- 1686, WZ4002, and combinations thereof.
437. The method of any one of claims 428-436, wherein the cancer is selected from the group consisting of: non-small cell lung cancer, pancreatic cancer, and colorectal cancer.
438. The method of any one of claims 428-437, wherein the cancer is associated with a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same.
439. The method of claim 438, wherein the dysregulation in a HER2 gene, a HER2 kinase protein, or expression or activity or level of any of the same is one or more point mutations in the HER2 gene.
440. The method of claim 439, wherein the one or more point mutations in a HER2 gene results in the translation of a HER2 protein having one or more amino acid substitutions at one or more of the following amino acid positions exemplified in Table 3.
441. The method of claim 440, wherein the one or more point mutations is selected from the mutations in Table 3 (e.g., S310F, S310Y, R678Q, R678W, R678P, I767M, V773M, V777L, and V842I).
442. A method for modulating EGFR in a mammalian cell, the method comprising contacting the mammalian cell with an effective amount of a compound of any one of claims 1-375, or a pharmaceutically acceptable salt thereof.
443. The method of claim 442, wherein the contacting occurs in vivo.
444. The method of claim 442, wherein the contacting occurs in vitro.
445. The method of any one of claims 442-444, wherein the mammalian cell is a mammalian cancer cell.
446. The method of claim 445, wherein the mammalian cancer cell is a mammalian EGFR-associated cancer cell.
447. The method of any one of claims 442-446, wherein the cell has a dysregulation of an EGFR gene, an EGFR kinase protein, or expression or activity or level of any of the same.
448. The method of claim 447, wherein the dysregulation in an EGFR gene, an EGFR kinase protein, or expression or activity or level of any of the same is one or more point mutations in the EGFR gene.
449. The method of claim 448, wherein the one or more point mutations in an EGFR gene results in the translation of an EGFR protein having one or more amino acid substitutions at one or more of the following amino acid positions exemplified in Table la and lb.
450. The method of claim 449, wherein the one or more point mutations is selected from the mutations in Table la and lb (e.g., L858R, G719S, G719C, G719A, L861Q, a deletion in exon 19 and/or an insertion in exon 20).
451. The method of claim 448, wherein the one or more point mutations is an EGFR inhibitor resistance mutation (e.g., L718Q, L747S, D761Y, T790M, C797S, T854A).
452. The method of claim 448, wherein the one or more point mutations in an EGFR gene include a deletion in exon 19 of a human EGFR gene.
453. The method of claim 448, wherein the one or more point mutations is an EGFR insertion in exon 20 of a human EGFR gene.
454. The method of claim 453, wherein the insertion in exon 20 of a human EGFR gene is selected from: A767_V769insX, V769_D770insX, D770_N771insX, N771_P772insX, P772_H773insX, and H773_V774insX.
455. The method of claim 454, wherein the insertion in exon 20 of a human
EGFR gene is selected from: A767_V769dupASV, V769_D770insASV, D770_N771insNPG, D770_N771insNPY, D770_N771insSVD, D770_N771insGL, N771_H773dupNPH, N771_P772insN, N771_P772insH, N771_P772insV,
P772_H773insDNP, P772_H773insPNP, H773_V774insNPH, H773_V774insH, H773_V774insPH, H773_V774insAH, and P772_H773insPNP.
456. A method for treating cancer in a subject in need thereof, the method comprising (a) determining that the cancer is associated with a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same; and (b) administering to the subject a therapeutically effective amount of a compound of any one of claims 1-375, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 376.
457. A method of treating a HER2-associated cancer in a subject, the method comprising administering to a subject identified or diagnosed as having a HER2-associated cancer a therapeutically effective amount of a compound of any one of claims 1-375 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 376.
458. A method of treating a HER2-associated cancer in a subject, the method comprising:
(a) determining that the cancer in the subject is a HER2-associated cancer; and
(b) administering to the subject a therapeutically effective amount of a compound of any one of claims 1-375 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 376.
459. A method of treating a subject, the method comprising administering a therapeutically effective amount of a compound of any one of claims 1-375 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 376, to a subject having a clinical record that indicates that the subject has a dysregulation of a HER2 gene, a HER2 kinase, or expression or activity or level of any of the same.
460. The method of any one of claims 456 and 458, wherein the step of determining that the cancer in the subject is a HER2-associated cancer includes performing an assay to detect dysregulation in a HER2 gene, a HER2 kinase protein, or expression or activity or level of any of the same in a sample from the subject.
461. The method of claim 460, further comprising obtaining a sample from the subject.
462. The method of claim 461, wherein the sample is a biopsy sample.
463. The method of any one of claims 456-462, wherein the assay is selected from the group consisting of sequencing, immunohistochemistry, enzyme-linked immunosorbent assay, and fluorescence in situ hybridization (FISH).
464. The method of claim 463, wherein the FISH is break apart FISH analysis.
465. The method of claim 463, wherein the sequencing is pyrosequencing or next generation sequencing.
466. The method of any one of claims 456, 459 or 460, wherein the dysregulation in a HER2 gene, a HER2 kinase protein, or expression or activity or level of any of the same is one or more point mutations in the HER2 gene.
467. The method of claim 446, wherein the one or more point mutations in a HER2 gene results in the translation of a HER2 protein having one or more amino acid substitutions at one or more of the following amino acid positions exemplified in Table 3.
468. The method of claim 446, wherein the one or more point mutations is selected from the mutations in Table 3 (e.g., S310F, S310Y, R678Q, R678W, R678P, I767M, V773M, V777L, and V842I).
469. The method of any one of claims 455, 458 or 459, wherein the dysregulation in a HER2 gene, a HER2 kinase protein, or expression or activity or level of any of the same is an insertion in exon 20 of the human HER2 gene.
470. The method of claim 469, wherein the insertion in exon 20 of the human HER2 gene is deletions at an amino acid position selected from: 774, 775, 776, 777, 778, and 780.
471. The method of claim 470, wherein the insertion in exon 20 of a human HER2 gene is selected from: M774AYVM, M774del insWLV, A775_G776insYVMA, A775_G776insAVMA, A775_G776insSVMA, A775_G776insVAG, A775insV G776C, A775_G776insI, G776del insVC2, G776del insW, G776del insLC, G776C V777insC, G776C V777insV, V777_G778insCG, G778_S779insCPG, and P780_Y781insGSP.
472. The method of any one of claims 457, 458 or 460, wherein the HER2- associated cancer is selected from the group consisting of: colon cancer, lung cancer, or breast cancer.
473. The method of claim 472, wherein the lung cancer is non-small cell lung cancer.
474. The method of any one of claims 457-473, further comprising administering an additional therapy or therapeutic agent to the subject.
475. The method of claim 474, wherein the additional therapy or therapeutic agent is selected from radiotherapy, cytotoxic chemotherapeutics, kinase targeted- therapeutics, apoptosis modulators, signal transduction inhibitors, immune-targeted therapies and angiogenesis-targeted therapies.
476. The method of claim 474, wherein said additional therapeutic agent is a second compound of any one of claims 1-375 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 376.
477. The method of claim 474, wherein said additional therapeutic agent is selected from one or more kinase targeted therapeutics.
478. The method of claim 474, wherein said additional therapeutic agent is a tyrosine kinase inhibitor.
479. The method of claim 474, wherein said additional therapeutic agent is an EGFR inhibitor.
480. The method of claim 474, wherein said additional therapeutic agent is selected from osimertinib, gefitinib, erlotinib, afatinib, lapatinib, neratinib, AZD-9291, CL-387785, CO-1686, WZ4002, and combinations thereof.
481. The method of claim 474, wherein said additional therapeutic agent is a HER2 inhibitor.
482. The method of claim 481, wherein the HER2 inhibitor is selected from trastuzumab, pertuzumab, trastuzumab emtansine, lapatinib, KU004, neratinib, dacomitinib, afatinib, tucatinib, erlotinib, pyrotinib, poziotinib, CP-724714, CUDC-101, sapitinib (AZD8931), tanespimycin (17-AAG), IPI-504, PF299, pelitinib, S- 22261 1, and AEE-788.
483. The method of any one of claims 477-482, wherein the compound of any one of claims 1-375 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 376, and the additional therapeutic agent are administered simultaneously as separate dosages.
484. The method of any one of claims 477-482, wherein the compound of any one of claims 1-375 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 376, and the additional therapeutic agent are administered as separate dosages sequentially in any order.
PCT/US2023/064133 2022-03-11 2023-03-10 Tetrahydroindole derivatives as egfr and/or her2 inhibtors useful for the treatment of cancer WO2023173083A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263319183P 2022-03-11 2022-03-11
US63/319,183 2022-03-11

Publications (1)

Publication Number Publication Date
WO2023173083A1 true WO2023173083A1 (en) 2023-09-14

Family

ID=85873762

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/064133 WO2023173083A1 (en) 2022-03-11 2023-03-10 Tetrahydroindole derivatives as egfr and/or her2 inhibtors useful for the treatment of cancer

Country Status (1)

Country Link
WO (1) WO2023173083A1 (en)

Citations (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6906194B2 (en) 2003-10-08 2005-06-14 Massachusetts Instititue Of Technology Fluorescence assay for kinase activity
WO2013050438A1 (en) 2011-10-06 2013-04-11 Bayer Pharma Aktiengesellschaft Substituted benzylindazoles for use as bub1 kinase inhibitors in the treatment of hyperproliferative diseases.
WO2013092512A1 (en) 2011-12-21 2013-06-27 Bayer Intellectual Property Gmbh Substituted benzylpyrazoles
WO2013167698A1 (en) 2012-05-11 2013-11-14 Bayer Pharma Aktiengesellschaft Substituted cycloalkenopyrazoles as bub1 inhibitors for the treatment of cancer
US8586570B2 (en) 2006-08-28 2013-11-19 Massachusetts Institute Of Technology Sox-based kinase sensor
WO2014147203A1 (en) 2013-03-21 2014-09-25 Bayer Pharma Aktiengesellschaft 3-heteroaryl substituted indazoles
WO2014147204A1 (en) 2013-03-21 2014-09-25 Bayer Pharma Aktiengesellschaft Heteroaryl substituted indazoles
WO2014176475A2 (en) 2013-04-26 2014-10-30 The Regents Of The University Of Michigan Egfr inhibitors and uses thereof
WO2014202584A1 (en) 2013-06-21 2014-12-24 Bayer Pharma Aktiengesellschaft Heteroaryl substituted pyrazoles
WO2014202583A1 (en) 2013-06-21 2014-12-24 Bayer Pharma Aktiengesellschaft Substituted benzylpyrazoles
WO2014202590A1 (en) 2013-06-21 2014-12-24 Bayer Pharma Aktiengesellschaft Substituted benzylpyrazoles
WO2014202588A1 (en) 2013-06-21 2014-12-24 Bayer Pharma Aktiengesellschaft Heteroaryl substituted pyrazoles
WO2015063003A1 (en) 2013-10-30 2015-05-07 Bayer Pharma Aktiengesellschaft Heteroaryl substituted pyrazoles
US9029502B2 (en) 2010-12-20 2015-05-12 The Regents Of The University Of Michigan Inhibitors of the epidermal growth factor receptor-heat shock protein 90 binding interaction
WO2015193339A1 (en) 2014-06-17 2015-12-23 Bayer Pharma Aktiengesellschaft 3-amino-1,5,6,7-tetrahydro-4h-indol-4-ones
WO2016202755A1 (en) 2015-06-17 2016-12-22 Bayer Pharma Aktiengesellschaft 3-amino-1,5,6,7-tetrahydro-4h-indol-4-ones
WO2017021348A1 (en) 2015-08-05 2017-02-09 Bayer Pharma Aktiengesellschaft 1h-pyrrol-3-amines
US20170166598A1 (en) 2014-05-13 2017-06-15 Ariad Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition
US9920060B2 (en) 2015-09-01 2018-03-20 Taiho Pharmaceutical Co., Ltd. Pyrazolo[3,4-d]pyrimidine compound or salt thereof
WO2018094225A1 (en) 2016-11-17 2018-05-24 Board Of Regents, The University Of Texas System Compounds with anti-tumor activity against cancer cells bearing egfr or her2 exon 20 mutations
WO2019046775A1 (en) 2017-08-31 2019-03-07 Dana-Farber Cancer Institute, Inc. Inhibitors of egfr and/or her2 and methods of use
WO2019081486A1 (en) 2017-10-24 2019-05-02 Bayer Aktiengesellschaft 4h-pyrrolo[3,2-c]pyridin-4-one derivatives
WO2019165385A1 (en) 2018-02-23 2019-08-29 Fulton Group N.A., Inc. Inward-firing premix fuel combustion burner
WO2019165358A1 (en) 2018-02-23 2019-08-29 The Regents Of The University Of Michigan Egfr dimer disruptors and use of the same
WO2019241715A1 (en) 2018-06-14 2019-12-19 Dana-Farber Cancer Institute, Inc. Cyano quinoline amide compounds as her2 inhibitors and methods of use
WO2019246541A1 (en) 2018-06-21 2019-12-26 Dana-Farber Cancer Institute, Inc. Inhibitors of egfr and methods of use thereof
WO2022066734A1 (en) * 2020-09-23 2022-03-31 Scorpion Therapeutics, Inc. Pyrrolo[3,2-c]pyridin-4-one derivatives useful in the treatment of cancer
WO2022072634A1 (en) * 2020-09-30 2022-04-07 Scorpion Therapeutics, Inc. Bicyclic compounds for use in the treatment cancer
WO2022094271A1 (en) * 2020-10-30 2022-05-05 Scorpion Therapeutics, Inc. Methods for treating cancer

Patent Citations (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6906194B2 (en) 2003-10-08 2005-06-14 Massachusetts Instititue Of Technology Fluorescence assay for kinase activity
US8586570B2 (en) 2006-08-28 2013-11-19 Massachusetts Institute Of Technology Sox-based kinase sensor
US9029502B2 (en) 2010-12-20 2015-05-12 The Regents Of The University Of Michigan Inhibitors of the epidermal growth factor receptor-heat shock protein 90 binding interaction
WO2013050438A1 (en) 2011-10-06 2013-04-11 Bayer Pharma Aktiengesellschaft Substituted benzylindazoles for use as bub1 kinase inhibitors in the treatment of hyperproliferative diseases.
WO2013092512A1 (en) 2011-12-21 2013-06-27 Bayer Intellectual Property Gmbh Substituted benzylpyrazoles
WO2013167698A1 (en) 2012-05-11 2013-11-14 Bayer Pharma Aktiengesellschaft Substituted cycloalkenopyrazoles as bub1 inhibitors for the treatment of cancer
WO2014147204A1 (en) 2013-03-21 2014-09-25 Bayer Pharma Aktiengesellschaft Heteroaryl substituted indazoles
WO2014147203A1 (en) 2013-03-21 2014-09-25 Bayer Pharma Aktiengesellschaft 3-heteroaryl substituted indazoles
WO2014176475A2 (en) 2013-04-26 2014-10-30 The Regents Of The University Of Michigan Egfr inhibitors and uses thereof
WO2014202584A1 (en) 2013-06-21 2014-12-24 Bayer Pharma Aktiengesellschaft Heteroaryl substituted pyrazoles
WO2014202583A1 (en) 2013-06-21 2014-12-24 Bayer Pharma Aktiengesellschaft Substituted benzylpyrazoles
WO2014202590A1 (en) 2013-06-21 2014-12-24 Bayer Pharma Aktiengesellschaft Substituted benzylpyrazoles
WO2014202588A1 (en) 2013-06-21 2014-12-24 Bayer Pharma Aktiengesellschaft Heteroaryl substituted pyrazoles
WO2015063003A1 (en) 2013-10-30 2015-05-07 Bayer Pharma Aktiengesellschaft Heteroaryl substituted pyrazoles
US20170166598A1 (en) 2014-05-13 2017-06-15 Ariad Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition
WO2015193339A1 (en) 2014-06-17 2015-12-23 Bayer Pharma Aktiengesellschaft 3-amino-1,5,6,7-tetrahydro-4h-indol-4-ones
WO2016202755A1 (en) 2015-06-17 2016-12-22 Bayer Pharma Aktiengesellschaft 3-amino-1,5,6,7-tetrahydro-4h-indol-4-ones
WO2017021348A1 (en) 2015-08-05 2017-02-09 Bayer Pharma Aktiengesellschaft 1h-pyrrol-3-amines
US9920060B2 (en) 2015-09-01 2018-03-20 Taiho Pharmaceutical Co., Ltd. Pyrazolo[3,4-d]pyrimidine compound or salt thereof
WO2018094225A1 (en) 2016-11-17 2018-05-24 Board Of Regents, The University Of Texas System Compounds with anti-tumor activity against cancer cells bearing egfr or her2 exon 20 mutations
WO2019046775A1 (en) 2017-08-31 2019-03-07 Dana-Farber Cancer Institute, Inc. Inhibitors of egfr and/or her2 and methods of use
WO2019081486A1 (en) 2017-10-24 2019-05-02 Bayer Aktiengesellschaft 4h-pyrrolo[3,2-c]pyridin-4-one derivatives
WO2019165385A1 (en) 2018-02-23 2019-08-29 Fulton Group N.A., Inc. Inward-firing premix fuel combustion burner
WO2019165358A1 (en) 2018-02-23 2019-08-29 The Regents Of The University Of Michigan Egfr dimer disruptors and use of the same
WO2019241715A1 (en) 2018-06-14 2019-12-19 Dana-Farber Cancer Institute, Inc. Cyano quinoline amide compounds as her2 inhibitors and methods of use
WO2019246541A1 (en) 2018-06-21 2019-12-26 Dana-Farber Cancer Institute, Inc. Inhibitors of egfr and methods of use thereof
WO2022066734A1 (en) * 2020-09-23 2022-03-31 Scorpion Therapeutics, Inc. Pyrrolo[3,2-c]pyridin-4-one derivatives useful in the treatment of cancer
WO2022072634A1 (en) * 2020-09-30 2022-04-07 Scorpion Therapeutics, Inc. Bicyclic compounds for use in the treatment cancer
WO2022094271A1 (en) * 2020-10-30 2022-05-05 Scorpion Therapeutics, Inc. Methods for treating cancer

Non-Patent Citations (81)

* Cited by examiner, † Cited by third party
Title
"Pharmaceutical Preformulation and Formulation", 2009, THE PHARMACEUTICAL PRESS AND THE AMERICAN PHARMACEUTICAL ASSOCIATION
AFRATIS KONSTANTINOS ET AL: "Regioselective Synthesis of Fully Substituted Fused Pyrroles through an Oxidant-Free Multicomponent Reaction", ORGANIC LETTERS, vol. 25, no. 3, 27 January 2023 (2023-01-27), US, pages 461 - 465, XP093047714, ISSN: 1523-7060, DOI: 10.1021/acs.orglett.2c03889 *
ARANVERONICAJASMINKA OMEROVIC, INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, vol. 20, no. 22, 2019, pages 5701
ARCILA ET AL., CLIN CANCER RES., vol. 18, no. 18, 15 September 2012 (2012-09-15)
BAGCHI ET AL., DRUG DES DEVEL THER., vol. 13, 2019, pages 3591 - 3605
BERHMAN REKLIEGMAN RARVIN AMNELSON WE.: "Nelson Textbook of Pediatrics", 1996, W.B. SAUNDERS COMPANY
BOLANOS-GARCIA VMBLUNDELL TL, TRENDS BIOCHEM. SCI., vol. 36, 2010, pages 141
BOSE ET AL., CANCER DISCOV., vol. 3, no. 2, February 2013 (2013-02-01), pages 224 - 37
CADENA GARCÍA JUAN MARTÍN ET AL: "EGFR and HER2 small molecules inhibitors as potential therapeutics in veterinary oncology", REVISTA COLOMBIANA DE CIENCIAS QUÍMICO-FARMACÉUTICAS, vol. 49, no. 2, 1 May 2020 (2020-05-01), XP093047820, ISSN: 0034-7418, Retrieved from the Internet <URL:https://revistas.unal.edu.co/index.php/rccquifa/article/viewFile/89898/75931> DOI: 10.15446/rcciquifa.v49n2.89898 *
CHEN ET AL., SCI REP., vol. 9, no. 1, 21 February 2019 (2019-02-21), pages 2516
CHO ET AL., NAT COMMUN., vol. 8, 2017, pages 15623
CHOI ET AL., BIOMED RES INT., 15 May 2018 (2018-05-15)
CHRISTGEN ET AL., VIRCHOWS ARCH., vol. 473, no. 5, November 2018 (2018-11-01), pages 577 - 582
DING ET AL., CANCER RES., vol. 63, no. 5, 1 March 2003 (2003-03-01), pages 1106 - 13
DOAN ET AL., J PHARMACOL EXP THER., vol. 303, no. 3, 2002, pages 1029 - 1037
FILIPSKI, K.J. ET AL., CURRENT TOPICS IN MEDICINAL CHEMISTRY, vol. 13, 2013, pages 776 - 802
GASTFRIEND ET AL., CURR OPIN BIOMED ENG., vol. 5, March 2018 (2018-03-01), pages 6 - 12
GHARIB ET AL., J CELL PHYSIOL., vol. 234, no. 8, August 2019 (2019-08-01), pages 13137 - 13144
GREENE, T.W.WUTS, P.G. M.: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
GROSSE AGROSSE CRECHSTEINER MSOLTERMANN A., DIAGN PATHOL., vol. 14, no. 1, 11 February 2019 (2019-02-11), pages 18
HANKER ET AL., CANCER DISCOV., vol. 7, no. 6, June 2017 (2017-06-01), pages 575 - 585
HYNESSTERN, BIOCHIM. BIOPHYS. ACTA, vol. 1198, 1994, pages 165 - 184
IRIE ET AL., MOL CANCER THER., vol. 18, no. 4, April 2019 (2019-04-01), pages 733 - 742
IWAKURANAWA., FRONT CELL NEUROSCI., vol. 13, no. 7, February 2013 (2013-02-01), pages 4
KARACHIALIOU ET AL.: "Real-time liquid biopsies become a reality in cancer treatment", ANN. TRANSL. MED., vol. 3, no. 3, 2016, pages 36, XP055630293, DOI: 10.3978/j.issn.2305-5839.2015.01.16
KAVURI ET AL., CANCER DISCOV., vol. 5, no. 8, August 2015 (2015-08-01), pages 832 - 841
KIM EYCHO ENPARK HS ET AL., CANCER BIOL THER., vol. 17, no. 3, 2016, pages 237 - 245
KING RW, BIOCHIM BIOPHYS ACTA, vol. 1786, 2008, pages 4
KLAPPER ET AL., ADV. CANCER RES., vol. 77, 2000, pages 25 - 79
KOHSAKASHINJI ET AL., SCIENCE TRANSLATIONAL MEDICINE, vol. 9, no. 416, 2017, pages eaan6566
KOPS GJ ET AL., NATURE REV. CANCER, vol. 5, 2005, pages 773
KOSAKA ET AL., CANCER RES., vol. 77, no. 10, 15 May 2017 (2017-05-15), pages 2712 - 2721
KRAWCZYK ET AL., ONCOL LETT., vol. 6, no. 4, October 2013 (2013-10-01), pages 1063 - 1067
L. FIESERM. FIESER: "Fieser and Fieser's Reagents for Organic Synthesis", 1994, JOHN WILEY AND SONS
LAI ET AL., EUR J CANCER., vol. 109, March 2019 (2019-03-01), pages 28 - 35
LAMMERS ET AL.: "Effect of Intratumoral Injection on the Biodistribution and the Therapeutic Potential of HPMA Copolymer-Based Drug Delivery Systems", NEOPLASIA, vol. 10, 2006, pages 788 - 795
LI ET AL., J THORAC ONCOL., vol. 1 1, no. 3, March 2016 (2016-03-01), pages 414 - 9
LI, SHIQING ET AL., CANCER CELL, vol. 7, no. 4, 2005, pages 301 - 311
LIU ET AL., J EXP CLIN CANCER RES., vol. 38, no. 1, 23 May 2019 (2019-05-23), pages 219
MARIN-ACEVEDO ET AL., JHEMATOL ONCOL., vol. 11, no. 39, 2018
MASOODASHIQRAMA KRISHNA KANCHAJANAKIRAMAN SUBRAMANIAN: "Seminars in oncology", 2019, WB SAUNDERS
MICHELE ET AL., BEAU-FALLER, 2012, pages 10507 - 10507
MOASSER, ONCOGENE, vol. 26, no. 45, 4 October 2007 (2007-10-04), pages 6469 - 6487
MONDALGOURISH ET AL., ACTA NEUROPATHOL., vol. 139, no. 6, 2020, pages 1071 - 1088
MORPHY., J. MED. CHEM., vol. 53, no. 4, 2010, pages 1413 - 1437
PAHUJA ET AL., CANCER CELL., vol. 34, no. 5, 12 November 2018 (2018-11-12), pages 792 - 806
PAPADIMITRAKOPOULOU, V.A. ET AL., ANNALS OF ONCOLOGY, vol. 29, 2018
PENG ET AL., CHEMRXIV., 2019
PETERS., J. MED. CHEM., vol. 56, no. 22, 2013, pages 8955 - 8971
PINES, GURWOLFGANG J. KOSTLERYOSEF YARDEN, FEBS LETTERS, vol. 584, no. 12, 2010, pages 2699 - 2706
R. LAROCK: "Comprehensive Organic Transformations", 1989, VCH PUBLISHERS
RABINDRAN ET AL., CANCER RES., vol. 64, no. 11, 1 June 2004 (2004-06-01), pages 3958 - 65
REXERARTEAGA, CRIT REV ONCOG., vol. 17, no. 1, 2012, pages 1
ROBICHAUX ET AL., NAT MED., vol. 24, no. 5, May 2018 (2018-05-01), pages 638 - 646
ROSS ET AL., CANCER., vol. 124, no. 7, 1 April 2018 (2018-04-01), pages 1358 - 1373
RUDOLPH AM ET AL.: "Rudolph's Pediatrics", 2002, MCGRAW-HILL
SALOMON ET AL., CRIT. REV. ONCOL. HEMATOL., vol. 19, 1995, pages 183 - 232
SCHLESSINGER J, COLD SPRING HARB PERSPECT BIOL, vol. 6, 2014, pages a008912
SCHMIDT MBASTIANS H, DRUG RES. UPDATES, vol. 10, 2007, pages 162
SCHMIDT MMEDEMA RH, CELL CYCLE, vol. 5, 2006, pages 159
SHAHRIYAZJASON F. LESTER., CLINICAL LUNG CANCER, 2019
SHAN ET AL., CELL, vol. 149, no. 4, 2012, pages 860 - 870
SI ET AL., CANCER BIOMARK., vol. 23, no. 2, 2018, pages 165 - 171
SMITH, M. B.MARCH, J.: "March' s Advanced Organic Chemistry: Reactions, Mechanisms, and Structure", 2001, JOHN WILEY & SONS
STAROSYLA ET AL., WORLD J PHARMACOL, vol. 3, no. 4, 9 December 2014 (2014-12-09), pages 162 - 173
STEWART ELTAN SZLIU GTSAO MS., TRANSL LUNG CANCER RES., vol. 4, no. 1, 2015, pages 67 - 81
STEWART ELTAN SZLIU GTSAO MS.TRANSL LUNG, CANCER RES., vol. 4, no. 1, 2015, pages 67 - 81
SUN ET AL., J CELL MOL MED., vol. 19, no. 12, December 2015 (2015-12-01), pages 2691 - 2701
UDAGERAARON M. ET AL., CANCER RES, vol. 75, no. 13, 2015, pages 2600 - 2606
VYSEHUANG ET AL., SIGNAL TRANSDUCT TARGET THER., vol. 4, no. 5, 8 March 2019 (2019-03-08), pages 5
WANG ET AL., AM J TRANSL RES., vol. 11, no. 2, 2019, pages 520 - 528
WANG ET AL., BIOTECHNOL BIOENG., vol. 114, no. 1, January 2017 (2017-01-01), pages 184 - 194
WANG ET AL., DRUG DELIV., vol. 26, no. 1, 2019, pages 551 - 565
WANG ET AL., SCI REP., vol. 7, 2017, pages 45917
WEAVER BACLEVELAND DW, CANCER RES., vol. 67, 2007, pages 10103
XU ET AL., THORAC CANCER., vol. 11, no. 3, March 2020 (2020-03-01), pages 679 - 685
YARDEN YPINES G, NAT REV CANCER, vol. 12, 2012, pages 553 - 563
YASUDAHIROYUKISUSUMU KOBAYASHIDANIEL B. COSTA., THE LANCET ONCOLOGY, vol. 13, no. 1, 2012, pages e23 - e31
YUN ET AL., CANCER CELL, vol. 11, no. 3, 2007, pages 217 - 227
YUN ET AL., CANCER CELL., vol. 11, no. 3, 2007, pages 217 - 227
YUN ET AL., PROC NATL ACAD SCI U S A., vol. 105, no. 6, 12 February 2008 (2008-02-12), pages 2070 - 5

Similar Documents

Publication Publication Date Title
KR102099159B1 (en) Acrylic acid derivatives, methods for their preparation and methods of use in medicine
JP2022507527A (en) KRAS G12C inhibitor and its usage
ES2753386T3 (en) 2-Hydroxy-1- {4 - [(4-phenyl) phenyl] carbonyl} piperazin-1-yl} ethane-1-one derivatives and related compounds as fatty acid synthase inhibitors (FASN) for the treatment of cancer
KR20130025860A (en) Heteroarylaminoquinolines as tgf-beta receptor kinase inhibitors
TW201319055A (en) Aminoquinazoline derivatives and their salts and methods of use
JP2023543528A (en) Pyrrolo[3,2-c]pyridin-4-one derivatives useful in the treatment of cancer
WO2022094271A1 (en) Methods for treating cancer
WO2022098992A1 (en) Use of macrocyclic compounds in methods of treating cancer
WO2022251497A1 (en) Heterocyclic compounds and methods of use
WO2022072634A1 (en) Bicyclic compounds for use in the treatment cancer
US11897871B1 (en) Methods for treating cancer
WO2022076831A2 (en) Methods for treating cancer
TW202320792A (en) Combination therapy comprising an fgfr inhibitor and a kras inhibitor
WO2023173083A1 (en) Tetrahydroindole derivatives as egfr and/or her2 inhibtors useful for the treatment of cancer
TWI565698B (en) Quinoxaline compounds, method for preparing the same and use thereof
WO2022197913A1 (en) Bicyclic derivatives which can be used to treat cancer
WO2022072645A2 (en) Methods for treating cancer
WO2022072632A1 (en) Bicyclic compounds for use in the treatment cancer
WO2023018636A1 (en) Compounds that inhibit pi3k isoform alpha and methods for treating cancer
KR20240051953A (en) Compounds that inhibit PI3K isoform alpha and methods for treating cancer
WO2024077036A1 (en) Methods for treating cancer
WO2023245015A2 (en) Phenyl amide compounds and methods of use
WO2023150291A2 (en) Heterocyclic compounds and methods of use

Legal Events

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

Ref document number: 23715379

Country of ref document: EP

Kind code of ref document: A1