COVALENT EGFR INHIBITORS AND METHODS OF USE THEREOF

Information

  • Patent Application
  • 20230295137
  • Publication Number
    20230295137
  • Date Filed
    March 28, 2023
    a year ago
  • Date Published
    September 21, 2023
    8 months ago
Abstract
The disclosure relates to compounds that act as covalent inhibitors of epidermal growth factor receptor (EGFR); pharmaceutical compositions comprising the compounds; and methods of treating or preventing kinase-mediated disorders, including cancer and other proliferation diseases.
Description
BACKGROUND

The epidermal growth factor receptor (EGFR, Erb-B1) belongs to a family of receptor tyrosine kinases that mediate the proliferation, differentiation, and survival of normal and malignant cells (Arteaga, C. L., J. Clin. Oncol. 19, 2001, 32-40). Deregulation of EGFR has been implicated in many types of human cancer, with overexpression of the receptor present in at least 70% of human cancers (Seymour, L. K., Curr. Drug Targets 2, 2001, 117-133), including non-small lung cell carcinomas, breast cancers, gliomas, squamous cell carcinomas of the head and neck, and prostate cancer (Raymond, E., et al., Drugs 60 (Suppl. 1), 2000, 15-23, discussion 41-2; Salomon, D. S., et al., Crit. Rev. Oncol. Hematol. 19, 1995, 183-232; Voldborg B. R., et al., Ann. Oncol. 8, 1997, 1197-1206). EGFR has, therefore, emerged as an attractive target for the design and development of diagnostic and therapeutic agents that can specifically bind and inhibit the receptor's tyrosine kinase activity and signal transduction pathway in cancer cells. For example, the EGFR tyrosine kinase (EGFR-TK) reversible inhibitor TARCEVA® is approved by the FDA for treatment of NSCLC and advanced pancreatic cancer. Other anti-EGFR targeted molecules have also been approved, including Lapatinib and IRESSA®.


Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) are effective clinical therapies for EGFR mutant advanced non-small cell lung cancer (NSCLC) patients (Mok, T. S., et al, N. Engl. J. Med. 361, 2009, 947-57; Paez, J. G., et al., Science 304, 2004, 1497-500; Lynch, T. J., et al., N. Engl. J. Med. 350, 2004, 2129.39; Rosell, R., et al., Lancet Oncol. 13, 2012, 239-46). Several randomized clinical trials have demonstrated that EGFR TKIs are more effective, as measured by response rate (RR) and progression free survival (PFS), than chemotherapy when used as initial systemic treatment for advanced EGFR mutant NSCLC (Mok, T. S., et al., N. Engl. J. Med. 361, 2009, 947-57; Rosell, R., et al., Lancet Oncol. 13, 2012, 239-46; Sequest, L. V. et al., J. Clin. Oncol. 31, 2013, 3327-34; Wu, Y. L., et al., Lancet Oncol. 15, 2014, 213-22; Maemondo, M., et al, N. Engl. J. Med. 362, 2010, 2380-8; Zhou, C., et al., Lancet Oncol. 12, 2011.735-42; Mitsudomi, T., et al., Lancet Oncol. 11, 2010, 121-8). However, the vast majority of patients will develop disease progression following successful treatment with an EGFR TKI. The most common mechanism of acquired resistance, detected in 60% of patients, is a secondary mutation in EGFR at position T790 (T790M) (Yu, H. A., et al., Clin. Cancer Res. 19, 2013, 2240-7). This mutation leads to an increase in ATP affinity, thus making it more difficult for reversible EGFR TKIs gefitinib and erlotinib to bind the EGFR TKI domain (Yun C. H., et al., Proc. Natl. Acad. Sci. USA 105, 2008, 2070-5).


Covalent EGFR inhibitors have emerged for inhibiting EGFR T790M-containing cancers. However, in lung cancer patients, afatinib is only effective in EGFR TKI naïve EGFR mutant cancers and has a RR of less than 10% in patients with NSCLC that have developed resistance to gefitinib or erlotinib (Miller, V. A., et al., Lancet Oncol. 13, 2012, 528-38). Afatinib is a potent inhibitor of both mutant and wild type (WT) EGFR. Inhibition of WT EGFR leads to toxicities, including skin rash and diarrhea, which limits the ability to escalate afatinib doses in patients to those necessary to inhibit EGFR T790M. Irreversible pyrimidine EGFR inhibitors including the tool compound WZ4002 and clinical compounds CO-1686 and AZD9291, overcome many of the limitations of afatinki (Zhou, W., et al., Nature 462, 2009, 1070-4; Walter, A. O., et al., Cancer Discov. 3, 2013, 1404-15; Cross, D. A. E., et al., Cancer Decov. 4, 2014, 1046-61). They are not only more potent on EGFR T790M, but also selectively inhibit mutant over WT EGFR and hence should lead to increased clinical efficacy and less toxicity compared with afatinib (Zhou, W., et al; Walter A. O., et al, Cross, D. A. E., et al.).


However, all current EGFR TKIs target the ATP site, and while third generation irreversible inhibitors can overcome T790M, they are all rendered impotent by the C797S mutation, which is already arising in treated patients. Cetuximab, an anti-EGFR antibody that blocks receptor dimerization, is not effective in EGFR-mutant NSCLC because mutational activation of the kinase is effectively “downstream” of receptor dimerization. Hence, alternative strategies to inhibit EGFR are needed. At present, suitable compounds with alternative mechanisms of action targeting mutant EGFR are not available. Thus, there is a need for potent small molecule EGFR inhibitors with alternative mechanisms of action targeting mutant EGFR.


SUMMARY

In an aspect, provided herein is a compound of Formula I:




embedded image


or a pharmaceutically acceptable salt thereof;


wherein the variables are defined herein.


In an embodiment, the compound of Formula I is a compound of Formula II:




embedded image


or a pharmaceutically acceptable salt thereof.


In another embodiment, the compound of Formula I is a compound of Formula III:




embedded image


or a pharmaceutically acceptable salt thereof.


In an aspect, provided herein is a method of treating cancer or a proliferation disease, comprising administering to a subject in need thereof an effective amount of a compound of disclosed herein or a pharmaceutical composition comprising a compound disclosed herein and a pharmaceutically acceptable carrier. In one embodiment, the cancer is lung cancer, breast cancer, glioma, squamous cell carcinoma, or prostate cancer. In another embodiment, the cancer is non-small cell lung cancer (NSCLC).


In another aspect, provided herein is a method of inhibiting the activity of EGFR, comprising administering to a subject in need thereof an effective amount of a compound of disclosed herein or a pharmaceutical composition comprising a compound disclosed herein and a pharmaceutically acceptable carrier. In an embodiment, the compound targets Cys775 on EGFR.


The disclosure also provides a kit comprising a compound capable of inhibiting EGFR activity selected from a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, and instructions for use in treating cancer. In one embodiment, the kit further comprises components for performing a test to determine whether a subject has an activating mutation in EGFR or a resistance mutation in EGFR







DETAILED DESCRIPTION
Definitions

Listed below are definitions of various terms used to describe the compounds and compositions disclosed herein. These definitions apply to the terms as they are used throughout this specification and claims, unless otherwise limited in specific instances, either individually or as part of a larger group.


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. Generally, the nomenclature used herein and the laboratory procedures in cell culture, molecular genetics, organic chemistry, and peptide chemistry are those well-known and commonly employed in the art.


As used herein, the articles “a” and “an” refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element. Furthermore, use of the term “including” as well as other forms, such as “include”, “includes,” and “included,” is not limiting.


As used herein, the term “about” will be understood by persons of ordinary skill in the art and will vary to some extent on the context in which it is used. As used herein when referring to a measurable value such as an amount, a temporal duration, and the like, the term “about” is meant to encompass variations of ±20% or ±10%, including ±5%, ±1%, and ±0.1% from the specified value, as such variations are appropriate to perform the disclosed methods.


The term “administration” or the like as used herein refers to the providing a therapeutic agent to a subject. Multiple techniques of administering a therapeutic agent exist in the art including, but not limited to, intravenous, oral, aerosol, parenteral, ophthalmic, pulmonary, and topical administration.


The term “treat,” “treated,” “treating,” or “treatment” includes the diminishment or alleviation of at least one symptom associated or caused by the state, disorder or disease being treated. In certain embodiments, the treatment comprises bringing into contact with wild-type or mutant EGFR an effective amount of a compound disclosed herein for conditions related to cancer.


As used herein, the term “prevent” or “prevention” means no disorder or disease development if none had occurred, or no further disorder or disease development if there had already been development of the disorder or disease. Also considered is the ability of one to prevent some or all of the symptoms associated with the disorder or disease.


As used herein, the term “patient,” “individual,” or “subject” refers to a human or a non-human mammal. Non-human mammals include, for example, livestock and pets, such as ovine, bovine, porcine, canine, feline and marine mammals. Preferably, the patient, subject, or individual is human.


As used herein, the terms “effective amount,” “pharmaceutically effective amount,” and “therapeutically effective amount” refer to a nontoxic but sufficient amount of an agent to provide the desired biological result. That result may be reduction or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. An appropriate therapeutic amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation.


As used herein, the term “pharmaceutically acceptable” refers to a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively non-toxic, i.e., the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.


As used herein, the term “pharmaceutically acceptable salt” refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts of the present disclosure include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present disclosure can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. The phrase “pharmaceutically acceptable salt” is not limited to a mono, or 1:1, salt. For example, “pharmaceutically acceptable salt” also includes bis-salts, such as a bis-hydrochloride salt. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science, 66, 2 (1977), each of which is incorporated herein by reference in its entirety.


As used herein, the term “composition” or “pharmaceutical composition” refers to a mixture of at least one compound useful within the disclosure with a pharmaceutically acceptable carrier. The pharmaceutical composition facilitates administration of the compound to a patient or subject. Multiple techniques of administering a compound exist in the art including, but not limited to, intravenous, oral, aerosol, parenteral, ophthalmic, pulmonary, and topical administration.


The term “pharmaceutical combination” as used herein means a product that results 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 the active ingredients, e.g., a compound of the disclosure and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage. The term “non-fixed combination” means that the active ingredients. e.g. a compound of the disclosure and a co-agent, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the two compounds in the body of the patient. The latter also applies to cocktail therapy, e.g., the administration of three or more active ingredients.


As used herein, the term “pharmaceutically acceptable carrier” means a pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound useful within the disclosure within or to the patient such that it may perform its intended function. Typically, such constructs are carried or transported from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation, including the compound useful within the disclosure, and not injurious to the patient. Some examples of materials that may serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyp's, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; surface active agents; alginic acid; pyrogen-free water; isotonic saline; Ringers solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations.


As used herein, “pharmaceutically acceptable carrier” also includes any and all coatings, antibacterial and antifungal agents, and absorption delaying agents, and the lice that are compatible with the activity of the compound useful within the present disclosure, and are physiologically acceptable to the patient. Supplementary active compounds may also be incorporated into the compositions. The “pharmaceutically acceptable carrier” may further include a pharmaceutically acceptable salt of the compound disclosed herein. Other additional ingredients that may be included in the pharmaceutical compositions are known in the art and described, for example, in Remington's Pharmaceutical Sciences (Genaro, Ed., Mack Publishing Co., 1985, Easton, PA), which is incorporated herein by reference.


As used herein, the term “EGFR” refers to epidermal growth factor receptor (alternately referred to as ErbB-1 or HER1) and may refer to the wild-type receptor or to a receptor containing one or more mutations.


As used herein, the term “HER” or Her” refers to members of the ErbB receptor tyrosine kinase family, including EGFR, ERBB2, HER3, and HER4.


As used herein, the term “allosteric site” refers to a site on EGFR other than the ATP binding site, such as that characterized in a crystal structure of EGFR. An “allosteric site” can be a site that is close to the ATP binding site, such as that characterized in a crystal structure of EGFR. For example, one allosteric site includes one or more of the following amino acid residues of epidermal growth factor receptor (EGFR): Lys745, Leu788, Ala743, Cys755, Leu777, Phe856, Asp855, Met766, Ile759, Glu762, and/or Ala763.


As used herein, the term “agent that prevents EGFR dimer formation,” or iterations thereof, refers to an agent that prevents dimer formation in which the C-lobe of the “activator” subunit impinges on the N-lobe of the “receiver” subunit. Examples of agents that prevent EGFR dimer formation include, but are not limited to, cetuximab, trastuzumab, panitumumab, and Mig6.


As used herein, the term “alkyl,” by itself or as part of another substituent means, unless otherwise stated, a straight or branched chain hydrocarbon having the number of carbon atoms designated (i.e., C1-C6alkyl means an alkyl having one to six carbon atoms) and includes straight and branched chains. Examples include methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert butyl, pentyl, neopentyl, and hexyl. Other examples of C1-C6 alkyl include ethyl, methyl, isopropyl, isobutyl, n-pentyl, and n-hexyl.


As used herein, the term “haloalkyl” refers to an alkyl group, as defined above, substituted with one or more halo substituents, wherein alkyl and halo are as defined herein. Haloalkyl includes, by way of example, chloromethyl, trifluoromethyl, bromoethyl, chlorofluoromethyl, and the like.


As used herein, the term “alkoxy” refers to the group —O-alkyl, wherein alkyl is as defined herein. Alkoxy includes, by way of example, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, t-butoxy and the like.


As used herein, the term “alkenyl” refers to a monovalent group derived from a hydrocarbon moiety containing, in certain embodiments, from two to six, or two to eight carbon atoms having at least one carbon-carbon double bond. The alkenyl group may or may not be the point of attachment to another group. The term “alkenyl” includes, but is not limited to, ethenyl, 1-propenyl, 1-butenyl, heptenyl, octenyl and the like.


As used herein, the term “alkynyl” refers to a monovalent group derived from a hydrocarbon moiety containing, in certain embodiments, from two to six, or two to eight carbon atoms having at least one carbon-carbon triple bond. The alkynyl group may or may not be the point of attachment to another group. The term “alkynyl” includes, but is not limited to, ethynyl, 1-propynyl, 1-butynyl, heptynyl, octynyl and the like.


As used herein, the term “halo” or “halogen” alone or as part of another substituent means, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom, preferably, fluorine, chlorine, or bromine, more preferably, fluorine or chlorine.


As used herein, the term “cycloalkyl” means a non-aromatic carbocylic system that is fully saturated having 1, 2 or 3 rings wherein such rings may be fused. The term “fused” means that a second ring is present (i.e., attached or formed) by having two adjacent atoms in common (i.e., shared) with the first ring. Cycloalkyl also includes bicyclic structures that may be bridged or spirocyclic in nature with each individual ring within the bicycle varying from 3-8 atoms. The term “cycloalkyl” includes, but is not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo[3.1.0]hexyl, spiro[3.3]heptanyl, and bicyclo[1.1.1]pentyl.


As used herein, the term ‘cycloalkenyl’ means a non-aromatic carbocyclic system that is partially saturated having 1, 2 or 3 rings wherein such rings may be fused, and wherein at least one ring contains an sp2 carbon-carbon bond. The term “cycloalkenyt” includes, but is not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, bicyclo[3.1.0]hexenyl, spiro[3.3]heptanenyl, and bicyclo[1.1.1]pentenyl.


As used herein, the term “heterocyclyl” or “heterocycloalkyl” means a non-aromatic carbocyclic system containing 1, 2, 3 or 4 heteroatoms selected independently from N, O, and S and having 1.2 or 3 rings wherein such rings may be fused, wherein fused is defined above. Heterocyclyl also includes bicyclic structures that may be bridged or spirocyclic in nature with each individual ring within the bicycle varying from 3-8 atoms, and containing 0, 1, or 2 N, O, or S atoms. The term “heterocyclyl” includes cyclic esters (i.e., lactones) and cyclic amides (i.e., lactams) and also specifically includes, but is not limited to, epoxidyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl (i.e., oxanyl), pyranyl, dioxanyl, aziridinyl, azetidinyl, pyrrolidinyl, 2,5-dihydro-1H-pyrrolyl, oxazolidinyl, thiazolidinyl, piperidinyl, morpholinyl, piperazinyl, thiomorpholinyl, 1,3-oxazinanyl, 1,3-thiazinanyl, 2-azabicyclo[2.1.1]-hexanyl, 5-azabicyclo[2.1.1]hexanyl, 6-azabicyclo[3.1.1] heptanyl, 2-azabicyclo[2.2.1]-heptanyl, 3-azabicyclo[3.1.1]heptanyl, 2-azabicyclo[3.1.1]heptanyl, 3-azabicyclo[3.1.0]-hexanyl, 2-azabicyclo[3.1.0]hexanyl, 3-azabicyclo[3.2.1]octanyl, 8-azabicyclo[3.2.1]octanyl, 3-oxa-7-azabicyclo[3.3.1]nonanyl, 3-oxa-9-azabicyclo[3.3.1]nonanyl, 2-oxa-5-azabicyclo-[2.2.1]heptanyl, 6-oxa-3-azabicyclo[3.1.1]heptanyl, 2-azaspiro[3.3]heptanyl, 2-oxa-6-azaspiro[3.3]heptanyl, 2-oxaspiro[3.3]heptanyl, 2-oxaspiro[3.5]nonanyl, 3-oxaspiro[5.3]-nonanyl, and 8-oxabicyclo[3.2.1]octanyl.


As used herein, the term “heterocycloalkenyl” means a non-aromatic carbocyclic system containing 1, 2, 3 or 4 heteroatoms selected independently from N, O, and S that is partially saturated having 1, 2 or 3 rings wherein such rings may be fused, and wherein at least one ring contains an sp2 carbon-carbon bond.


As used herein, the term “aromatic” refers to a carbocycle or heterocycle with one or more polyunsaturated rings and having aromatic character, i.e., having (4n+2) delocalized π (pi) electrons, where n is an integer.


As used herein, the term “aryl” means an aromatic carbocyclic system containing 1, 2 or 3 rings, wherein such rings may be fused, wherein fused is defined above. If the rings are fused, one of the rings must be fully unsaturated and the fused ring(s) may be fully saturated, partially unsaturated or fully unsaturated. The term “aryl” includes, but is not limited to, phenyl, naphthyl, indanyl, and 1,2,3,4-tetrahydronaphthalenyl. In some embodiments, aryl groups have 6 carbon atoms. In some embodiments, aryl groups have from six to ten carbon atoms. In some embodiments, aryl groups have from six to sixteen carbon atoms.


As used herein, the term “heteroaryl” means an aromatic carbocyclic system containing 1, 2, 3, or 4 heteroatoms selected independently from N, O, and S and having 1, 2, or 3 rings wherein such rings may be fused, wherein fused is defined above. The term “heteroaryl” includes, but is not limited to, furanyl, thienyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, thiadiazoyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, imidazo[1,2-a]pyridinyl, pyrazolo[1,5-a]pyridinyl, 5,6,7,8-tetrahydroisoquinolinyl, 5,6,7,8-tetrahydroquinolinyl, 6,7-dihydro-5H-cyclopenta[b]pyridinyl, 6,7-dihydro-5H-cyclopenta-[c]pyridinyl, 1,4,5,6-tetrahydrocyclopenta[c]pyrazolyl, 2,4,5,6 tetrahydrocyclopenta[c]pyrazoyl, 5,6-dihydro-4H-pyrrolo[1,2-b]pyrazolyl, 6,7-dihydro-5H-pyrrolo[1,2-b][1,2,4]triazolyl, 5,6,7,8-tetrahydro-[1,2,4]triazolo[1,5-a]pyridinyl, 4,5,6,7-tetrahydropyrazolo[1,5-a]pyridinyl, 4,5,6,7-tetrahydro-1H-indazolyl and 4,5,6,7-tetrahydro-2H-indazolyl.


It is to be understood that if an aryl, heteroaryl, cycloalkyl, or heterocyclyl moiety may be bonded or otherwise attached to a designated moiety through differing ring atoms (i.e., shown or described without denotation of a specific point of attachment), then all possible points are intended, whether through a carbon atom or, for example, a trivalent nitrogen atom. For example, the term “pyridinyl” means 2-, 3- or 4-pyridinyl, the term “thienyl” means 2- or 3-thienyl, and so forth.


As used herein, the phrase “nitrogen protecting group” refers to a functional group bound to a nitrogen atom to obtain chemoselectivity in a subsequent chemical reaction. Examples of nitrogen protecting groups include, but are not limited to, carbobenzyloxy (Cbz), tert-butyloxycarbonyl (Boc), 9-fluorenylmethyloxycarbonyl (Fmoc), acetyl (Ac), benzoyl (Bz), benzyl (Bn), tosyl (Ts), and p-methoxybenzyl (PMB).


As used herein, the term “substituted” means that an atom or group of atoms has replaced hydrogen as the substituent attached to another group.


Compounds

Provided herein are compounds that are covalent inhibitors of epidermal growth factor receptor (EGFR) useful in the treatment of kinase-mediated disorders, including cancer and other proliferation diseases.


In an aspect, provided herein is a compound of Formula I:




embedded image




    • or a pharmaceutical y acceptable salt thereof;


      wherein:

    • X is selected from the group consisting of absent, NH, SO2, and C1-C6 alkyl, wherein alkyl is optionally substituted with one or two R6;

    • A is selected from the group consisting of C6-C10 aryl, 5-10 membered heteroaryl, C3-C10 cycloalkyl, 3-10 membered heterocycloalkyl, C3-C10 cycloalkenyl, and 3-10 membered heterocycloalkenyl;

    • R1 is selected from the group consisting of H, halo, CN, OH, NO2, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 alkyl-N(R7)2, C1-C6 alkyl-OH, N(R7)2, NHC(O)R7, C(O)N(R7)2, NHC(O)N(R7)2, SO2N(R7)2, OC(O)N(R7)2, NHC(O)OR7, C6-C10 aryl, 5-10 membered heteroaryl, C3-C10 cycloalkyl, 3-10 membered heterocycloalkyl, C3-C10 cycloalkenyl, and 4-10 membered heterocycloalkenyl;

    • R3 is selected from the group consisting of C1-C6 alkyl, C2-C6 alkenyl, C6-C10 aryl, 5-10 membered heteroaryl, C3-C10 cycloalkyl, and 3-10 membered heterocycloalkyl, wherein alkyl, aryl, heteroaryl, cycloalkyl, and heterocycloalkyl are each optionally substituted with one or two R6;

    • R4 is selected from the group consisting of H, halo, OH, CN, NO2, NH2, NH(C1-C6 alkyl), N(C1-C6 alkyl)2, C1-C6 alkyl-N(R7)2, C1-C6 alkyl, C1-C6 haloalkyl, C(O)—C1-C6 alkyl, C(O)NH2, C(O)NH—C1-C6 alkyl, C1-C6 alkyl-OH, P(O)(C1-C6 alkyl)2, C6-C10 aryl, 5-10 membered heteroaryl, C3-C10 cycloalkyl, and 3-10 membered heterocycloalkyl; wherein C6-C10 aryl, 5-10 membered heteroaryl, C3-C10 cycloalkyl, and 3-10 membered heterocycloalkyl are each optionally substituted with one or two R6;

    • each R5 is independently selected from the group consisting of C1-C6 alkyl, OH, CN, NO2, C1-C6 haloalkyl, NH2, NH(C1-C6 alkyl), N(C1-C6 alkyl)2, C1-C6 alkyl-N(R7)2, C(O)—C1-C6 alkyl, C(O)NH2, C(O)NH—C1-C6 alkyl, C1-C6 alkyl-OH, P(O)(C1-C6 alkyl)2, C6-C10 aryl, 5-10 membered heteroaryl, C3-C10 cycloalkyl, and 3-10 membered heterocycloalkyl; wherein C6-C10 aryl, 5-10 membered heteroaryl, C3-C10 cycloalkyl, and 3-10 membered heterocycloalkyl are each optionally substituted with one or two R6;

    • alternatively, R6 is







embedded image




    • each R6 is independently selected from the group consisting of halo, OH, C(O)—C1-C6 alkyl, NH2, NH(C1-C6 alkyl), N(C1-C6 alkyl)2, and P(O)(C1-C6 alkyl)2;

    • each R7 is independently selected from the group consisting of H, OH, halo, C1-C6 alkyl, C6-C10 aryl, 5-10 membered heteroaryl, C3-C10 cycloalkyl, and 3-10 membered heterocycloalkyl;

    • R2 is selected from the group consisting of:







embedded image


embedded image


embedded image


embedded image


embedded image




    • L3 is a bond, —NH—, —N(C1-C4 alkyl)-, or C1-C4 alkylene, optionally wherein one or more carbon is independently replaced with —C(O)—, —O—, —S—, —NRL3a—, —NRL3aC(O)—, —C(O)NRL3a—, —SC(O)—, —C(O)S—, —OC(O)—, —C(O)O—, —NRL3aC(S)—, —C(S)NRL3a—, trans-CRL3b═CRL3b—, cis-CRL3b═CRL3b—, —C≡C—, —S(O)—, —S(O)O—, —OS(O)—, —S(O)NRL3a—, —NRL3aS(O)—, —S(O)2—, —S(O)2O—, —OS(O)2—, —S(O)2NRL3a—, or —NRL3aS(O)2—;

    • RL3a is hydrogen, C1-C6 alkyl optionally substituted with R9, or a nitrogen protecting group;

    • RL3b is independently, at each occurrence, selected from the group consisting of hydrogen, halogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3-12 membered heterocycloalkyl, C6-C10 aryl, and 5-8 membered heteroaryl, wherein alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl are optionally substituted with one, two, or three R9;

    • or, alternatively, two RL3b groups, together with the atoms to which they are attached, form a C3-C8 cycloalkyl or 4-7 membered heterocycloalkyl, both of which are optionally substituted with one, two, or three R9;

    • L4 is a bond or C1-C6 alkyl optionally substituted with one, two, or three R9;

    • each of RE1, RE2, RE3, and RE4 is independently selected from the group consisting of hydrogen, halogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocycloalkyl, C6-C10 aryl, and 5-12 membered heteroaryl, CN, CH2OREE, CH2N(REE)2, CH2SREE, OREE, N(REE)2, SREE, wherein alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl are optionally substituted with one, two, or three R9;

    • or, alternatively, RE1, and RE3, or RE2 and RE3, or RE1 and RE2 are joined to form C3-C6 cycloalkyl or 4-7 membered heterocycloalkyl, both of which are optionally substituted with one, two, or three R9;

    • each REE is independently selected from the group consisting of hydrogen, C1-C6 alkyl, C1-C6 alkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3-8 membered heterocycloalkyl, C6-C10 aryl, and 5-10 membered heteroaryl, wherein alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl are optionally substituted with one, two, or three R9;

    • or, alternatively, two REE groups, together with the atom to which they are attached, form 4-7 membered heterocycloalkyl;

    • RE6 is hydrogen, C1-C6 alkyl, or a nitrogen protecting group;

    • each Y is independently O, S, CH2, or NRE7;

    • RE7 is hydrogen, C1-C6 alkyl, or a nitrogen protecting group;

    • each R9 is independently selected from the group consisting of halo, OH, NH2, NH(C1-C6 alkyl), and N(C1-C6 alkyl)2;

    • a is 0, 1, or 2; and

    • z is 1, 2, or 3.





In an aspect of Formula I, or a pharmaceutically acceptable salt thereof,

    • R1 is selected from the group consisting of H, halo, CN, OH, NO2, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 alkyl-N(R7)2, C1-C6 alkyl-OH, N(R7)2, NHC(O)R7, C(O)N(R7)2. NHC(O)N(R7)2, SO2N(R7)2, OC(O)N(R7)2, NHC(O)OR7, C6-C10 aryl, 5-10 membered heteroaryl, C3-C10 cycloalkyl, 3-10 membered heterocycloalkyl, C3-C10 cycloalkenyl, and 4-10 membered heterocycloalkenyl; wherein said aryl, heteroaryl, cycloalkyl, heterocycloalkyl, cycloalkenyl, and heterocycloalkenyl are each optionally substituted with one, two, or three substituents selected from the group consisting of C1-C6 alkyl, OH, halo, OC1-C6 alkyl, and NH2, ═O; and
    • R4 is selected from the group consisting of H, halo, OH, CN, NO2, NH2, NH(C1-C6 alkyl), N(C1-C6 alkyl)2, C1-C6 alkyl-N(R7)2, C1-C6 alkyl, C1-C6 haloalkyl, C(O)—C1-C6 alkyl, C(O)NH2, C(O)NH—C1-C6 alkyl, C1-C6 alkyl-OH, P(O)(C1-C6 alkyl)2, C6-C10 aryl, 5-10 membered heteroaryl, C3-C10 cycloalkyl, and 3-10 membered heterocycloalkyl; wherein C1-C6 alkyl, C6-C10 aryl, 5-10 membered heteroaryl, C3-C10 cycloalkyl, and 3-10 membered heterocycloalkyl are each optionally substituted with one or two R6;
    • each R6 is independently selected from the group consisting of halo, OH, C(O)—C1-C6 alkyl, NH2, NH(C1-C6 alkyl), N(C1-C6 alkyl)2, P(O)(C1-C6 alkyl)2, and 4-6 membered heterocycloalkyl, wherein heterocycloalkyl is optionally substituted with C1-C6 alkyl; and
    • each R7 is independently selected from the group consisting of H, OH, halo, C1-C6 alkyl, C0-C6 alkyl-C6-C10 aryl, C0-C6 alkyl-5-10 membered heteroaryl, C0-C6 alkyl-C3-C10 cycloalkyl, and C0-C6 alkyl-3-10 membered heterocycloalkyl;
    • wherein all other variables are defined supra.


In an embodiment,

    • A is selected from the group consisting of phenyl, 5-6 membered heteroaryl, C3-C8 cycloalkyl, and 3-8 membered heterocycloalkyl;
    • R1 is selected from the group consisting of H, C1-C6 alkyl, and 5-6 membered heteroaryl;
    • R3 is selected from the group consisting of C1-C3 alkyl, C2-C4 alkenyl, SO2(C1-C3 alkyl), phenyl, 5-6 membered heteroaryl, C5-C6 cycloalkyl, and 4-8 membered heterocycloalkyl all of which are optionally substituted with one or two R5;
    • R4 is selected from the group consisting of H, halo, and C1-C3 alkyl;
    • R5 is independently selected from the group consisting of C1-C4 alkyl, OH, NH2, N(C1-C3 alkyl)2, C(O)—C1-C3 alkyl, C(O)NH2, C(O)NH—C1-C4 alkyl, C1-C4 alkyl-OH, P(O)(C1-C3 alkyl)2, phenyl, 5-6 membered heteroaryl, 4-8 membered heterocycloalkyl, C5-C6 cycloalkyl; wherein phenyl, heteroaryl, heterocycloalkyl, and cycloalkyl are each optionally substituted with one or two R6;
    • R6 is independently selected from the group consisting of halo, OH, C(O)—C1-C3 alkyl, NH2, N(C1-C3 alkyl)2, and P(O)(C1-C3 alkyl)2;
    • R2 is selected from the group consisting of




embedded image




    • wherein L3 is a bond, —NH—, or —N(C1-C4 alkyl)-;

    • each of RE1, RE2, and RE3 is independently selected from the group consisting of hydrogen, halogen, and C1-C6 alkyl, wherein alkyl is optionally substituted with one, two, or three R9;

    • each R9 is independently selected from the group consisting of NH2, NH(C1-C6 alkyl), and N(C1-C6 alkyl)2;

    • each Y is independently O or CH2; and

    • a is 1 or 2.





In another embodiment,

    • A is selected from the group consisting of phenyl, piperidine, thiophene, pyrrolidine, isoxazole, pyrrole, pyridine, isothiazole, pyrazole, imidazole, thiazole, and oxazole;
    • R1 is selected from the group consisting of H, C1-C3 alkyl, and 5-6 membered heteroaryl;
    • R3 is selected from the group consisting of C1-C3 alkyl, C2-C4 alkenyl, SO2(C1-C3 alkyl), azetidine, azepane, diazepane, oxazepane, oxepane, piperidine, pyrrolidine, cycloheptane, cyclohexane, phenyl, pyridizine, pyridine, and pyrazine, all of which are optionally substituted with R5;
    • R4 is selected from the group consisting of H, halo, and C1-C3 alkyl;
    • R5 is independently selected from the group consisting of C1-C4 alkyl, OH, NH2, N(C1-C3 alkyl)2, C(O)—C1-C3 alkyl, C(O)NH2, C(O)NH—C1-C4 alkyl, C1-C4 alkyl-OH, P(O)(C1-C3 alkyl)2, phenyl, 5-6 membered heteroaryl, 4-8 membered heterocycloalkyl, C5-C6 cycloalkyl; wherein phenyl, heteroaryl, heterocycloalkyl, and cycloalkyl are each optionally substituted with one or two R6;
    • R6 is independently selected from the group consisting of halo, OH, C(O)—C1-C3 alkyl, NH2, N(C1-C3 alkyl)2, and P(O)(C1-C3 alkyl); and
    • R2 is selected from the group consisting of




embedded image


In another embodiment,

    • A is selected from the group consisting of phenyl, piperidine, thiophene, pyrrolidine, isoxazole, pyrrole, pyridine, isothiazole, pyrazole, imidazole, thiazole, and oxazole;
    • R1 is selected from the group consisting of H, C1-C3 alkyl, and 5-6 membered heteroaryl;
    • R3 is selected from the group consisting of C1-C4 alkyl, C2-C4 alkenyl, SO2(C1-C3 alkyl), azetidine, azepane, diazepane, oxazepane, oxepane, piperidine, pyrrolidine, cycloheptane, cyclohexane, phenyl, pyridizine, pyridine, and pyrazine, all of which are optionally substituted with R5;
    • R4 is selected from the group consisting of H, halo, and C1-C3 alkyl;
    • R5 is independently selected from the group consisting of C1-C4 alkyl, OH, NH2, N(C1-C3 alkyl)2, C(O)—C1-C3 alkyl, C(O)NH2, C(O)NH—C1-C4 alkyl, C1-C4 alkyl-OH, P(O)(C1-C3 alkyl)2, phenyl, 5-6 membered heteroaryl, 4-8 membered heterocycloalkyl, C5-C6 cycloalkyl; wherein phenyl, heteroaryl, heterocycloalkyl, and cycloalkyl are each optionally substituted with one or two R6;
    • R6 is independently selected from the group consisting of halo, OH, C(O)—C1-C3 alkyl, NH2, N(C1-C3 alkyl)2, and P(O)(C1-C3 alkyl)2; and
    • R2 is selected from the group consisting of




embedded image


In yet another embodiment, X is absent. In another embodiment, X is NH. In yet another embodiment, X is SO2. In still another embodiment, X is C1-C3 alkyl optionally substituted with one a two R6.


In an embodiment, A is selected from the group consisting of phenyl, 5-6 membered heteroaryl, C4-C7 cycloalkyl, and 4-10 membered heterocycloalkyl. In still another embodiment, A is 5-6 membered heteroaryl. In an embodiment, A is 4-6 membered heterocycloalkyl. In another embodiment, A is C6-C10 aryl. In yet another embodiment. A is selected from the group consisting of phenyl, piperidine, thiophene, pyrrolidine, isoxazole, pyrrole, pyridine, isothiazole, pyrazole, imidazole, thiazole, and oxazole.


In an embodiment, A is thiophene. In another embodiment, A is piperidine. In yet another embodiment, A is pyrrolidine. In still another embodiment, A is pyrrole. In an embodiment, A is phenyl.


In another embodiment, R1 is selected from the group consisting of H, halo, CN, OH, NO2, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 alkyl-N(R7)2, C1-C6 alkyl-OH, N(R7)2, NHC(O)R7, C(O)N(R7)2, NHC(O)N(R7)2, OC(O)N(R7)2, NHC(O)OR7, C6-C10 aryl, 5-10 membered heteroaryl, C3-C10 cycloalkyl, and 3-10 membered heterocycloalkyl.


In yet another embodiment, R1 is selected from the group consisting of H, halo, CN, OH, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C8 alkyl-N(R7)2, C1-C6 alkyl-OH, N(R7)2, NHC(O)R7, C(O)N(R7)2, NHC(O)N(R7)2, OC(O)N(R7)2, NHC(O)OR7, C6-C10 aryl, 5-10 membered heteroaryl, C3-C10 cycloalkyl, and 3-10 membered heterocycloalkyl.


In still another embodiment, R1 is selected from the group consisting of H, halo, CN, OH, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 alkyl-OH, N(R7)2, NHC(O)R7, C(O)N(R7)2, C6-C10 aryl, 5-10 membered heteroaryl, C3-C10 cycloalkyl, and 3-10 membered heterocycloalkyl.


In an embodiment, R1 is selected from the group consisting of H, 5-6 membered heteroaryl, and C1-C3 alkyl.


In yet another embodiment, R3 is selected from the group consisting of C1-C3 alkyl, C2-C4 alkenyl, SO2(C1-C6 alkyl), phenyl, 5-6 membered heteroaryl, C4-C8cycloalkyl, and 4-8 membered heterocycloalkyl all of which are optionally substituted with one or two R5. In still another embodiment, R3 is 5-9 membered heteroaryl optionally substituted with R5. In an embodiment, R3 is C4-C8 cycloalkyl optionally substituted with R6. In another embodiment, R3 is 4-8 membered heterocycloalkyl optionally substituted with R5.


In still another embodiment, R3 is selected from the group consisting of C1-C4 alkyl. C2-C4 alkenyl, SO2(C1-C3 alkyl), azetidine, azepane, diazepane, oxazepane, oxepane, piperidine, pyrrolidine, cycloheptane, cyclohexane, phenyl, pyridizine, pyridine, and pyrazine, all of which are optionally substituted with R5.


In an embodiment, R4 is selected from the group consisting of H, halo, OH, CN, NH2, NH(C1-C6 alkyl), N(C1-C6 alkyl)2, C1-C6 alkyl, C1-C6 haloalkyl, C(O)—C1-C6 alkyl, C(O)NH2, C(O)NH—C1-C6 alkyl, C6-C10 aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, and 3-8 membered heterocycloalkyl; wherein C6-C10 aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, and 3-8 membered heterocycloalkyl are each optionally substituted with one or two R6.


In another embodiment, R4 is selected from the group consisting of H, halo, OH, CN, NH2, NH(C1-C6 alkyl), N(C1-C6 alkyl)2, C1-C8 alkyl, C1-C6 haloalkyl, C(O)—C1-C6 alkyl, C(O)NH2, and C(O)NH—C1-C6 alkyl. In yet another embodiment, R4 is selected from the group consisting of H, halo, and C1-C3 alkyl.


In another embodiment, each R5 is independently selected from the group consisting of C1-C4 alkyl, OH, NH2, N(C1-C6 alkyl)2, C(O)—C1-C3 alkyl, C(O)NH2, C(O)NH—C1-C4 alkyl, C1-C4 alkyl-OH, P(O)(C1-C3 alkyl)2, phenyl, 5-6 membered heteroaryl, 4-8 membered heterocycloalkyl, C4-C8 cycloalkyl; wherein phenyl, heteroaryl, heterocycloalkyl, and cycloalkyl are each optionally substituted with one or two R6.


In yet another embodiment, R5 is




embedded image


In still another embodiment, each R6 is independently selected from the group consisting of halo, OH, C(O)—C1-C3 alkyl, NH2, N(C1-C3 alkyl)2, and P(O)(C1-C3 alkyl)2.


In an embodiment, R2 is selected from the group consisting of




embedded image


In another embodiment, R2 is selected from the group consisting of




embedded image




    • wherein L3 is a bond, —NH—, or —N(C1-C4 alkyl)-;

    • each of RE1, RE2, and RE3 is independently selected from the group consisting of hydrogen, halogen, and C1-C6 alkyl, wherein alkyl is optionally substituted with one, two, or three R9;

    • each R9 is independently selected from the group consisting of NH2, NH(C1-C6 alkyl), and N(C1-C6 alkyl)2;

    • Y is O; and

    • a is 2.





In yet another embodiment, R2 is




embedded image




    • wherein

    • wherein L3 is a bond, —NH—, or —N(C1-C4 alkyl)-;

    • each of RE1, RE2, and RE3 is independently selected from the group consisting of hydrogen, halogen, and C1-C6 alkyl;

    • Y is O.





In yet another embodiment, R2 is selected from the group consisting of




embedded image


In still another embodiment, R2 is selected from the group consisting of




embedded image


In another embodiment, R2 is selected from the group consisting of




embedded image


In yet another embodiment, the compound of Formula I is a compound of Formula II:




embedded image


or a pharmaceutically acceptable salt thereof.


In still another embodiment, the compound of Formula I is a compound of Formula III:




embedded image


or a pharmaceutically acceptable salt thereof.


In an embodiment,

    • R1 is H or 5-6 membered heteroaryl;
    • R3 is selected from the group consisting of C1-C4 alkyl, C2-C4 alkenyl, SO2(C1-C3 alkyl), azetidine, azepane, diazepane, oxazepane, oxepane, piperidine, pyrrolidine, cycloheptane, cyclohexane, phenyl, pyridizine, pyridine, and pyrazine, all of which are optionally substituted with R5;
    • R4 is selected from the group consisting of H, halo, and C1-C3 alkyl;
    • R5 is independently selected from the group consisting of C1-C4 alkyl, OH, NH2, N(C1-C3 alkyl)2, C(O)—C1-C3 alkyl, C(O)NH2, C(O)NH—C1-C4 alkyl, C1-C4 alkyl-OH, P(O)(C1-C3 alkyl)2, phenyl, 5-6 membered heteroaryl, 4-8 membered heterocycloalkyl, C3-C6 cycloakyl; wherein phenyl, heteroaryl, heterocycloalkyl, and cycloalkyl are each optionally substituted with one or two R6;
    • R6 is independently selected from the group consisting of halo, OH, C(O)—C1-C3 alkyl, NH2, N(C1-C3 alkyl)2, and P(O)(C1-C3 alkyl)2; and
    • R2 is selected from the group consisting of




embedded image




    • wherein, L3 is a bond or —NH—;

    • each of RE1, RE2, and RE3 is independently selected from the group consisting of hydrogen, halogen, and C1-C6 alkyl;

    • each Y is O; and

    • a is 2.





In another embodiment,

    • R2 is selected from the group consisting of




embedded image


In an embodiment, the compound of Formula I is selected from the group consisting of a compound in Table 1.










TABLE 1





Compound



No.
Structure







001


embedded image







002


embedded image







003


embedded image







004


embedded image







005


embedded image







006


embedded image







007


embedded image







008


embedded image







074


embedded image







075


embedded image







076


embedded image











or a pharmaceutically acceptable salt thereof.


In another embodiment, the compound of Formula I is selected from the group consisting of:










TABLE 2





Compound No.
Structure







009


embedded image







010


embedded image







011


embedded image







012


embedded image







013


embedded image







014


embedded image







015


embedded image







016


embedded image







017


embedded image







018


embedded image







019


embedded image







020


embedded image







021


embedded image







022


embedded image







023


embedded image







024


embedded image







025


embedded image







026


embedded image







027


embedded image







028


embedded image







029


embedded image







030


embedded image







031


embedded image







032


embedded image







033


embedded image







034


embedded image







035


embedded image







036


embedded image







037


embedded image







038


embedded image







039


embedded image







040


embedded image







041


embedded image







042


embedded image







043


embedded image







044


embedded image







045


embedded image







046


embedded image







047


embedded image







048


embedded image







049


embedded image







050


embedded image







051


embedded image







052


embedded image







062


embedded image







063


embedded image







064


embedded image







065


embedded image







066


embedded image







067


embedded image







068


embedded image







069


embedded image







070


embedded image







071


embedded image







072


embedded image







073


embedded image







077


embedded image











or a pharmaceutically acceptable salt thereof.


In yet another embodiment, the compound of Formula I is selected from the group consisting of:










TABLE 3





Compound No.
Structure







053


embedded image







054


embedded image







055


embedded image







056


embedded image







057


embedded image







058


embedded image







059


embedded image







060


embedded image







061


embedded image











or a pharmaceutically acceptable salt thereof.


The compounds disclosed herein may exist as tautomers and optical isomers (e.g., enantiomers, diastereomers, diastereomeric mixtures, racemic mixtures, and the like).


It is generally well known in the art that any compound that will be convened in vivo to provide a compound disclosed herein is a prodrug within the scope of the present disclosure.


Compounds provided herein can also include all isotopes of atoms occurring in the intermediates or final compounds. Isotopes include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include tritium and deuterium. One or more constituent atoms of the compounds of the invention can be replaced or substituted with isotopes of the atoms in natural or non-natural abundance. In some embodiments, the compound includes at least one deuterium atom. For example, one or more hydrogen atoms in a compound of the present disclosure can be replaced or substituted by deuterium. In some embodiments, the compound includes two or more deuterium atoms. In some embodiments, the compound includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 deuterium atoms. Synthetic methods for including isotopes into organic compounds are known in the art (Deuterium Labeling in Organic Chemistry by Alan F. Thomas (New York, N.Y., Appleton-Century-Crofts, 1971; The Renaissance of H/D Exchange by Jens Atzrodt, Volker Derdau, Thorsten Fey and Jochen Zimmermann, Angew. Chem. Int. Ed. 2007, 7744-7765; The Organic Chemistry of Isotopic Labelling by James R. Hanson, Royal Society of Chemistry, 2011). Isotopically labeled compounds can used in various studies such as NMR spectroscopy, metabolism experiments, and/or assays.


In the compounds provided herein, any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom. Unless otherwise stated, when a position is designated specifically as “H” or “hydrogen,” the position is understood to have hydrogen at its natural abundance isotopic composition. Also, unless otherwise stated, when a position is designated specifically as “D” or “deuterium”, the position is understood to have deuterium at an abundance that is at least 3000 times greater than the natural abundance of deuterium, which is 0.015% (i.e., at least 45% incorporation of deuterium).


In embodiments, the compounds provided herein have an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).


In an aspect, provided herein is a pharmaceutical composition comprising any one of the compounds disclosed herein, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier.


In an embodiment, the composition further comprises a second active agent. In another embodiment, the second active agent is selected from the group consisting of a MEK inhibitor, a PI3K inhibitor, and an mTor inhibitor. In yet another embodiment, the second active agent prevents EGFR dimer formation in a subject. In still another embodiment, the second active agent is selected from the group consisting of cetuximab, trastuzumab, and panitumumab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib, gefitinib, or erlotinib. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


In another aspect, provided herein are pharmaceutical compositions comprising a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.


In another aspect, provided herein is a method of inhibiting the activity of EGFR, comprising administering to a subject in need thereof an effective amount of a compound of disclosed herein or a pharmaceutical composition comprising a compound disclosed herein and a pharmaceutically acceptable carrier. In an embodiment, the compound targets Cys775 on EGFR.


In another aspect, the pharmaceutical composition further comprises a second active agent, wherein said second active agent prevents EGFR dimer formation, and a pharmaceutically acceptable carrier. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR diner formation is cetuximab.


A compound that binds to an allosteric site in EGFR, such as the compounds of the present disclosure (e.g., the compounds of the formulae disclosed herein), optionally in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, are capable of modulating EGFR activity. In some embodiments, the compounds of the present disclosure are capable of inhibiting or decreasing EGFR activity without a second active agent (e.g., an antibody such as cetuximab, trastuzumab, or panitumumab). In other embodiments, the compounds of the present disclosure in combination with a second active agent. In an embodiment, the second active agent prevents EGFR dimer formation and/or are capable of inhibiting or decreasing EGFR activity. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib, gefitinib or erlotinib. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


Methods of Treatment

In an aspect, provided herein is a method of treating cancer in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound disclosed herein. In an embodiment, the cancer is selected from the group consisting of lung cancer, colon cancer, breast cancer, endometrial cancer, thyroid cancer, glioma, squamous cell carcinoma, and prostate cancer. In another embodiment, the cancer is non-small cell lung cancer (NSCLC).


In another aspect, provided herein is a method of inhibiting a kinase in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound provided herein. In an embodiment, the kinase is EGFR.


In yet another aspect, provided herein is a method of treating or preventing a kinase-mediated disorder in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound of the present disclosure. In an embodiment, the kinase-mediated disorder is resistant to an EGFR-targeted therapy. In another embodiment, the EGFR-treated therapy is selected from the group consisting of gefitinib, erlotinib, osimertinib, CO-1686, and WZ4002.


In some embodiments, the compounds of the present disclosure are capable of modulating (e.g., inhibiting or decreasing) the activity of EGFR containing one or more mutations. In some embodiments, the mutant EGFR contains one or more mutations selected from T790M, L718Q, L844V, V948R, L858R, I941R, C797S, and Del. In other embodiments, the mutant EGFR contains a combination of mutations, wherein the combination is selected from Del/L718Q, Del/L844V, Del/T790M, Del/T790M/L718Q, Del/T790M/L844V, L858R/L718Q, L858R/L844V, L858R/T790M, L858R/T790M/I941R, Del/T790M, Del/T790M/C797S, L858R/T790M/C797S, and L858R/T790M/L718Q. In other embodiments, the mutant EGFR contains a combination of mutations, wherein the combination is selected from Del/L844V, L858R/L844V, L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M, Del/T790M/C797S, and L858R/T790M. In other embodiments, the mutant EGFR contains a combination of mutations, wherein the combination is selected from L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M.


In some embodiments, the compounds of the present disclosure in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, are capable of modulating (e.g., inhibiting or decreasing) the activity of EGFR containing one or more mutations. In some embodiments, the mutant EGFR contains one or more mutations selected from T790M, L718Q, L844V, V948R, L858R, I941R, C797S, and Del. In other embodiments, the mutant EGFR contains a combination of mutations, wherein the combination is selected from Del/L718Q, Del/L844V, Del/T790M, Del/T790M/L718Q, Del/T790M/L844V, L858R/L718Q, L858R/L844V, L858R/T790M, L858R/T790M/I941R, Del/T790M, Del/T790M/C797S, L858R/T790M/C797S, and L858R/T790M/L718Q. In other embodiments, the mutant EGFR contains a combination of mutations, wherein the combination is selected from Del/L844V, L858R/L844V, L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M. In other embodiments, the mutant EGFR contains a combination of mutations, wherein the combination is selected from L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib, gefitinib or erlotinib.


In some embodiments, the compounds of the present disclosure are capable of modulating (e.g., inhibiting or decreasing) the activity of EGFR containing one or more mutations, but do not affect the activity of a wild-type EGFR.


In other embodiments, the compounds of the present disclosure in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, are capable of modulating (e.g., inhibiting or decreasing) the activity of EGFR containing one or more mutations, but do not affect the activity of a wild-type EGFR. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib, gefitinib or erlotinib. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


Modulation of EGFR containing one or more mutations, such as those described herein, but not a wild-type EGFR, provides an approach to the treatment, prevention, or amelioration of diseases including, but not limited to, cancer and metastasis, inflammation, arthritis, systemic lupus erythematosus, skin-related disorders, pulmonary disorders, cardiovascular disease, ischemia, neurodegenerative disorders, liver disease, gastrointestinal disorders, viral and bacterial infections, central nervous system disorders, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, spinal cord injury, and peripheral neuropathy.


In some embodiments, the compounds of the disclosure exhibit greater inhibition of EGFR containing one or more mutations as described herein relative to a wild-type EGFR. In certain embodiments, the compounds of the disclosure 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 a wild-type EGFR. In various embodiments, the compounds of the disclosure exhibit up to 1000-fold greater inhibition of EGFR containing one or more mutations as described herein relative to a wild-type EGFR. In various embodiments, the compounds of the disclosure exhibit up to 10000-fold greater inhibition of EGFR having a combination of mutations described herein (e.g., L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M) relative to a wild-type EGFR.


In other embodiments, the compounds of the disclosure in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, exhibit greater inhibition of EGFR containing one or more mutations as described herein relative to a wild-type EGFR. In certain embodiments, the compounds of the disclosure in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, 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 a wild-type EGFR. In various embodiments, the compounds of the disclosure in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, exhibit up to 1000-fold greater inhibition of EGFR containing one or more mutations as described herein relative to a wild-type EGFR. In various embodiments, the compounds of the disclosure in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, exhibit up to 10000-fold greater inhibition of EGFR having a combination of mutations described herein (e.g., L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M) relative to a wild-type EGFR. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dinner formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib, gefitinib or erlotinib. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


In some embodiments, the compounds of the disclosure exhibit from about 2-fold to about 10-fold greater inhibition of EGFR containing one or more mutations as described herein relative to a wild-type EGFR. In various embodiments, the compounds of the disclosure exhibit from about 10-fold to about 100-fold greater inhibition of EGFR containing one or more mutations as described herein relative to a wild-type EGFR. In various embodiments, the compounds of the disclosure exhibit from about 100-fold to about 1000-fold greater inhibition of EGFR containing one or more mutations as described herein relative to a wild-type EGFR. In various embodiments, the compounds of the disclosure exhibit from about 1000-fold to about 10000-fold greater inhibition of EGFR containing one or more mutations as described herein relative to a wild-type EGFR.


In other embodiments, the compounds of the disclosure in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, exhibit from about 2-fold to about 10-fold greater inhibition of EGFR containing one or more mutations as described herein relative to a wild-type EGFR. In other embodiments, the compounds of the disclosure in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, exhibit from about 10-fold to about 100-fold greater inhibition of EGFR containing one or more mutations as described herein relative to a wild-type EGFR. In other embodiments, the compounds of the disclosure in combination with a second active agent wherein said second active agent prevents EGFR dieter formation exhibit from about 100-fold to about 1000-fold greater inhibition of EGFR containing one or more mutations as described herein relative to a wild-type EGFR. In other embodiments, the compounds of the disclosure in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, exhibit from about 1000 fold to about 10000-fold greater inhibition of EGFR containing one or more mutations as described herein relative to a wild-type EGFR. In other embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dieter formation is cetuximab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib, gefitinib or erlotinib. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


In certain embodiments, the compounds of the disclosure exhibit at least 2-fold greater inhibition of EGFR having a combination of mutations selected from L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M relative to a wild-type EGFR. In certain embodiments, the compounds of the disclosure exhibit at least 3-fold greater inhibition of EGFR having a combination of mutations selected from L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M relative to a wild-type EGFR. In certain embodiments, the compounds of the disclosure exhibit at least 5-fold greater inhibition of EGFR having a combination of mutations selected from L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M relative to a wild-type EGFR.


In certain embodiments, the compounds of the disclosure exhibit at least 10 fold greater inhibition of EGFR having a combination of mutations selected from L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M relative to a wild-type EGFR. In certain embodiments, the compounds of the disclosure exhibit at least 25-fold greater inhibition of EGFR having a combination of mutations selected from L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M relative to a wild-type EGFR. In certain embodiments, the compounds of the disclosure exhibit at least 5-fold greater inhibition of EGFR having a combination of mutations selected from L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M relative to a wild-type EGFR. In certain embodiments, the compounds of the disclosure exhibit at least 100 fold greater inhibition of EGFR having a combination of mutations selected from L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M relative to a wild-type EGFR.


In certain embodiments, the compounds of the disclosure in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, exhibit at least 2-fold greater inhibition of EGFR having a combination of mutations selected from L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M relative to a wild-type EGFR. In certain embodiments, the compounds of the disclosure in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, exhibit at least 3-fold greater inhibition of EGFR having a combination of mutations selected from L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M relative to a wild-type EGFR. In certain embodiments, the compounds of the disclosure in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, exhibit at least 5-fold greater inhibition of EGFR having a combination of mutations selected from L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M relative to a wild-type EGFR.


In certain embodiments, the compounds of the disclosure in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, exhibit at least 10-fold greater inhibition of EGFR having a combination of mutations selected from L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M relative to a wild-type EGFR. In certain embodiments, the compounds of the disclosure in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, exhibit at least 25-fold greater inhibition of EGFR having a combination of mutations selected from L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M relative to a wild-type EGFR. In certain embodiments, the compounds of the disclosure in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, exhibit at least 5-fold greater inhibition of EGFR having a combination of mutations selected from L L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M relative to a wild-type EGFR. In certain embodiments, the compounds of the disclosure in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, exhibit at least 100-fold greater inhibition of EGFR having a combination of mutations selected from L858R/T790M, L858R/T790M/I941R, L858R/T790M/C797S, Del/T790M, Del/T790M/C797S, and L858R/T790M relative to a wild-type EGFR.


In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


In some embodiments, the inhibition of EGFR activity is measured by IC50.


In some embodiments, the inhibition of EGFR activity is measured by EC50.


In some embodiments, the inhibition of EGFR by a compound of the disclosure can be measured via a biochemical assay. By illustrative and non-limiting example, a homogenous time-resolved fluorescence (HTRF) assay may be used to determine inhibition of EGFR activity using conditions and experimental parameters disclosed herein. The HTRF assay may, for example, employ concentrations of substrate (e.g., biotin-Lck-peptide substrate) of about 1 μM; concentrations of EGFR (mutant or WT) from about 0.2 nM to about 40 nM; and concentrations of inhibitor from about 0.000282 μM to about 50 μM. A compound of the disclosure screened under these conditions may, for example, exhibit an IC50 value from about 1 nM to >1 μM; from about 1 nM to about 400 nM; from about 1 nM to about 150 nM; from about 1 nM to about 75 nM; from about 1 nM to about 40 nM; from about 1 nM to about 25 nM; from about 1 nM to about 15 nM; or from about 1 nM to about 10 nM. In certain embodiments, a compound of the disclosure screened under the above conditions for inhibition of EGFR having a mutation or combination of mutations selected from L858R/T790M, L858R, and T790M may, for example, exhibit an IC50 value from about 1 nM to >1 μM; from about 1 nM to about 400 nM; from about 1 nM to about 150 nM; from about 1 nM to about 75 nM; from about 1 nM to about 40 nM; from about 1 nM to about 25 nM; from about 1 nM to about 15 nM; or from about 1 nM to about 10 nM.


In some embodiments, the compounds of the disclosure bind to an allosteric site in EGFR. In some embodiments, the compounds of the disclosure interact with at least one amino acid residue of epidermal growth factor receptor (EGFR) selected from Lys745, Leu788, and Ala 743. In other embodiments, the compounds of the disclosure interact with at least one amino acid residue of epidermal growth factor receptor (EGFR) selected from Cys755, Leu777, Phe856, and Asp855. In other embodiments, the compounds of the disclosure interact with at least one amino acid residue of epidermal growth factor receptor (EGFR) selected from Met766, Ile759, Glu762, and Ala763. In other embodiments, the compounds of the disclosure interact with at least one amino acid residue of epidermal growth factor receptor (EGFR) selected from Lys745, Leu788, and Ala 743; at least one amino acid residue of epidermal growth factor receptor (EGFR) selected from Cys755, Leu777, Phe856, and Asp855; and at least one amino acid residue of epidermal growth factor receptor (EGFR) selected from Met766, Ile759, Glu762, and Ala763. In other embodiments, the compounds of the disclosure do not interact with any of the amino acid residues of epidermal growth factor receptor (EGFR) selected from Met793, Gly796, and Cys797.


In some embodiments, the disclosure provides a compound comprising an allosteric kinase inhibitor, wherein the compound is a more potent inhibitor of a drug-resistant EGFR mutant relative to a wild type EGFR. For example, the compound can be at least about 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or about 100-fold more potent at inhibiting the kinase activity of the drug-resistant EGFR mutant relative to a wild-type EGFR. In some embodiments, the drug-resistant EGFR mutant is resistant to one or more known EGFR inhibitors, including but not limited to gefitinib, erlotinib, Iapatinib, WZ4002, HKI-272, CL-387785, and osimertinib.


In some embodiments, the drug-resistant EGFR mutant comprises a sensitizing mutation, such as Del and L858R.


In some embodiments, the disclosure provides a compound comprising an allosteric kinase inhibitor in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, wherein the compound is a more potent inhibitor of a drug-resistant EGFR mutant relative to a wild type EGFR. For example, the compound in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, can be at least about 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or about 100-fold more potent at inhibiting the kinase activity of the drug-resistant EGFR mutant relative to a wild-type EGFR. In some embodiments, the drug-resistant EGFR mutant is resistant to one or more known EGFR inhibitors, including but not limited to gefitinib, erlotinib, lapatinib, WZ4002, HKI-272, CL-387785, and osimertinib. In some embodiments, the drug-resistant EGFR mutant comprises a sensitizing mutation, such as Del and L858R. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


In some embodiments, the disclosure provides a compound comprising an allosteric kinase inhibitor, wherein the compound inhibits kinase activity of a drug-resistant EGFR mutant harboring a sensitizing mutation (e.g., Del and L858R) and a drug-resistance mutation (e.g., T790M, L718Q, C797S, and L844V) with less than a 10-fold difference in potency (e.g., as measured by IC50) relative to an EGFR mutant harboring the sensitizing mutation but not the drug-resistance mutation. In some embodiments, the difference in potency is less than about 9-fold, 8-fold, 7-fold, 6-fold, 5-fold, 4-fold, 3-fold, or 2-fold.


In other embodiments, the disclosure provides a compound comprising an allosteric kinase inhibitor in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, wherein the compound in combination with the second active agent inhibits kinase activity of a drug-resistant EGFR mutant harboring a sensitizing mutation (e.g., Del and L858R) and a drug-resistance mutation (e.g., T790M, L718Q, C797S, and L844V) with less than a 10-fold difference in potency (e.g., as measured by IC50) relative to an EGFR mutant harboring the sensitizing mutation but not the drug-resistance mutation. In some embodiments, the difference in potency is less than about 9 fold, 8-fold, 7-fold, 6-fold, 5-fold, 4-fold, 3-fold, or 2-fold. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib, gefitinib or erlotinib. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


In some embodiments, the disclosure provides a compound comprising an allosteric kinase inhibitor, wherein the compound is more potent than one or more known EGFR inhibitors, including but not limited to gefitinib, erlotinib, lapatinib, WZ4002, HKI-272, CL-387785, and osimertinib, at inhibiting the activity of EGFR containing one or more mutations as described herein, such as T790M, L718Q, L844V, L858R, C797S, and Del. For example, the compound can be at least about 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or about 100-fold more potent (e.g., as measured by IC50) than gefitinib, erlotinib, lapatinib, WZ4002, HKI-272, CL-387785, and osimertinib at inhibiting the activity of the EGFR containing one or more mutations as described herein.


In other embodiments, the disclosure provides a compound comprising an allosteric kinase inhibitor in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, wherein the compound in combination with the second active agent is more potent than one or more known EGFR inhibitors, including but not limited to gefitinib, erlotinib, lapatinb, WZ4002, HKI-272, CL-387785, and osimertinib, at inhibiting the activity of EGFR containing one or more mutations as described herein, such as T790M, L718Q, L844V, L858R, C797S, and Del. For example, the compound in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, can be at least about 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or about 100-fold more potent (e.g., as measured by IC50) than gefitinib, erlotinib, lapatinib, WZ4002, HKI-272, CL-387785, and osimertinib at inhibiting the activity of the EGFR containing one or more mutations as described herein. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR diner formation is cetuximab. In an embodiment, the second active agent is art ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib, gefitinib or erlotinib. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


In some embodiments, the disclosure provides a compound comprising an allosteric kinase inhibitor, wherein the compound is less potent than one or more known EGFR inhibitors, including but not limited to gefitinib, erlotinib, lapatinib, WZ4002, HKI-272, CL-387785, and osimertinib, at inhibiting the activity of a wild-type EGFR. For example, the compound can be at least about 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or about 100-fold less potent (e.g., as measured by IC50) than gefitinib, erlotinib, lapatinib, WZ4002, HKI-272, CL-387785, and osimertinib, at inhibiting the activity of a wild-type EGFR.


In other embodiments, the disclosure provides a compound comprising an allosteric kinase inhibitor in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, wherein the compound in combination with the second active agent is less potent than one or more known EGFR inhibitors, including but not limited to gefitinib, erlotinib, lapatinib, WZ4002, HKI-272, CL-387785, and osimertinib, at inhibiting the activity of a wild-type EGFR. For example, the compound in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation can be at least about 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or about 100-fold less potent (e.g., as measured by IC50) than gefitinib, erlotinb, lapatinib, WZ4002, HKI-272, CL-387785, and osimertinib, at inhibiting the activity of a wild-type EGFR. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib, gefitinib or erlotinib. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


Potency of the inhibitor can be determined by EC50 value. A compound with a lower EC50 value, as determined under substantially similar conditions, is a more potent inhibitor relative to a compound with a higher EC50 value. In some embodiments, the substantially similar conditions comprise determining an EGFR-dependent phosphorylation level, in vitro or in vivo (e.g., in 3T3 cells expressing a wild type EGFR, a mutant EGFR, or a fragment of any thereof).


Potency of the inhibitor can also be determined by 1050 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 3T3 cells expressing a wild type EGFR, a mutant EGFR, or a fragment of any thereof).


An EGFR sensitizing mutation comprises without limitation L858R, G719S, G719C, G719A, L861Q, a deletion in exon 19 and/or an insertion in exon 20. A drug-resistant EGFR mutant can have without limitation a drug resistance mutation comprising T790M, T854A, L718Q, C797S, or D761 Y.


The selectivity between wild-type EGER 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 WT 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, or Exon 19 deletion/T790M can be used. Proliferation assays are performed at a range of inhibitor concentrations (10 μM, 3 μM, 1.1 μM, 330 nM, 110 nM, 33 nM, 11 nM, 3 nM, 1 nM) and an EC50 is calculated.


An alternative method to measure effects on EGFR activity is to assay EGFR phosphorylation. Wild type 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 NIH-3T3 cells (which do not normally express endogenous EGFR) and the ability of the inhibitor (using concentrations as above) to inhibit EGFR phosphorylation can be assayed. Cells are exposed to increasing concentrations of inhibitor for 6 hours and stimulated with EGF for 10 minutes. The effects on EGFR phosphorylation are assayed by Western Blotting using phospho-specific (Y1068) EGFR antibodies.


In another aspect, the present disclosure relates to a compound that binds to an allosteric site in EGFR, wherein the compound exhibits greater than 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold, 100-fold, or 1000-fold inhibition of EGFR containing one or more mutations as described herein (e.g., L858R/T790M, Del/T790M, Del/T790M/L718Q, L858R/T790M/C797S, Del/T790M/C797S, L858R/T790M/I941R, or L858R/T790M/L718Q) relative to a wild-type EGFR.


In other embodiments, the disclosure provides a compound that binds to an allosteric site in EGFR in combination with a second active agent, wherein said second active agent prevents EGFR dimer formation, wherein the compound in combination with the second active agent greater than 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold, 100-fold, or 1000-fold inhibition of EGFR containing one or more mutations as described herein (e.g., L858R/T790M, Del/T790M, Del/T790M/L718Q, Del/T790M/C797S, L858R/T790M/C797S, L858R/T790M/I941R, or L858R/T790M/L718Q) relative to a wild-type EGER. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib, gefitinib or erlotinib. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


In still another aspect, the disclosure provides a method of inhibiting epidermal growth factor receptor (EGFR), the method comprising administering to a subject in need thereof an effective amount of a compound of disclosed herein, or a pharmaceutically acceptable salt thereof. In some embodiments, the method further comprises administering a second active agent, wherein said second active agent prevents EGFR dimer formation. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib, gefitinib or erlotinib. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


In another aspect, provided herein is a method of treating or preventing a disease, the method comprising administering to a subject in need thereof an effective amount of a compound of disclosed herein, or a pharmaceutically acceptable salt thereof. In some embodiments, the disease is mediated by a kinase. In further embodiments, the kinase comprises a mutated cysteine residue. In further embodiments, the mutated cysteine residue is located in or near the position equivalent to Cys 797 in EGFR, including such positions in Jak3, Blk, Bmx, Btk, HER2 (ErbB2), HER4 (Erb64), Ilk, Tec, and Txk. In some embodiments, the method further comprises administering a second active agent, wherein said second active agent prevents dimer formation of the kinase. In some embodiments, the second active agent that prevents kinase dimer formation is an antibody. In further embodiments, the second active agent prevents EGFR dimer formation. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib, gefitinib or erlotinib. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


In some embodiments, the disease is mediated by EGFR (e.g., EGFR plays a role in the initiation or development of the disease). In some embodiments, the disease is mediated by a Her-kinase. In further embodiments, the Her-kinase is HER1, HER2, or HER4.


In certain embodiments, the disease is resistant to a known EGFR inhibitor, including but not limited to, gefitinib, erlotinib, osimertinib, CO-1686, or WZ4002. In certain embodiments, a diagnostic test is performed to determine if the disease is associated with an activating mutation in EGFR. In certain embodiments, a diagnostic test is performed to determine if the disease is associated with an EGFR harboring an activating mutation and/or a drug resistance mutation. Activating mutations comprise without limitation L858R, G719S, G719C, G719A, L718Q, L861Q, a deletion in exon 19 and/or an insertion in exon 20. Drug resistant EGFR mutants can have without limitation a drug resistance mutation comprising T790M, T854A, L718Q, C797S, or D761Y. The diagnostic test can comprise sequencing, pyrosequencing, PCR, RT-PCR, or similar analysis techniques known to those of skill in the art that can detect nucleotide sequences.


In certain embodiments, the disease is cancer or a proliferation disease.


In further embodiments, the disease is lung cancer, colon cancer, breast cancer, prostate cancer, liver cancer, pancreas cancer, brain cancer, kidney cancer, ovarian cancer, stomach cancer, skin cancer, bone cancer, gastric cancer, breast cancer, pancreatic cancer, glioma, glioblastoma, hepatocellutar carcinoma, papillary renal carcinoma, head and neck squamous cell carcinoma, leukemias, lymphomas, myelomas, or solid tumors. In further embodiments, the disease is lung cancer, breast cancer, glioma, squamous cell carcinoma, or prostate cancer. In still further embodiments, the disease is non-small cell lung cancer.


In certain embodiments, the disease is resistant to a known EGFR inhibitor, including but not limited to, gefitinib, erlotinib, osimertinib, CO-1686, or WZ4002. In certain embodiments, a diagnostic test is performed to determine if the disease is associated with an activating mutation in EGFR. In certain embodiments, a diagnostic test is performed to determine if the disease is associated with an EGFR harboring an activating mutation and/or a drug resistance mutation. Activating mutations comprise without limitation L858R, G719S, G719C, G719A, L718Q, L861Q, a deletion in exon 19 and/or an insertion in exon 20. Drug resistant EGFR mutants can have without limitation a drug resistance mutation comprising T790M, T854A, L718Q, C797S, or D761 Y. The diagnostic test can comprise sequencing, pyrosequencing, PCR. RT-PCR, or similar analysis techniques known to those of skill in the art that can detect nucleotide sequences.


In yet another aspect, provided herein is a method of treating a kinase-mediated disorder comprising administering to a subject in need thereof an effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof. In some embodiments, the compound is an inhibitor of HER1, HER2, or HER4. In other embodiments, the subject is administered an additional therapeutic agent. In other embodiments, the compound and the additional therapeutic agent are administered simultaneously or sequentially.


In another aspect, the disclosure provides a method of treating a kinase mediated disorder, the method comprising administering to a subject in need thereof an effective amount of a compound of disclosed herein, or a pharmaceutically acceptable salt thereof, and a second active agent, wherein said second active agent prevents EGFR dimer formation. In some embodiments, the compound is an inhibitor of HER1, HER2, or HER4. In other embodiments, the subject is administered an additional therapeutic agent. In other embodiments, the compound, the second active agent that prevents EGFR dimer formation, and the additional therapeutic agent are administered simultaneously or sequentially. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib, gefitinib or erlotinib. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


In other embodiments, the disease is cancer. In further embodiments, the cancer is lung cancer, colon cancer, breast cancer, prostate cancer, liver caner, pancreas cancer, brain cancer, kidney cancer, ovarian cancer, stomach cancer, skin cancer, bone cancer, gastric cancer, breast cancer, pancreatic cancer, glioma, gliobastoma, hepatocellular carcinoma, papillary renal carcinoma, head and neck squamous cell carcinoma, leukemias, lymphomas, myelomas, or solid tumors. In further embodiments, the disease is lung cancer, breast cancer, glioma, squamous cell carcinoma, or prostate cancer. In still further embodiments, the disease is non-small cell lung cancer.


In another aspect, provided herein is a method of treating cancer, wherein the cancer cell comprises activated EGFR, comprising administering to a subject in need thereof an effective amount of a compound of disclosed herein, or a pharmaceutically acceptable salt thereof.


In another aspect, provided herein is a method of treating cancer, wherein the cancer cell comprises activated EGFR, comprising administering to a subject in need thereof an effective amount of a compound of disclosed herein, or a pharmaceutically acceptable salt thereof and a second active agent, wherein said second active agent prevents EGFR dimer formation. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib, gefitinib or erlotinib. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


In certain embodiments, the EGFR activation is selected from mutation of EGFR, amplification of EGFR, expression of EGFR, and ligand mediated activation of EGFR.


In further embodiments, the mutation of EGFR is selected from G719S, G719C, G719A, L858R, L861 Q, an exon 19 deletion mutation, and an exon 20 insertion mutation.


In still another aspect, provided herein is a method of treating cancer in a subject, wherein the subject is identified as being in need of EGFR inhibition for the treatment of cancer, comprising administering to the subject an effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof.


In certain embodiments, the subject identified as being in need of EGFR inhibition is resistant to a known EGFR inhibitor, including but not limited to, gefitinib, erlotinib, osimertinib, CO-1686, or WZ4002. In certain embodiments, a diagnostic test is performed to determine if the subject has an activating mutation in EGFR. In certain embodiments, a diagnostic test is performed to determine if the subject has an EGFR harboring an activating mutation and/or a drug resistance mutation. Activating mutations comprise without limitation L858R, G7193, G719C, G719A, L718Q, L861 Q, a deletion in exon 19 and/or an insertion in exon 20. Drug resistant EGFR mutants can have without limitation a drug resistance mutation comprising T790M, T854A, L718Q, C7973, or D761Y. The diagnostic test can comprise sequencing, pyrosequencing, PCR, RT-PCR, or similar analysis techniques known to those of skill in the art that can detect nucleotide sequences.


In an aspect, provided herein is a method of preventing resistance to a known EGFR inhibitor (including but not limited to gefitinib, erlotinib, osimertinib, CO-1686, or WZ4002) in a subject, comprising administering to a subject in need thereof an effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof.


In another aspect, provided herein is a method of preventing resistance to a known EGFR inhibitor (including but not limited to gefitinib, erlotinib, osimertinib, CO-1686, or WZ4002) in a disease, comprising administering to a subject in need thereof an effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof, and a second active agent, wherein said second active agent prevents EGFR dimer formation. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab.


In an embodiment of the methods disclosed herein, the subject is a human.


In another aspect, the disclosure provides a compound disclosed herein, or a pharmaceutically acceptable salt thereof, for use in the manufacture of a medicament for treating or preventing a disease in which EGFR plays a role.


In an aspect, provided herein is a method of treating or preventing a condition selected from the group consisting of autoimmune diseases, inflammatory diseases, proliferative and hyperproliferative diseases, immunologically-mediated diseases, bone diseases, metabolic diseases, neurological and neurodegenerative diseases, cardiovascular diseases, hormone related diseases, allergies, asthma, and Alzheimer's disease. In other embodiments, said condition is selected from a proliferative disorder and a neurodegenerative disorder.


One aspect of this disclosure provides compounds that are useful for the treatment of diseases, disorders, and conditions characterized by excessive or abnormal cell proliferation. Such diseases include, but are not limited to, a proliferative or hyperproliferative disease, and a neurodegenerative disease. Examples of proliferative and hyperproliferative diseases include, without limitation, cancer. The term “cancer” includes, but is not limited to, the following cancers: breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach, skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, small cell carcinoma, lung adenocarcinoma, bone, colon, colorectal, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder carcinoma, liver carcinoma and biliary passages, kidney carcinoma, myeloid disorders, lymphoid disorders, Hodgkin's, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx, small intestine, colon, rectum, large intestine, rectum, brain and central nervous system, chronic myeloid leukemia (CML), and leukemia. The term “cancer” includes, but is not limited to, the following cancers: myeloma, lymphoma, or a cancer selected from gastric, renal, head and neck, oropharangeal, non-small cell lung cancer (NSCLC), endometrial, hepatocarcinoma, non-Hodgkin's lymphoma, and pulmonary.


The term “cancer” refers to any cancer caused by the proliferation of malignant neoplastic cells, such as tumors, neoplasms, carcinomas, sarcomas, leukemias, lymphomas and the like. For example, cancers include, but are not limited to, mesothelioma, leukemias and lymphomas such as cutaneous T-cell lymphomas (CTCL), noncutaneous peripheral T-cell lymphomas, lymphomas associated with human T-cell lymphotrophic virus (HTLV) such as adult T-cell leukemia/lymphoma (ATLL), B-cell lymphoma, acute nonlymphocytic leukemias, chronic lymphocytic leukemia, chronic myelogenous leukemia, acute myelogenous leukemia, lymphomas, and multiple myeloma, non-Hodgkin lymphoma, acute lymphatic leukemia (ALL), chronic lymphatic leukemia (CLL), Hodgkin's lymphoma, Burkitt lymphoma, adult T-cell leukemia lymphoma, acute-myeloid leukemia (AML), chronic myeloid leukemia (CML), or hepatocellular carcinoma. Further examples include myelodysplastic syndrome, childhood solid tumors such as brain tumors, neuroblastoma, retinoblastoma, Wilms' tumor, bone tumors, and soft-tissue sarcomas, common solid tumors of adults such as head and neck cancers (e.g., oral, laryngeal, nasopharyngeal and esophageal), genitourinary cancers (e.g., prostate, bladder, renal, uterine, ovarian, testicular), lung cancer (e.g., small-cell and non-small cell), breast cancer, pancreatic cancer, melanoma and other skin cancers, stomach cancer, brain tumors, tumors related to Gorlin syndrome (e.g., medulloblastoma, meningioma, etc.), and liver cancer. Additional exemplary forms of cancer which may be treated by the subject compounds include, but are not limited to, cancer of skeletal or smooth muscle, stomach cancer, cancer of the small intestine, rectum carcinoma, cancer of the salivary gland, endometrial cancer, adrenal cancer, anal cancer, rectal cancer, parathyroid cancer, and pituitary cancer.


Additional cancers that the compounds described herein may be useful in preventing, treating and studying are, for example, colon carcinoma, familial adenomatous polyposis carcinoma and hereditary non-polyposis colorectal cancer, or melanoma. Further, cancers include, but are not limited to, labial carcinoma, larynx carcinoma, hypopharynx carcinoma, tongue carcinoma, salivary gland carcinoma, gastric carcinoma, adenocarcinoma, thyroid cancer (medullary and papillary thyroid carcinoma), renal carcinoma, kidney parenchyma carcinoma, cervix carcinoma, uterine corpus carcinoma, endometrium carcinoma, chorion carcinoma, testis carcinoma, urinary carcinoma, melanoma, brain tumors such as glioblastoma, astrocytoma, meningioma, medulloblastoma and peripheral neuroectodermal tumors, gall bladder carcinoma, bronchial carcinoma, multiple myeloma, basalioma, teratoma, retinoblastoma, choroidea melanoma, seminoma, rhabdomyosarcoma, craniopharyngeoma, osteosarcoma, chondrosarcoma, myosarcoma, liposarcoma, fibrosarcoma, Ewing sarcoma, and plasmocytoma. In one aspect of the disclosure, the present disclosure provides for the use of one or more compounds of the disclosure in the manufacture of a medicament for the treatment of cancer, including without limitation the various types of cancer disclosed herein.


In some embodiments, the compounds of this disclosure are useful for treating cancer, such as colorectal, thyroid, breast, and lung cancer; and myeloproliferative disorders, such as polycythemia vera, thrombocythemia, myeloid metaplasia with myelofibrosis, chronic myelogenous leukemia, chronic myelomonocytic leukemia, hypereosinophilic syndrome, juvenile myelomonocytic leukemia, and systemic mast cell disease. In some embodiments, the compounds of this disclosure are useful for treating hematopoietic disorders, in particular, acute-myelogenous leukemia (AML), chronic-myelogenous leukemia (CML), acute-promyelocytic leukemia, and acute lymphocytic leukemia (ALL).


The term “cancerous cell” as provided herein, includes a cell afflicted by any one of the above-identified conditions.


The disclosure further provides a method for the treatment or prevention of cell proliferative disorders such as hyperplasias, dysplasias and pre-cancerous lesions. Dysplasia is the earliest form of pre-cancerous lesion recognizable in a biopsy by a pathologist. The subject compounds may be administered for the purpose of preventing said hyperplasias, dysplasias, or pre-cancerous lesions from continuing to expand or from becoming cancerous. Examples of pre-cancerous lesions may occur in skin, esophageal tissue, breast and cervical intra-epithelial tissue.


Examples of neurodegenerative diseases include, without limitation, adrenoleukodystrophy (ALD), Alexander's disease, Alper's disease, Alzheimer's disease, amyotrophic lateral sclerosis (Lou Gehrig's Disease), ataxia telangiectasia, Batten disease (also known as Spielmeyer-Vogt-Sjogren-Batten disease), bovine spongiform encephalopathy (BSE), Canavan disease, Cockayne syndrome, corticobasal degeneration, Creutzfeldt-Jakob disease, familial fatal insomnia, frontotemporal lobar degeneration. Huntington's disease, HIV-associated dementia, Kennedy's disease, Krabbe's disease, Lewy body dementia, neuroborreliosis, Machado-Joseph disease (spinocerebellar ataxia type 3), multiple system atrophy, multiple sclerosis, narcolepsy, Niemann Pick disease, Parkinson's disease, Pelizaeus-Merzbacher disease, Pick's disease, primary lateral sclerosis, prion diseases, progressive supranuclear palsy, Refsum's disease, Sandhoff disease, Schilder's disease, subacute combined degeneration of spinal cord secondary to pernicious anaemia, Spielmeyer-Vogt-Sjogren-Batten disease (also known as Batten disease), spinocerebellar ataxia (multiple types with varying characteristics), spinal muscular atrophy, Steele-Richardson-Olszewski disease, tabes dorsalis, and toxic encephalopathy.


Another aspect of this disclosure provides a method for the treatment or lessening the seventy of a disease selected from a proliferative or hyperproliferative disease, or a neurodegenerative disease, comprising administering an effective amount of a compound, or a pharmaceutically acceptable composition comprising a compound, to a subject in need thereof. In other embodiments, the method further comprises administering a second active agent, wherein said second active agent prevents EGFR dimer formation. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib, gefitinib or erlotinib. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


The activity of the compounds and compositions of the present disclosure as EGFR kinase inhibitors may be assayed in vitro, in vivo, or in a cell line. In vitro assays include assays that determine inhibition of either the kinase activity or ATPase activity of the activated kinase. Alternate in vitro assays quantitate the ability of the inhibitor to bind to the protein kinase and may be measured either by radio labelling the inhibitor prior to binding, isolating the inhibitor/kinase complex and determining the amount of radio label bound, or by running a competition experiment where new inhibitors are incubated with the kinase bound to known radioligands. Detailed conditions for assaying a compound utilized in this disclosure as an inhibitor of various kinases are set forth in the Examples below.


In accordance with the foregoing, the present disclosure further provides a method for preventing or treating any of the diseases or disorders described above in a subject in need of such treatment, which method comprises administering to said subject a therapeutically effective amount of a compound of the disclosure, or a pharmaceutically acceptable salt thereof, and optionally a second active agent, wherein said second active agent prevents EGFR dimer formation. For any of the above uses, the required dosage will vary depending on the mode of administration, the particular condition to be treated and the effect desired.


In other embodiments, the compound and the second active agent that prevents EGFR dimer formation are administered simultaneously or sequentially.


Administration/Dosages/Formulations

Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.


Injectable preparations (for example, sterile injectable aqueous or oleaginous suspensions) may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension, or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P., and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.


In order to prolong the effect of a drug, it is often desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.


Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this disclosure 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 or vaginal cavity and release the active compound.


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 active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings, and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets, and pills, the dosage forms may also comprise buffering agents.


Dosage forms for topical or transdermal administration of a compound of this disclosure include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, ear drops, eye ointments, powders and solutions are also contemplated as being within the scope of this disclosure.


The ointments, pastes, creams and gels may contain, in addition to an active compound of this disclosure, excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.


Powders and sprays can contain, in addition to the compounds of this disclosure, excipients such as lactose, talc, silicic add, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants such as chlorofluorohydrocarbons.


Transdermal patches have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.


According to the methods of treatment of the present disclosure, disorders are treated or prevented in a subject, such as a human or other animal, by administering to the subject a therapeutically effective amount of a compound of the disclosure, in such amounts and for such time as is necessary to achieve the desired result. The term “therapeutically effective amount” of a compound of the disclosure, as used herein, means a sufficient amount of the compound so as to decrease the symptoms of a disorder in a subject. As is well understood in the medical arts a therapeutically effective amount of a compound of this disclosure will be at a reasonable benefit/risk ratio applicable to any medical treatment.


In general, compounds of the disclosure will be administered in therapeutically effective amounts via any of the usual and acceptable modes known in the art, either singly or in combination with one or more therapeutic agents. A therapeutically effective amount may vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors. In general, satisfactory results are indicated to be obtained systemically at daily dosages of from about 0.03 to 2.5 mg/kg per body weight. An indicated daily dosage in the larger mammal, e.g., humans, is in the range from about 0.5 mg to about 100 mg, conveniently administered, e.g., in divided doses up to four times a day or in retard form. Suitable unit dosage forms for oral administration comprise from ca. 1 to 50 mg active ingredient.


In certain embodiments, a therapeutic amount or dose of the compounds of the present disclosure may range from about 0.1 mg/Kg to about 500 mg/Kg, alternatively from about 1 to about 50 mg/Kg. In general, treatment regimens according to the present disclosure comprise administration to a patient in need of such treatment from about 10 mg to about 1000 mg of the compound(s) of this disclosure per day in single or multiple doses. Therapeutic amounts or doses will also vary depending on route of administration, as well as the possibility of co-usage with other agents.


Upon improvement of a subject's condition, a maintenance dose of a compound, composition or combination of this disclosure may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained; when the symptoms have been alleviated to the desired level, treatment should cease. The subject may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.


It will be understood, however, that the total daily usage of the compounds and compositions of the present disclosure will be decided by the attending physician within the scope of sound medical judgment. The specific inhibitory dose for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and lice factors well known in the medical arts.


The disclosure also provides for a pharmaceutical combination, e.g., a kit, comprising a) a first agent which is a compound of the disclosure as disclosed herein, in free form or in pharmaceutically acceptable salt form, and b) at least one co-agent. The kit can comprise instructions for its administration.


In certain embodiments, these compositions optionally further comprise one or more additional therapeutic agents. For example, an agent that prevents EGFR dimer formation, chemotherapeutic agents or other antiproliferative agents may be combined with the compounds of this disclosure to treat proliferative diseases and cancer.


Some examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers; alumina; aluminum stearate; lecithin; serum proteins, such as human serum albumin; buffer substances such as phosphates, glycine, sorbic acid, or 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; polyacrylates; waxes; polyethylenepolyoxypropylene-block polymers; wool fat; sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; glycols, such a propylene glycol or polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar, buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen free water, isotonic saline; Ringer's solution; ethyl alcohol; and phosphate buffer solutions. Further, non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator. The protein kinase inhibitors or pharmaceutical salts thereof may be formulated into pharmaceutical compositions for administration to animals or humans. These pharmaceutical compositions, which comprise an amount of the protein inhibitor effective to treat or prevent a protein kinase-mediated condition and a pharmaceutically acceptable carrier, are other embodiments of the present disclosure.


Kits

In an aspect, provided herein is a kit comprising a compound capable of inhibiting kinase activity selected from one or more compounds of disclosed herein, or pharmaceutically acceptable salts thereof, and instructions for use in treating cancer. In certain embodiments, the kit further comprises components for performing a test to determine whether a subject has activating and/or drug resistance mutations in EGFR.


In another aspect, the disclosure provides a kit comprising a compound capable of inhibiting EGFR activity selected from a compound disclosed herein, or a pharmaceutically acceptable salt thereof.


In another aspect, the disclosure provides a kit comprising a compound capable of inhibiting kinase activity selected from one or more compounds of disclosed herein, or pharmaceutically acceptable salts thereof; a second active agent, wherein said second active agent prevents EGFR dimer formation; and instructions for use in treating cancer. In certain embodiments, the kit further comprises components for performing a test to determine whether a subject has activating and/or drug resistance mutations in EGFR. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab.


In another aspect, the disclosure provides a kit comprising a compound capable of inhibiting EGFR activity selected from a compound of disclosed herein, or a pharmaceutically acceptable salt thereof and a second active agent, wherein said second active agent prevents EGFR dimer formation. In some embodiments, the second active agent that prevents EGFR dimer formation is an antibody. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab, trastuzumab, or panitumumab. In further embodiments, the second active agent that prevents EGFR dimer formation is cetuximab. In an embodiment, the second active agent is an ATP competitive EGFR inhibitor. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib, gefitinib or erlotinib. In another embodiment, the ATP competitive EGFR inhibitor is osimertinib.


The disclosure is further illustrated by the following examples and synthesis schemes, which are not to be construed as limiting this disclosure in scope or spirit to the specific procedures herein described. It is to be understood that the examples are provided to illustrate certain embodiments and that no limitation to the scope of the disclosure is intended thereby. It is to be further understood that resort may be had to various other embodiments, modifications, and equivalents thereof which may suggest themselves to those skilled in the art without departing from the spirit of the present disclosure and/or scope of the appended claims.


EXAMPLES

The application is further illustrated by the following examples, which should not be construed as further limiting. The practice of the present disclosure will employ, unless otherwise indicated, conventional techniques of organic synthesis, cell biology, cell culture, and molecular biology, which are within the skill of the art.


Abbreviations





    • ACN acetonitrile

    • Ac2O acetic anhydride

    • DCM dichloromethane

    • DMF dimethylformamide

    • DIEA diisopropylethylamine

    • EtOAc ethyl acetate

    • HATU (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate

    • LCMS liquid chromatography mass spectrometry

    • MeOH methanol

    • pet. ether petroleum ether

    • SM starting material

    • TEA triethylamine

    • TFA trifluoroacetic acid





Example 1: Synthetic Protocol



embedded image


embedded image


A solution of 2-fluoro-1-methyl-3-nitrobenzene (7.23 g, 46.67 mmol) and tert-butyl (R)-3-aminoazepane-1-carboxylate (10 g, 46.67 mmol) in DMF (80 mL) was added DIEA (30.0 g, 233.3 mmol). The reaction was heated at 120° C. with stirring for 2h. LCMS showed SM was consumed. The reaction was cooled to rt, diluted with EtOAc (800 mL), washed with water (800 mL×2), brine (500 mL×2), dried over Na2SO4. The crude product was purified through silica gel column (EtOAc/pet·ether=1/20) to afford tert-butyl (R)-3-((2-methyl-6-nitrophenyl)amino)-azepane-1-carboxylate (5.1 g, yield 31%) as yellow solid. ESI-MS (EI+, m/z): 350.15




embedded image


To a solution of tert-butyl (R)-3-((2-methyl-6-nitrophenyl)amino)azepane-1-carboxylate (5.1 g, 1 eq mmol) in MeOH (50 mL) was added Pd/C (500 mg, 10 w % wet). The reaction flask was degassed and replaced with H2 (g) three times. The reaction was stirred for 16h at r.t. LCMS showed SM consumed. The reaction was filtered through a silica pad. The cake was washed with EtOAc (100 mL×2). The combined organic phase was concentrated under reduce pressure to afford tert-butyl (R)-3-((2-amino-6-methylphenyl)amino)azepane-1-carboxylate (4.4 g, yield 95%) as yellow solid and used directly in the next step without further purification. ESI-MS (EI+, m/z): 320.19




embedded image


A solution of tert-butyl (R)-3-((2-amino-6-methylphenyl)amino)azepane-1-carboxylate (4.40 g, 144.20 mmol) in methanol (18 mL) was added cyanic bromid (1.82 g, 173.04 mmol) in ACN (6 mL) dropwise at rt. The reaction was heated to 50° C. with stirring for 5h. LCMS showed SM consumed. The reaction was diluted with water (300 mL), concentrated to ⅔ volume. The mixture was extracted with EtOAc (300 mL×3), dried over Na2SO4, concentrated under reducer pressure. The crude product was purified through SGC (EtOAc/pet·ether=1/1 & methanol/DCM=1/10) to afford tert-butyl (R)-3-(2-amino-7-methyl-1H-benzo[d]imidazol-1-yl)azepane-1-carboxylate (2.3 g, yield 51%) as light yellow solid. ESI-MS (EI+, m/z): 345.25; 1H NMR (400 MHz, DMSO-d6) δ 6.98 (dd, J=7.9, 1.2 Hz, 1H), 6.82 (t, J=7.6 Hz, 1H), 6.62 (d, J=7.8 Hz, 1H), 6.02 (d, J=19.5 Hz, 2H), 4.71 (d, J=50.5 Hz, 1H), 3.73 (s, 1H), 3.52 (s, 1H), 2.58 (d, J=6.6 Hz, 3H), 2.23 (d, J=12.8 Hz, 1H), 1.98 (s, 1H), 1.84 (d, J=33.0 Hz, 4H), 1.43 (s, 7H), 1.34-1.28 (m, 4H).




embedded image


A solution of tart-butyl (R)-3-(2-amino-7-methyl-1H-benzo[d]imidazol-1-yl)azepane-1-carboxylate (180 mg, 0.52 mmol), compound 5-acrylamidothiophene-3-carboxylic acid (112 mg, 0.57 mmol), HATU (237 mg, 0.62 mmol) and DIEA (201 mg, 1.56 mmol) in DMF (5 mL) was stirred at 20° C., for 1 h. LCMS showed reaction was completed. The reaction was diluted with EtOAc (60 mL) and washed with brine (3*50 mL). The organic layer was dried over Na2SO4, filtered. The filtrate was concentrated and the residue was purified by prep-TLC to afford compound tert-butyl (R)-3-(2-(5-acrylamidothiophene-3-carboxamido)-7-methyl-1H-benzo[d]imidazol-1-yl)azepane-1-carboxylate (150 mg, 55%) as a light yellow solid.




embedded image


A solution of compound (R)-3-(2-(5-acrylamidothiophene-3-carboxamido)-7-methyl-1H-benzo[d]imidazol-1-yl)azepane-1-carboxylate (150 mg, 0.29 mmol) in DCM (2 mL) and TFA (1 mL) was stirred at 20° C. for 2 h. TLC showed reaction was completed. The reaction was concentrated, the residue was diluted EtOAc (50 mL) and washed with saturated NaHCO3 solution (2×20 mL) and brine (30 mL). The organic layer was concentrated to afford (R)-5-acrylamido-N-(1-(azepan-3-yl)-7-methyl-1H-benzo[d]imidazol-2-yl)thiophene-3-carboxamide (100 mg, 83%) as a yellow solid.




embedded image


To a solution of (R)-5-acrylamido-N-(1-(azepan-3-yl)-7-methyl-1H-benzo[d]imidazol-2-yl)thiophene-3-carboxamide (80 mg, 236 umol) in DCM (2 mL) at rt was added TEA (71 mg, 708 umol) and Ac2O (48 mg, 472 umol). The reaction was stirred for 2 h. LCMS showed SM consumed. The reaction solvent was concentrated under reduce pressure. The crude product was dissolved in EtOAc (20 mL), washed with water (10 mL×20), brine (10 mL×2), dried over Na2SO4, concentrated under reduce pressure. The residue was purified through prep-HPLC to afford 002 (28 mg, yield 25%) as white solid.




embedded image


embedded image


General Procedure for Preparation of tert-butyl (R)-3-((2-nitro-5-(((triisopropylsilyl)oxy)methyl)phenyl)amino)azepane-1-carboxylate



embedded image


A solution of (3-fluoro-4-nitro-phenyl)methoxy-triisopropyl-silane (1.64 g, 5.0 mmol, 1 eq), tert-butyl (3R)-3-aminoazepane-1-carboxylate (1.20 g, 5.60 mmol, 1.12 eq) and DIPEA (969.32 mg, 7.50 mmol, 1.31 mL, 1.5 eq) in ACN (25 mL) was added K2CO3 (138.21 mg, 1.00 mmol, 0.2 eq). The reaction was stirred at 65° C. for 16 hrs. LCMS showed consumption of reactant and formation of the desired product mass. The reaction mixture was poured onto NH4Cl solution (20 mL) and extracted with EtOAc (15 mL*3). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (12 g SepaFlash® Silica Flash Column, Eluent of 0˜15% Ethyl acetate/Petroleum ether gradient @ 40 mL/min) to yield tert-butyl (3R)-3-[2-nitro-5-(triisopropylsilyloxymethyl)anilino]azepane-1-carboxylate (1.43 g, 2.60 mmol, 52.07% yield, 95% purity) as an orange oil.


General Procedure for Preparation of tert-butyl (R)-3-((2-amino-5-(((triisopropylsilyl)oxy)methyl)phenyl)amino)azepane-1-carboxylate



embedded image


To a solution of tert-butyl (3R)-3-[2-nitro-5-(triisopropylsilyloxymethyl)anilino]azepane-1-carboxylate (1.1 g, 2.11 mmol, 1 eq) in THF (30 mL) was added Pd/C (3 g, 10% purity, 1.00 eq) under H2 (2.30 mmol) (50 psi). The mixture was stirred at 25° C. for 3 hr under H2 (2.30 mmol) (50 psi). The reaction was monitored by LCMS which showed the reactant was consumed completely and the desired mass peak was detected. The reaction mixture was filtered through a layer of diatomite and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Plate 1, SiO2, Petroleum ether/Ethyl acetate=2/1) to yield tert-butyl (3R)-3-[2-amino-5-(triisopropylsilyloxymethyl)anilino]azepane-1-carboxylate (300 mg, 610.03 umol, 28.94% yield) as a yellow oil.


General Procedure for Preparation of tert-butyl (3R)-3-[2-amino-6-(triisopropylsilyloxymethyl)benzimidazol-1-yl]azepane-1-carboxylate



embedded image


To a solution of tert-butyl (3R)-3-[2-amino-5-(triisopropylsilyloxymethyl)anilino]azepane-1-carboxylate (2 g, 4.07 mmol, 1 eq) in MeOH (30 mL) was added BrCN (0.510 g, 4.81 mmol, 354.17 uL, 1.18 eq). The mixture was stirred at 50° C. for 4 hr under N2. The reaction was monitored by LCMS which showed complete consumption of reactant and detection of the desired mass peak. The reaction solution was added Na2CO3 solution (50 mL) and stirred under 25° C. for another 1 hr. The reaction mixture was then concentrated under reduced pressure to remove MeOH. The residue was poured onto NaHCO3 solution (50 mL) and extracted with EtOAc 180 mL (60 mL*3). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Plate 1, SiO2, DCM:Methanol=10/1) to yield compound tert-butyl (3R)-3-[2-amino-6-(triisopropylsilyloxymethyl)benzimidazol-1-yl]azepane-1-carboxylate (1.55 g, 3.00 mmol, 73.75% yield) was obtained as a yellow oil.


General Procedure for Preparation of tert-butyl (3R)-3-[2-[(4-nitrothiophene-2-carbonyl)amino]-6-(triisopropylsilyloxymethyl)benzimidazol-yl]azepane-1-carboxylate



embedded image


To a solution of tert-butyl (3R)-3-[2-amino-6-(triisopropylsilyloxymethyl)benzimidazol-1-yl]azepane-1-carboxylate (201 mg, 388.94 umol, 1 eq) in THF (3 mL) were added DIPEA (150.80 mg, 1.17 mmol, 203.24 uL, 3 eq), 4-nitrothiophene-2-carboxylic add (80.81 mg, 466.73 umol, 1.2 eq) and HATU (221.83 mg, 583.41 umol, 1.5 eq). The mixture was stirred at 25° C. for 2 hr. The reaction was monitored by LCMS which showed complete consumption of reactant and detection of the desired mass peak. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was diluted with H2O 10 mL and extracted with EtOAc 30 mL (10 mL*3). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Plate 1, SiO2, Petroleum ether/Ethyl acetate=4/1) to yield compound tert-butyl (3R)-3-[2-[(4-nitrothiophene-2-carbonyl)amino]-6-(triisopropylsilyloxymethyl)benzimidazol-1-yl]azepane-1-carboxylate (400 mg) as a yellow solid.


General Procedure for Preparation of tert-butyl (3R)-3-[6-(hydroxyl methyl)-2-[(4-nitrothiophene-2-carbonyl)amino]benzimidazol-1-yl]azepane-1-carboxylate



embedded image


To a solution of tert-butyl (3R)-3-[2-[(4-nitrothiophene-2-carbonyl)amino]-6-(triisopropylsilyloxymethyl) benzimidazol-1-yl]azepane-1-carboxylate (201 mg, 299.14 umol, 1 eq) in THF (3 mL) was added TBAF (1 M, 603.00 uL, 2.02 eq) (1 M THF) at 0° C. The mixture was stirred at 25° C. for 3 hrs. The reaction was monitored by LCMS and TLC which showed complete consumption of reactant and detection of the desired product. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was diluted with H2O 5 mL and extracted with EtOAc 15 mL (5 mL*3). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give compound tert-butyl (3R)-3-[6-(hydroxyl methyl)-2-[(4-nitrothiophene-2-carbonyl)amino]benzimidazol-1-yl]azepane-1-carboxylate (200 mg, crude) as a yellow oil. The compound is used to the next step without purification.


General Procedure for Preparation of tert-butyl (3R)-3-[6-formyl-2-[(4-nitrothiophene-2-carbonyl)amino]benzimidazol-1-yl] azepane-1-carboxylate



embedded image


To a solution of tert-butyl (3R)-3-[6-(hydroxymethyl)-2-[(4-nitrothiophene-2-carbonyl)amino]benzimidazol-1-yl] azepane-1-carboxylate (200 mg, 387.91 umol, 1 eq) in CHCl3 (9 mL) and THF (3 mL) was added MnO2 (1.01 g, 11.64 mmol, 30 eq). The suspension was degassed under vacuum and purged with N2 3 times. The mixture was stirred at 40° C. for 12 hr under N2. The reaction was monitored by LCMS which showed complete consumption of reactant and detection of the desired mass peak. The reaction mixture was filtered through a layer of diatomite and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Plate 1, SiO2, Petroleum ether/Ethyl acetate=1/1) to yield tert-butyl (3R)-3-[6-formyl-2-[(4-nitrothiophene-2-carbonyl)amino] benzimidazol-1-yl] azepane-1-carboxylate (137 mg, 266.76 umol, 68.77% yield) was obtained as a yellow solid.


General Procedure for Preparation of tert-butyl (3R)-3-[6-[(4-methylpiperazin-1-yl)methyl]-2-[(4-nitrothiophene-2-carbonyl)amino]benzimidazol-1-yl]azepane-1-carboxylate



embedded image


To a solution of tert-butyl (3R)-3-[6-formyl-2-[(4-nitrothiophene-2-carbonyl)amino]benzimidazol-1-yl]azepane-1-carboxylate (137 mg, 266.76 umol, 1 eq) in DCM (30 mL) and MeOH (30 mL) were added 1-methylpiperazine (53.44 mg, 533.53 umol, 59.18 uL, 2 eq), NaBH3CN (83.82 mg, 1.33 mmol, 5 eq) and AcOH (160.20 mg, 2.67 mmol, 152.57 uL, 10 eq). The mixture was stirred at 25° C. for 12 hr. The reaction was monitored by LCMS which showed formation of the desired mass peak. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was diluted with sat. aq. NaHCO3 5 mL and extracted with DCM 15 mL (5 mL*3). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2 column, DCM/MeOH=9/1) to yield compound tert-butyl (3R)-3-[6-[(4-methylpiperazin-1-yl)methyl]-2-[(4-nitrothiophene-2-carbonyl)amino]benzimidazol-1-yl]azepane-1-carboxylate (110 mg, 184.03 umol, 68.99% yield) as a yellow solid.


General Procedure for Preparation of N-[1-[(3R)-1-acetylazepan-3-yl]-6-[(4-methylpiperazin-1-yl) methyl]benzimidazol-2-yl]-4-nitro-thiophene-2-carboxamide



embedded image


A mixture of tert-butyl (3R)-3-[6-[(4-methylpiperazin-1-yl)methyl]-2-[(4-nitrothiophene-2-carbonyl)amino] benzimidazol-1-yl]azepane-1-carboxylate (170 mg, 284.41 umol, 1 eq) in HCl/MeOH (2 mL) (4 M) was stirred at 25° C. for 1 hr. LCMS detected complete consumption of compound 10 and formation of compound 11 peak. The reaction mixture was concentrated under reduced pressure to give a residue. To a solution of the above residue and DIPEA (108.90 mg, 842.58 umol, 146.76 uL, 3 eq) in DCM (3 mL) was added Ac2O (34.41 mg, 337.03 umol, 31.57 uL, 1.2 eq). The mixture was stirred at 25° C. for 12 hr. The reaction was monitored by LCMS which showed complete consumption of reactant and detection of the desired mass peak. The reaction mixture was diluted with sat. aq. NaHCO3 10 mL and extracted with EtOAc 30 mL (10 mL*3). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure and was purified by prep-HPLC (column: Phenomenex C18 80*40 mm*3 um; mobile phase: [water(NH4HCO3)-ACN]; B %: 10%-50%, 8 min) to yield compound N-[1-[(3R)-1-acetylazepan-3-yl]-6-[(4-methylpiperazin-1-yl) methyl]benzimidazol-2-yl]-4-nitro-thiophene-2-carboxamide (70 mg, 129.71 umol, 46.18% yield) as a yellow solid.


General Procedure for Preparation of Compound N-[1-[(3R)-1-acetylazepan-3-yl]-6-[(4-methylpiperazin-1-yl)methyl]benzimidazol-2-yl]-4-amino-thiophene-2-carboxamide



embedded image


To a solution of N-[1-[(3R)-1-acetylazepan-3-yl]-6-[(4-methylpiperazin-1-yl)methyl]benzimidazol-2-yl]-4-nitro-thiophene-2-carboxamide (70 mg, 129.71 umol, 1 eq) in THF (5 mL) and MeOH (5 mL) was added Pd(OH)2 (18.22 mg, 25.94 umol, 20% purity, 0.2 eq) and. The mixture was stirred at 25° C. under H2 (15 psi) for 4 hr. The reaction was monitored by LCMS which showed complete consumption of reactant and detection of the desired mass peak. The reaction mixture was filtered through a layer of diatomite and concentrated under reduced pressure to afford compound N-[1-[(3R)-1-acetylazepan-3-yl]-6-[(4-methylpiperazin-1-yl)methyl]benzimidazol-2-yl]-4-amino-thiophene-2-carboxamide (70 mg, crude) was obtained as a yellow solid. The compound is used to the next step without purification.


General Procedure for Preparation of N-[1-[(3R)-1-acetylazepan-3-yl]-6-[(4-methylpiperazin-1-yl)methyl]benzimidazol-2-yl]-4-(prop-2-enoylamino)thiophene-2-carboxamide



embedded image


To a solution of N-[1-[(3R)-1-acetylazepan-3-yl]-6-[(4-methylpiperazin-1-yl)methyl]benzimidazol-2-yl]-4-amino-thiophene-2-carboxamide (70 mg, 137.35 umol, 1 eq) in dioxane (0.5 mL) and H2O (0.3 mL) was added NaHCO3 (34.61 mg, 412.04 umol, 16.02 uL, 3 eq) and prop-2-enoyl chloride (24.86 mg, 274.69 umol, 2240 uL, 2 eq) at 0° C. under N2. The mixture was stirred at 0° C. for 0.5 hr under N2. The reaction was monitored by LCMS which showed complete consumption of reactant and detection of the desired mass peak. The reaction solution purified by prep-HPLC (column: Waters Xbridge BEH C18 100*30 mm*10 um; mobile phase: [water(NH4HCO3)-ACN]; B %: 10%-45%, 8 min) to yield compound N-[1-[(3R)-1-acetylazepan-3-yl]-6-[(4-methyl]piperazin-1-yl)methyl]benzimidazol-2-yl)-4-(prop-2-enoylamino)thiophene-2-carboxamide (8.59 mg, 15.17 umol, 11.04% yield, 99.55% purity) as a yellow solid. Analytic data HNMR and QC were consistent with the desired product.


The Synthetic Route of 5-cyano-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-4-(prop-2-enoylamino)thiophene-2-carboxamide



embedded image


General Procedure for Preparation of ethyl 5-cyano-4-nitro-thiophene-2-carboxylate



embedded image


To a solution of ethyl 5-bromo-4-nitro-thiophene-2-carboxylate (5 g, 17.85 mmol, 1 eq) in DMF (20 mL) was added CuCN (1.92 g, 21.42 mmol, 4.68 mL, 1.2 eq). The mixture was stirred at 80° C. for 2 hr under N2. The reaction was monitored by TLC and LCMS which showed formation of a new spot and detection of the desired mass peak. The reaction mixture was diluted with brine 50 mL and extracted with EtOAc 100 mL (50 mL*2). The combined organic layers were washed with brine 150 mL (50 mL*3), dried over Na2SO4, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 1/1) to yield compound ethyl 5-cyano-4-nitro-thiophene-2-carboxylate (2.2 g, 9.73 mmol, 54.48% yield) was obtained as a white solid.


General Procedure for Preparation of ethyl 4-amino-5-cyano-thiophene-2-carboxylate



embedded image


To a solution of ethyl 5-cyano-4-nitro-thiophene-2-carboxylate (0.1 g, 442.07 umol, 1 eq) in EtOH (1.5 mL) was added a solution of NH4Cl (23.65 mg, 442.07 umol, 1 eq) in H2O (0.5 mL), then was added Zn (144.53 mg, 2.21 mmol, 5 eq). The mixture was stirred at 20° C. for 1 hr under N2. The reaction was monitored by LCMS which showed complete consumption of reactant and detection of the desired mass peak. The pH of the reaction mixture was adjusted to pH˜7 using 0.5M HCl solution under N2. The mixture was stirred at 20° C. for 1h under N2. The reaction mixture was diluted with brine 20 mL and extracted with EtOAc 60 mL (30 mL*2). The combined organic layers were concentrated under reduced pressure to yield compound ethyl 4-amino-5-cyano-thiophene-2-carboxylate (0.08 g, crude) was obtained as a white solid.


General Procedure for Preparation of 5-cyano-4-amino-thiophene-2-carboxylic acid



embedded image


To a solution of ethyl 5-cyano-4-amino-thiophene-2-carboxylate (2.18 g, 9.64 mmol, 1 eq) in THF (12 mL) and H2O (4 mL) was added LiOH·H2O (1.21 g, 28.91 mmol, 3 eq). The mixture was stirred at 25° C. for 5 hr under N2. The reaction was monitored by TLC and LCMS which showed formation of a new spot and detection of the desired mass peak. The reaction mixture was concentrated under reduced pressure to remove most of THF. The pH of the solution was adjusted to 4˜5 using 0.5M HCl solution. The mixture was then diluted with brine 50 mL and extracted with EtOAc 100 mL (50 mL*2). The combined organic layers were dried over Na2SO4, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, DCM/MeOH=50/1 to 8/1) to yield compound 5-cyano-4-amino-thiophene-2-carboxylic acid (1.1 g, 5.55 mmol, 57.60% yield) as a white solid.


General Procedure for Preparation of 4-amino-5-cyano-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]thiophene-2-carboxamide



embedded image


To a solution of 4-amino-5-cyano-thiophene-2-carboxylic acid (0.3 g, 1.7 mmol, 1 eq) in DMF (2 mL) was added DIPEA (461.10 mg, 3.57 mmol, 621.42 uL, 2 eq) and HATU (746.11 mg, 1.96 mmol, 1.1 eq), then was added 4-(2-amino-7-methylbenzimidazol-1-yl)cyclohexanol (218.81 mg, 891.94 umol, 0.5 eq). The mixture was stirred at 20° C. for 12 hrs under N2. LCMS indicated desired compound was detected. TLC showed one main new spot was formed. The reaction mixture was diluted with H2O (50 mL) and extracted with EtOAc (50 mL×2). The combined organic layers were concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=20/1 to 1/1) to afford 4-amino-5-cyano-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]thiophene-2-carboxamide (0.15 g, 379.29 umol, 21.26% yield) as a white solid.


General Procedure for Preparation of 5-cyano-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-4-(prop-2-enoylamino)thiophene-2-carboxamide



embedded image


To a solution of 4-amino-5-cyano-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]thiophene-2-carboxamide (10 mg, 25.29 umol, 1 eq) in DCM (1 mL) was added TEA (2.56 mg, 25.29 umol, 3.52 uL, 1 eq) and prop-2-enoyl chloride (2.29 mg, 25.29 umol, 2.06 uL, 1 eq). The mixture was stirred at 20° C. for 30 min under N2. LCMS indicated desired compound was detected.


The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by Prep-HPLC (column: Phenomenex Luna 80*30 mm*3 um; mobile phase: [wa2ter (0.1% TFA)-ACN]; B %: 30%-50%, 8 min) to afford 5-cyano-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-4-(prop-2-enoylamino)thiophene-2-carboxamide (2.3 mg, 5.12 umol, 20.23% yield) as a yellow solid.


General Procedure for Preparation of 4-(2-amino-7-methyl-benzimidazol-1-yl)cyclohexanol and ethyl 5-bromo-4-nitro-thiophene-2-carboxylate



embedded image


General Procedure for Preparation of 4-(2-methyl-6-nitro-anilino)cyclohexanol



embedded image


To a solution of 2-fluoro-1-methyl-3-nitro-benzene (50 g, 322.32 mmol, 1 eq) in DMF (500 mL) was added DIPEA (62.48 g, 483.48 mmol, 84.21 mL, 1.5 eq) and trans-4-amino cyclohexanol (37.12 g, 322.32 mmol, eq). The mixture was stirred at 100° C. for 8 hr. The reaction was monitored by TLC and LCMS which showed formation of a new spot and detection of the desired mass peak. The reaction mixture was diluted with H2O 500 mL and extracted with DCM 1500 mL (500 mL*3). The combined organic layers were washed with brine 2000 mL (500 mL*4), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=50/1 to 0/1) to yield compound 4-(2-methyl-6-nitro-anilino)cyclohexanol (60 g, 220.54 mmol, 68.42% yield, 92% purity) as a yellow oil.


General Procedure for Preparation of 4-(2-amino-6-methyl-anilino)cyclohexanol



embedded image


To a solution of Pd/C (6 g, 239.72 mmol, 10% purity, 1 eq) in MeOH (600 mL) was added 4-(2-methyl-6-nitro-anilino)cyclohexanol (60 g, 239.72 mmol, 1 eq). The mixture was stirred at 15° C. for 24 hr under H2 (15 psi). The reaction was monitored by TLC, which indicated complete conversion of the reaction. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=50/1 to 0/1) to yield compound 4-(2-amino-6-methyl-anilino)cyclohexanol (42 g, 171.58 mmol, 71.57% yield, 90% purity) was obtained as a yellow solid.


General Procedure for Preparation of 4-(2-amino-7-methyl benzimidazol-1-yl)cyclohexanol



embedded image


To a solution of 4-(2-amino-6-methyl-anilino)cyclohexanol (21 g, 95.32 mmol, 1 eq) in H2O (60 mL) was added BrCN (14.11 g, 133.21 mmol, 9.80 mL, 1.40 eq) in ACN (60 mL) and MeOH (180 mL) slowly at 5° C. under N2. The mixture was stirred at 50° C. for 2 hr under N2. Then the mixture was added Na2CO3 (10.10 g, 95.32 mmol, 1 eq) in H2O (20 mL). The mixture was stirred at 25° C. for 1 hr under N2. The reaction was monitored by LCMS which indicated complete conversion of the reaction. The reaction mixture was diluted with H2O 100 mL and extracted with EtOAc 600 mL (200 mL*3). The combined organic layers were dried over (Na2SO4), filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, DCM/MeOH=50/1 to 5/1) to yield compound 4-(2-amino-7-methyl-benzimidazol-1-yl)cyclohexanol (9.5 g, 38.73 mmol, 40.63% yield) was obtained as a purple solid.


General Procedure for Preparation of 5-bromo-4-nitro-thiophene-2-carboxylate



embedded image


To a solution of ethyl 5-bromothiophene-2-carboxylate (50 g, 212.68 mmol, 1 eq) in H2SO4 (250 mL, 98%) was added KNO3 (26.9 g, 266.07 mmol, 1.25 eq) slowly at 0° C. under N2. The mixture was stirred at 15° C. for 4 hr under N2. The reaction was monitored by TLC and LCMS which showed formation of a new spot and detection of the desired mass peak. The reaction mixture was dropwise added to aq Na2CO3 solution (˜500 mL) at 0° C. to adjust the pH=5˜7 and stirred at 15° C. for 1 hr under N2. The mixture was filtered and the filter cake was collected and dried under reduced pressure to yield compound ethyl 5-bromo-4-nitro-thiophene-2-carboxylate (40 g, 128.53 mmol, 60.43% yield, 90% purity) as a pale yellow solid.


The Synthetic Route of 5-[(3S)-3-hydroxyazepan-1-yl]-N-[1-(4-hydroxy cyclohexyl)-7-methyl-benzimidazol-2-yl]-4-(prop-2-enoylamino)thiophene-2-carboxamide



embedded image


embedded image


General Procedure for Preparation of 5-[(3S)-3-hydroxyazepan-1-yl]-4-nitro-thiophene-2-carboxylate



embedded image


To a solution of tert-butyl (3S)-3-hydroxyarepane-1-carboxylate (3.5 g, 16.26 mmol, 1 eq) in EtOAc (10 mL) was added HCl/EtOAc (4 M, 10 mL, 2.46 eq). The mixture was stirred at 20° C. for 10 min under N2. The reaction was monitored by LCMS which showed consumption of compound 1 mass and detection of compound 2 mass peak. The reaction mixture was concentrated under reduced pressure to yield (3S)-azepan-3-ol (3 g, 19.78 mmol, 1 eq, HCl) as a white solid.


To a solution of (3S)-azepan-3-ol (3 g, 19.78 mmol, 1 eq, HCl) in ACN (30 mL) was added TEA (8.01 g, 79.14 mmol, 11.01 mL, 4 eq) and ethyl 5-bromo-4-nitro-thiophene-2-carboxylate (5.54 g, 19.78 mmol, 1 eq). The mixture was stirred at 80° C. for 12 hr under N2. The reaction was monitored by TLC and LCMS which showed formation of a new spot and detection of the desired mass peak.


The reaction mixture was diluted with H2O 100 mL and extracted with EtOAc 200 mL (100 mL*2). The combined organic layers were dried over Na2SO4, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=0/1) to yield compound ethyl 5-[(3S)-3-hydroxyazepan-1-yl]-4-nitro-thiophene-2-carboxylate (2.2 g, 7.00 mmol, 35.37% yield) was obtained as a white solid.


General Procedure for Preparation of 5-[(3S)-3-hydroxyazepan-1-yl]-4-nitro-thiophene-2-carboxylic acid



embedded image


To a solution of ethyl 5-[(3S)-3-hydroxyazepan-1-yl]-4-nitro-thiophene-2-carboxylate (2.2 g, 7.00 mmol, 1 eq) in THF (9 mL) and H2O (3 mL) was added LiOH·H2O (881.04 mg, 21.00 mmol, 3 eq). The mixture was stirred at 20° C. for 1 hr. The reaction was monitored by TLC and LCMS which showed formation of a new spot and detection of the desired mass peak.


The reaction mixture was concentrated under reduced pressure to remove most of THF. The pH of the solution was adjusted to 3˜4 using 1M HCl solution. The mixture was then diluted with brine 50 mL and extracted with EtOAc 100 mL (50 mL*2). The combined organic layers were dried over Na2SO4, and concentrated under reduced pressure to yield compound 5-[(3S)-3-hydroxyazepan-1-yl]-4-nitro-thiophene-2-carboxylic acid (1.1 g, 3.84 mmol, 54.90% yield) as a white solid. The compound was directly used to the next step without further purification.


General Procedure for Preparation of 5-[(3S)-3-hydroxyazepan-1-yl]-N-[1-(4-hydroxycyclohexyl)-7 methyl-benzimidazol-2-yl]-4-nitro-thiophene-2-carboxamide



embedded image


To a solution of 5-[(3S)-3-hydroxyazepan-1-yl]-4-nitro-thiophene-2-carboxylic acid (1.1 g, 3.84 mmol, 1.1 eq) in DMF (10 mL) was added DIPEA (1.35 g, 10.48 mmol, 1.83 mL, 3 eq), HATU (1.59 g, 4.19 mmol, 12 eq) and 4-(2-amino-7-methyl-benzimidazol-1-yl)cyclohexanol (856.85 mg, 3.49 mmol, 1 eq). The mixture was stirred at 20° C. for 12 hr under N2. The reaction was monitored by TLC and LCMS which showed formation of a new spot and detection of the desired mass peak.


The reaction mixture was diluted with H2O 10 mL and extracted with EtOAc 20 mL (10 mL*2). The combined organic layers were washed with brine 15 mL (5 mL*3), dried over Na2SO4, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, DCM/MeOH=50/1 to 10/1) to yield compound 5-[(3S)-3-hydroxyazepan-1-yl]-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-4-nitro-thiophene-2-carboxamide (0.3 g, 584.10 umol, 16.72% yield) as a yellow solid.


General Procedure for Preparation of 4-amino-5-[(3S)-3-hydroxyoxepan-1-yl]-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]thiophene-2-carboxamide



embedded image


To a solution of 5-[(3S)-3-hydroxyazepan-1-yl]-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-4-nitro-thiophene-2-carboxamide (50 mg, 97.35 umol, 1 eq) in EtOH (1.5 mL) was added a solution of NH4Cl (5.21 mg, 97.35 umol, 1 eq) in H2O (0.5 mL), the mixture was then added Zn (31.83 mg, 486.75 umol, 5 eq). The mixture was stirred at 20° C. for 2 hr under N2. The reaction was monitored by LCMS which showed complete consumption of reactant and detection of the desired mass peak.


The pH of the reaction mixture was adjusted to PH˜7 using 0.5M HCl solution under N2. The mixture was stirred at 20° C. for 1 h under N2. The reaction mixture was diluted with brine 20 mL and extracted with EtOAc 60 mL (30 mL*2). The combined organic layers were concentrated under reduced pressure to yield 4-amino-5-[(3S)-3-hydroxyazepan-1-yl]-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]thiophene-2-carboxamide (30 mg, crude) as a brown solid.


General Procedure for Preparation of 5-[(3S)-3-hydroxyazepan-1 yl]-N-[1-(4-hydroxy cyclohexyl)-7-methyl-benzimidazol-2-yl]-4-(prop-2-enoylamino)thiophene-2-carboxamide



embedded image


To a solution of 4-amino-5-[(3S)-3-hydroxyazepan-1-yl]-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]thiophene-2-carboxamide (30 mg, 62.03 umol, 1 eq) and DIPEA (24.05 mg, 186.09 umol, 32.41 uL, 3 eq) in DCM (2 mL) was added prop-2-enoyl chloride (6.18 mg, 68.23 umol, 5.56 uL, 1.1 eq) at 0° C. The mixture was stirred at 0° C. for 2 hr under N2. The reaction was monitored by LCMS which showed complete consumption of reactant and detection of the desired mass peak.


The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Waters Xbridge BEH C18 100*25 mm*5 um; mobile phase: [water(10 mM NH4HCO3)-ACN]; B %: 25%˜55%, 10 min) to yield compound 5-[(3S)-3-hydroxyazepan-1-yl]-N-[1-(4-hydroxy cyclohexyl)-7-methyl-benzimidazol-2-yl]-4-(prop-2-enoylamino)thiophene-2-carboxamide (5 mg, 9.30 umol, 14.99% yield) was obtained as a yellow solid. Analytic data HNMR and QC is consistent with the desired structure.



1H NMR (DMSO-d6) δ: 7.85-8.05 (m, 1H), 7.60 (br d, 1H), 7.36 (br d, 1H), 6.94-7.09 (m, 1H), 6.66-6.93 (m, 1H), 6.31-6.47 (m, 1H), 6.13-6.30 (m, 1H), 5.71 (br d, 1H), 5.50-5.67 (m, 1H), 4.56-4.75 (m, 2H), 3.90 (br s, 1H), 2.89-3.21 (m, 8H), 2.63 (br s, 3H), 2.59-2.59 (m, 1H), 1.73 (br d, 8H), 1.24-1.55 (m, 4H)


MS (ESI); m/z=538.2 [M+H]+


The Synthetic Route of N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-5-(3-oxopiperazin-1-yl)-4-(prop-2-enoylamino)thiophene2-carboxamide



embedded image


General Procedure for Preparation of 5-bromo-4-nitro-thiophene-2-carboxylic acid



embedded image


A mixture of ethyl 5-bromo-4-nitro-thiophene-2-carboxylate (2 g, 7.14 mmol, 1 eq), KOH (801.24 mg, 14.28 mmol, 2 eq) in EtOH (20 mL) and H2O (10 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 15° C. for 1 hr under N2 atmosphere. The reaction was monitored by LCMS which showed complete consumption of reactant and detection of the desired mass peak.


The reaction mixture was concentrated under reduced pressure. The reaction mixture was partitioned between HCl (50 mL, 1N) and EtOAc (50 mL×2). The organic phase was separated, washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate at 50/1 to 0/1) to afford 5-bromo-4-nitro-thiophene-2-carboxylic acid (1.5 g, 5.95 mmol, 83.35% yield) as a white solid.


General Procedure for Preparation of 4-nitro-5-(3-oxopiperazin-1-yl)thiophene-2-carboxylic acid



embedded image


A mixture of 5-bromo-4-nitro-thiophene-2-carboxylic acid (1.5 g, 5.95 mmol, 1 eq), piperazin-2-one (893.77 mg, 8.93 mmol, 1.5 eq), TEA (1.81 g, 17.85 mmol, 2.49 mL, 3 eq) in THF (15 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 80° C. for 12 hr under N2 atmosphere. TLC indicated 5-bromo-4-nitro-thiophene-2-carboxylic acid was consumed completely. The reaction mixture was concentrated under reduced pressure to get the residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=1/0 to 0/1) to afford 4-nitro-5-(3-oxopiperazin-1-yl)thiophene-2-carboxylic acid (1 g, 3.69 mmol, 61.95% yield) as a yellow solid.


General Procedure for Preparation of N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-4-nitro-5-(3-oxopiperazin-1-yl)thiophene-2-carboxamide



embedded image


A mixture of 4-nitro-5-(3-oxopiperazin-1-yl)thiophene-2-carboxylic acid (1 g, 3.69 mmol, 1.3 eq), 4(2-amino-7-methylbenzimidazol-1-yl)cyclohexanol (695.70 mg, 2.84 mmol, 1 eq), DIPEA (1.10 g, 8.51 mmol, 1.48 mL, 3 eq), HATU (1.62 g, 4.25 mmol, 1.5 eq) in DMF (20 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 15° C. for 12 hr under N2 atmosphere. The reaction was monitored by TLC and LCMS which showed one main new spot was formed and detection of the desired mass peak.


The reaction mixture was partitioned between DCM (50 mL*2) and H2O (50 mL). The organic phase was separated, washed with brine (50 mL), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=1/0 to 0/1) to afford N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-4-nitro-5-(3-oxopiperazin-1-yl)thiophene-2-carbxamide (350 mg, 702.03 umol, 24.76% yield) was obtained as a yellow solid.


General Procedure for Preparation of 4-amino-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-5-(3-oxopiperazin-1-yl)thiophene-2-carboxamide



embedded image


To a solution of N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-4-nitro-5-(3-oxopiperazin-1-yl) thiophene-2-carboxamide (100 mg, 200.58 umol, 1 eq) in EtOH (1.5 mL) was added a solution of NH4Cl (10.73 mg, 200.58 umol, 1 eq) in H2O (1.5 mL), then was added Zn (65.58 mg, 1.00 mmol, 5 eq) slowly under N2. The mixture was stirred at 20° C. for 2 hr under N2. The reaction was monitored by TLC and LCMS which showed one main new spot was formed and detection of the desired mass peak.


The mixture was adjusted pH=7 with HCl (1M) under N2. The mixture was stirred at 20° C. for 1 h under N2. The reaction mixture was diluted with H2O (20 mL) and extracted with EtOAc (30 mL×2). The combined organic layers were concentrated under reduced pressure to afford 4-amino-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-5-(3-oxopiperazin-1-yl)thiophene-2-carboxamide (55.8 g, crude) as a white solid.


General Procedure for Preparation of N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-5-(3-oxopiperazin-1-yl)-4-(prop-2-enoylamino)thiophene2-carboxamide



embedded image


To a solution of 4-amino-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-5-(3-oxopiperazin-1-yl)thiophene-2-carboxamide (60 mg, 128.05 umol, 1 eq) in DCM (2 mL) was added DIPEA (49.65 mg, 384.15 umol, 66.91 uL, 3 eq) and prop2-enoyl chloride (12.75 mg, 140.85 umol, 11.49 uL, 1.1 eq). The mixture was stirred at 20° C. for 2 hr under N2. LCMS indicated the desired product mass peak was detected.


The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by Prep-HPLC (column: Waters Xbridge BEH C18 100*25 mm*5 um; mobile phase: [water(10 mM NH4HCO3)-ACN]; B %: 20%-50%, 10 min) to afford N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-5-(3-oxopiperazin-1-yl)-4-(prop-2-enoylamino)thiophene2-carboxamide (1.4 mg, 2.68 umol, 2.09% yield) as a pale yellow solid. Analytic data HNMR and QC is consistent with the desired structure.


The Synthetic Route of N-[1(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-5-[methyl (pyrimidin-2-ylmethyl)amino]-4-(prop-2-enoylamino)thiophene-2-carboxamide



embedded image


embedded image


General Procedure for Preparation of ethyl 4-nitro-5-(pyrimidin-2-ylmethytamino) thiophene-2-carboxylate



embedded image


To a solution of pyrimidin-2-ylmethanamine (4 g, 36.65 mmol, 1 eq) in THF (40 mL) was added TEA (4.45 g, 43.98 mmol, 6.12 mL, 1.2 eq) and ethyl 5-bromo-4-nitro-thiophene-2-carboxylate (10.27 g, 36.65 mmol, 1 eq). The mixture was stirred at 80° C. for 8 hr under N2. The reaction was monitored by TLC and LCMS which shaved formation of a new spot and detection of the desired mass peak.


The reaction mixture was diluted with H2O 100 mL and extracted with EtOAc 200 mL (100 mL*2). The combined organic layers were washed with brine 150 mL (50 mL*3), dried over Na2SO4, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=3/1 to 1/6) to yield compound ethyl 4-nitro-5-(pyrimidin-2-ylmethylamino) thiophene-2-carboxylate (3.5 g, 11.35 mmol, 30.97% yield) was obtained as a white solid.


General Procedure for Preparation of ethyl 5-[methyl(pyrimidin-2-ylmethyl) amino]-4-nitrothiophene-2-carboxylate



embedded image


A solution of ethyl 4-nitro-5-(pyrimidin-2-ylmethylamino) thiophene-2-carboxylate (3.5 g, 11.35 mmol, 1 eq) in DMF (50 mL) was added was added NaH (544.8 mg, 13.62 mmol, 60% purity, 1.2 eq) at 0° C. The mixture was then stirred for 30 min. Then Mel (1.77 g, 12.48 mmol, 776.92 uL, 1.1 eq) was added to the reaction mixture. The reaction was then stirred at at 25° C. for 4 hr under N2. The reaction was monitored by LCMS which showed complete consumption of reactant and detection of the desired mass peak.


The reaction mixture was slowly poured onto brine 50 mL and extracted with EtOAc 90 mL (30 mL*3). The combined organic layers were washed with brine 150 mL (50 mL*3), dried over Na2SO4, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=3/1 to 1/9) to yield compound ethyl 5-[methyl(pyrimidin-2-ylmethyl) amino]-4-nitrothiophene-2-carboxylate (1.9 g, 5.89 mmol, 52% yield) as a light yellow solid.


General Procedure for Preparation of 5-[methyl(pyrimidin-2-ylmethyl)amino]-4-nitro-thiophene-2-carboxylic acid



embedded image


To a solution of ethyl 5-[methyl(pyrimidin-2-ylmethyl) amino]-4-nitrothiophene-2-carboxylate (1.9 g, 5.89 mmol, 1 eq) in THF (20 mL) and H2O (10 mL) was added LiOH·H2O (1.05 g, 17.67 mmol, 3 eq). The mixture was stirred at 205° C. for 5 hr. The reaction was monitored by LCMS which showed detection of the desired mass peek.


The reaction mixture was concentrated under reduced pressure to remove most of THF. The pH of the solution was adjusted to 3˜4 using 1M HCl solution. The mixture was then diluted with brine 50 mL and extracted with EtOAc 60 mL (20 mL*3). The combined organic layers were dried over Na2SO4, and concentrated under reduced pressure to yield compound 5-[methyl(pyrimidin-2-ylmethyl)amino]-4-nitro-thiophene-2-carboxylic acid (0.58 g crude) as yellow solid. The product was directly used to the next step without further purification.


General Procedure for Preparation of N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-5-[methyl(pyrimidin-2-ylmethyl) amino]-4-nitro-thiophene-2-carboxamide



embedded image


To a solution of 5-[methyl(pyrimidin-2-ylmethyl)amino]-4-nitro-thiophene-2-carboxylic acid (0.58 g, 1.97 mmol, 1 eq) and 4-(2-amino-7-methyl-benzimidazol-1-yl)cyclohexanol (483.49 mg, 1.97 mmol, 1 eq) in DMF (5 mL) was added HATU (824.32 mg, 2.17 mmol, 1.1 eq) and DIPEA (509.44 mg, 3.94 mmol, 686.58 uL, 2 eq). The mixture was stirred at 25° C. for 12 hr under N2. The reaction was monitored by LCMS which showed detection of the desired mass peak. The reaction mixture was diluted with H2O 10 mL and extracted with EtOAc 20 mL (10 mL*2). The combined organic layers were washed with brine 15 mL (5 mL*3), dried over Na2SO4, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, DCM/MeOH=10/1) to yield compound N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-5-[methyl(pyrimidin-2-ylmethyl) amino]-4-nitro-thiophene-2-carboxamide (0.2 g, 383.44 umol, 19.46% yield) was obtained as a white solid.


General Procedure for Preparation of 4-amino-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-5-[methyl(pyrimidin-2-ylmethyl)amino]thiophene-2-carboxamide



embedded image


To a solution of N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-5-[methyl (pyrimidin-2-ylmethyl) amino]-4-nitro-thiophene-2-carboxamide (40 mg, 76.69 umol, 1 eq) in EtOH (1.5 mL) was added a solution of NH4Cl (4.10 mg, 76.69 umol, 1 eq) in H2O (0.5 mL), then was added Zn (25.07 mg, 383.44 umol, 5 eq) slowly under N2. The mixture was stirred at 20° C. for 2 hr under N2. The reaction was monitored by TLC and LCMS which showed one main new spot was formed and detection of the desired mass peak.


The reaction mixture was added 1M HCl and stirred at 20° C. for 1h under N2. The reaction was added NaHCO3 solution to PH=7 under N2, diluted with H2O 20 mL and extracted with EtOAc 40 Ml (20 Ml*2). The combined organic layers were concentrated under reduced pressure to give compound 4-amino-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-5-[methyl(pyrimidin-2-ylmethyl)amino]thiophene-2-carboxamide (30 mg, crude) was obtained as a white solid.


General Procedure for Preparation of N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-5-[methyl (pyrimidin-2-ylmethyl)amino]-4-(prop-2-enoylamino)thiophene-2-carboxamide



embedded image


To a solution of 4-amino-N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-5-[methyl(pyrimidin-2-yl methyl)amino]thiophene-2-carboxamide (30 mg, 61.02 umol, 1 eq) in DCM (2 Ml) was added DIPEA (23.66 mg, 183.07 umol, 31.89 Ul, 3 eq) and prop-2-enoyl chloride (6.08 mg, 67.13 umol, 5.47 Ul, 1.1 eq). The mixture was stirred at 0° C. for 5 min under N2. The reaction was monitored by LCMS which showed complete consumption of reactant and detection of the desired mass peak.


The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Waters Xbridge BEH C18 100*25 mm*5 um; mobile phase: [water(10 Mm NH4HCO3)-CAN]; B %: 20%-50%, 10 min) to yield compound N-[1-(4-hydroxycyclohexyl)-7-methyl-benzimidazol-2-yl]-5-[methyl (pyrimidin-2-ylmethyl)amino]-4-(prop-2-enoylamino)thiophene-2-carboxamide (5.2 mg, 9.53 umol, 15.62% yield) was obtained as a pale yellow solid. Analytic data HNMR and QC is consistent with the desired structure.















Compound

LCMS



No
Structure
(M + H)

1H NMR








057


embedded image


438.58

1H NMR (500 MHz, DMSO) δ 12.65 (s, 1H), 7.38 (d, J = 8.1 Hz, 1H), 7.12- 7.04 (m, 2H), 6.98 (d, J = 7.5 Hz, 1H), 6.69-6.53 (m, 1H), 6.14 (dt, J = 16.8, 2.8 Hz, 1H), 5.66 (ddd, J = 10.6, 6.6, 2.7 Hz, 1H), 4.76 (s, 1H), 4.21-3.96 (m, 2H), 3.93-3.56 (m, 6H), 3.56- 3.36 (m, 3H), 3.19 (dd, J = 13.5, 6.6 Hz, 1H), 3.13-3.05 (m, 1H), 2.63 (s, 3H), 2.60-2.54 (m, 1H), 2.22 (dd, J = 12.6, 6.9 Hz, 1H), 2.14 (dd, J = 13.9, 6.9 Hz, 1H), 2.08-2.05 (s, 3H), 1.95- 1.87 (m, 3H), 1.84-1.72 (m, 1H).






002


embedded image


466.20

1H NMR (400 MHz, DMSO-d6) δ 12.75 (s, 1H), 11.41 (s, 1H), 7.66 (dd, J = 5.6, 1.5 Hz, 1H), 7.40 (d, J = 8.0 Hz, 1H), 7.20 (dd, J = 7.1, 1.6 Hz, 1H), 7.08 (t, J = 7.6 Hz, 1H), 7.00 (dd, J = 13.8, 7.5 Hz, 1H), 6.42 (dd, J = 17.0, 9.9 Hz, 1H), 6.31 (dd, J = 17.1, 2.1 Hz, 1H), 5.83 (dd, J = 9.9, 2.1 Hz, 1H), 4.78 (d, J = 10.7 Hz, 1H), 4.23 (dd, J = 13.1. 10.2 Hz, 1H), 4.11 (d, J = 12.9 Hz, 1H), 3.71 (dt, J = 14.9, 7.8 Hz, 1H), 3.56 (s, 1H), 2.66 (d, J = 15.9 Hz, 4H), 2.07 (d, J = 16.1 Hz, 4H), 2.02-1.87 (m, 3H), 1.40 (s, 1H).






059


embedded image


463.30

1H NMR (400 MHz, DMSO-d6) δ 12.60 (s, 1H), 10.04 (d, J = 11.8 Hz, 1H), 7.42-7.23 (m, 2H), 7.06 (q, J = 7.4 Hz, 1H), 7.00-6.84 (m, 2H), 6.37 (dd, J = 17.0, 10.1 Hz, 1H), 6.18 (dd, J = 17.0, 2.1 Hz, 1H), 5.67 (dd, J = 10.1, 2.1 Hz, 1H), 4.72 (dt, J = 44.0, 10.8 Hz, 1H), 4.27-4.06 (m, 2H), 3.96 (s, 3H), 3.75 (dt, J = 14.9, 7.6 Hz, 1H), 3.52 (dt, J = 14.0, 5.6 Hz, 1H), 2.76-2.59 (m, 4H), 2.08 (d, J = 10.1 Hz, 4H), 1.96 (d, J = 10.9 Hz, 3H), 1.47-1.32 (m, 1H).






009


embedded image


466.30

1H NMR (400 MHz, DMSO-d6) δ 12.72 (s, 1H), 10.59 (s, 1H), 7.80 (d, J = 1.7 Hz, 1H), 7.70 (s, 1H), 7.41 (d, J = 8.0 Hz, 1H), 7.10 (t, J = 7.6 Hz, 1H), 7.00 (d, J = 7.8 Hz, 1H), 6.38 (dd, J = 17.0, 9.9 Hz, 1H), 6.27 (dd, J = 17.0, 2.2 Hz, 1H), 5.77 (dd, J = 9.9, 2.2 Hz, 1H), 4.86-4.58 (m, 1H), 4.23 (dd, J = 13.0, 10.2 Hz, 1H), 4.10 (d, J = 12.8 Hz, 1H), 3.87-3.68 (m, 1H), 3.57-3.48 (m, 1H), 2.80-2.59 (m, 4H), 2.12-1.89 (m, 6H), 1.37 (q, J = 12.8 Hz, 1H).






001


embedded image


466.30

1H NMR (400 MHz, DMSO-d6) δ 12.78 (d, J = 17.6 Hz, 1H), 11.59 (d, J = 10.9 Hz, 1H), 8.29 (dd, J = 7.8, 3.6 Hz, 1H), 7.99 (d, J = 3.5 Hz, 1H), 7.45 (d, J = 8.0 Hz, 1H), 7.14 (q, J = 7.7, 7.2 Hz, 1H), 7.02 (d, J = 7.6 Hz, 1H), 6.46- 6.21 (m, 2H), 5.85 (dd, J = 9.3, 2.4 Hz, 1H), 4.81 (s, 1H), 4.29-4.03 (m, 2H), 3.66 (dd, J = 13.9, 6.7 Hz, 1H), 3.56 (d, J = 5.7 Hz, 1H), 3.14-3.03 (m, 1H), 2.68 (d, J = 14.8 Hz, 4H), 2.08 (s, 3H), 2.04-1.87 (m, 4H), 1.38 (q, J = 11.7 Hz, 1H)






011


embedded image


486.20

1H NMR (400 MHz, DMSO-d6) δ 10.70 (s, 1H), 8.30 (s, 1H), 7.85 (d, J = 12.8 Hz, 1H), 7.72 (s, 1H), 7.59 (d, J = 44.6 Hz, 1H), 7.28-7.15 (m, 1H), 7.01 (s, 1H), 6.40 (dd, J = 17.0, 9.6 Hz, 1H), 6.27 (d, J = 17.2 Hz, 1H), 5.76 (d, J = 10.1 Hz, 1H), 5.35 (d, J = 30.4 Hz, 1H), 4.60 (t, J = 13.1 Hz, 1H), 4.32 (q, J = 13.7, 11.0 Hz, 1H), 4.16 (d, J = 13.5 Hz, 1H), 3.96 (d, J = 14.3 Hz, 1H), 2.07 (d, J = 10.8 Hz, 3H), 1.98 (s, 4H), 1.31 (dd, J = 40.7, 22.6 Hz, 2H).






023


embedded image


459.15

1H NMR (400 MHz, DMSO-d6) δ 12.86 (s, 1H), 10.56 (s, 1H), 7.91-7.68 (m, 2H), 7.58-7.47 (m, 1H), 7.32-7.18 (m, 2H), 6.45-6.24 (m, 2H), 5.77 (dd, J = 9.8, 2.3 Hz, 1H), 4.49 (s, 1H), 4.32 (d, J = 7.1 Hz, 2H), 3.37 (s, 1H), 1.91- 1.78 (m, 3H), 1.63 (d, J = 12.8 Hz, 2H), 1.27-1.14 (m, 2H), 1.11-1.00 (m, 2H).






014


embedded image


444.20

1H NMR (400 MHz, DMSO-d6) δ 10.64 (s, 1H), 7.85 (d, J = 1.6 Hz, 1H), 7.75 (d, J = 1.6 Hz, 1H), 7.54 (dd, J = 7.8, 1.2 Hz, 1H), 7.31 (dd, J = 8.1, 1.2 Hz, 1H), 7.24 (t, J = 8.0 Hz, 1H), 6.38 (dd, J = 17.0, 9.9 Hz, 1H), 6.28 (dd, J = 17.0, 2.2 Hz, 1H), 5.78 (dd, J = 9.9, 2.2 Hz, 1H), 4.27 (dd, J = 13.8, 8.5 Hz, 1H), 3.73 (dd, J = 13.9, 3.0 Hz, 1H), 3.36 (s, 1H), 3.28-3.22 (m, 1H), 2.55 (d, J = 10.5 Hz, 1H), 2.24-2.13 (m, 1H), 2.07-1.91 (m, 4H), 1.73 (d, J = 10.6 Hz, 1H).






020


embedded image


445.15

1H NMR (400 MHz, DMSO) δ 12.91 (s, 1H), 10.58 (s, 1H), 7.76 (s, 2H), 7.52 (d, J = 7.7 Hz, 1H), 7.27 (d, J = 8.3 Hz, 1H), 7.19 (t, J = 7.9 Hz, 1H), 6.38 (dd, J = 17.0, 9.9 Hz, 1H), 6.27 (dd, J = 17.0, 2.2 Hz, 1H), 5.77 (dd, J = 9.9, 2.3 Hz, 1H), 5.36-5.23 (m, 1H), 4.71 (d, J = 4.3 Hz, 1H), 3.70 (d, J = 4.4 Hz, 1H), 2.97 (q, J = 12.5 Hz, 2H), 2.06-1.99 (m, 2H), 1.82 (d, J = 12.2 Hz, 2H), 1.35 (g, J = 12.1 Hz, 2H).






015


embedded image


458.20

1H NMR (400 MHz, DMSO) δ 12.89 (s, 1H), 10.58 (s, 1H), 7.80 (d, J = 1.6 Hz, 1H), 7.74 (d, J = 1.6 Hz, 1H), 7.51 (dd, J = 7.9, 1.2 Hz, 1H), 7.27 (dd, J = 8.1, 1.2 Hz, 1H), 7.19 (t, J = 7.9 Hz, 1H), 6.38 (dd, J = 17.0, 9.9 Hz, 1H) 6.27 (dd, J = 17.0, 2.2 Hz, 1H), 5.77 (dd, J = 9.9, 2.2 Hz, 1H), 5.62 (q, J = 10.6, 9.5 Hz. 1H), 3.91-3.79 (m, 1H), 2.85 (s, 1H), 2.82 (s, 1H), 2.59 (s, 1H), 2.37 (s, 3H), 1.97 (d, J = 7.2 Hz, 1H), 1.92- 1.75 (m, 4H), 1.73 (s, 1H).






010


embedded image


452.25

1H NMR (400 MHz, DMSO-d6) δ 12.71 (d, J = 13.7 Hz, 1H), 10.57 (d, J = 3.8 Hz, 1H), 7.83-7.47 (m, 4H), 7.23 (dddd, J = 9.3, 7.4, 5.9, 4.0 Hz, 2H), 6.44-6.23 (m, 2H), 5.77 (dd, J = 9.9, 2.2 Hz, 1H), 4.70 (d, J = 103.2 Hz, 1H), 4.30-3.96 (m, 2H), 3.81 (dd, J = 14.4, 3.7 Hz, 1H), 3.64 (ddt, J = 43.1, 13.2, 6.5 Hz, 1H), 3.20 (dt, J = 13.6, 5.7 Hz, 1H), 2.54 (s, 1H), 2.11 (d, J = 18.8 Hz, 3H), 1.95 (td, J = 28.2, 27.0, 16.7 Hz, 4H), 1.45 (dt, J = 25.9, 12.5 Hz, 1H).






052


embedded image


452.25

1H NMR (400 MHz, DMSO-d6) δ 12.70 (s, 1H), 10.53 (s, 1H), 7.72 (d, J = 1.6 Hz, 1H), 7.65 (d, J = 1.5 Hz, 1H), 7.41 (d, J = 7.8 Hz, 1H), 7.10 (t, J = 7.7 Hz, 1H), 7.01 (d, J = 7.5 Hz, 1H), 6.44- 6.20 (m, 2H), 5.77 (dd, J = 9.8, 2.3 Hz, 1H), 4.93 (s, 1H), 4.59 (d, J = 13.0 Hz, 1H), 4.08-3.95 (m, 1H), 3.26-3.19 (m, 1H), 3.14 (t, J = 12.9 Hz, 1H), 2.87 (d, J = 12.2 Hz, 1H), 2.73 (s, 3H), 2.13 (s, 3H), 1.88 (dd, J = 25.3, 11.6 Hz, 2H).






062


embedded image


520.25

1H NMR (400 MHz, DMSO) δ 13.18 (s, 1H), 10.59 (s, 1H), 7.97-7.70 (m, 3H), 7.64 (d, J = 18.9 Hz, 1H), 7.42 (d, J = 11.5 Hz, 1H), 6.51-6.16 (m, 2H), 5.77 (d, J = 12.0 Hz, 1H), 4.33 (s, 2H), 4.21 (s, 1H), 3.74 (d, J = 47.1 Hz, 1H), 3.59 (s, 1H), 3.39 (s, 1H), 2.85-2.60 (m, 1H), 2.01 (d, J = 31.3 Hz, 6H), 1.35- 1.20 (m, 1H).






063


embedded image


480.3

1H NMR (400 MHz, DMSO) δ 12.68 (d, J = 17.4 Hz, 1H), 10.56 (s, 1H), 7.77 (dd, J = 5.6, 1.6 Hz, 1H), 7.71 (d, J = 1.6 Hz, 1H), 7.41 (dd, J = 7.8, 3.9 Hz, 1H), 7.11 (td, J = 7.7, 5.6 Hz, 1H), 7.02 (t, J = 8.3 Hz, 1H), 6.38 (dd, J = 17.0, 9.9 Hz, 1H), 6.27 (dd, J = 16.9, 2.2 Hz, 1H), 5.81-5.75 (m, 1H), 4.55-4.14 (m, 2H), 3.91 (dd, J = 13.6, 4.5 Hz, 1H), 3.62-3.50 (m, 1H), 3.25-3.14 (m, 1H), 2.99 (td, J = 14.0, 8.4 Hz, 1H), 2.86 (d, J = 3.6 Hz, 3H), 2.21 (d, J = 10.6 Hz, 1H), 2.04-1.96 (m, 1H), 1.93 (d, J = 16.7 Hz, 2H), 1.78 (dd, J = 6.6, 4.4 Hz, 2H), 1.73 (s, 1H), 1.64 (d, J = 5.3 Hz, 1H), 1.53 (s, 1H), 1.42-1.34 (m, 1H).






064


embedded image


549.35

1H NMR (400 MHz, DMSO) δ 12.59 (s, 1H), 9.42 (s, 1H), 7.94 (d, J = 9.0 Hz, 1H), 7.38 (dd, J = 8.0, 1.2 Hz, 1H), 7.07 (t, J = 7.7 Hz, 1H), 6.97 (d, J = 7.5 Hz, 1H), 6.62 (dd, J = 17.0, 10.2 Hz, 1H), 6.23 (dt, J = 16.9, 1.9 Hz, 1H), 5.72 (dd, J = 10.2, 2.1 Hz, 1H), 4.80 (s, 1H), 4.27 (dd, J = 13.1, 10.4 Hz, 1H), 4.12-4.06 (m, 1H), 3.80-3.74 (m, 1H), 3.52 (td, J = 7.0, 4.6 Hz, 1H), 2.96 (t, J = 5.3 Hz, 4H), 2.73 (d, J = 11.3 Hz, 1H), 2.64 (s, 3H), 2.08 (s, 4H), 1.97 (td, J = 12.6, 6.2 Hz, 3H), 1.69 (t, J = 5.8 Hz, 4H), 1.54 (q, J = 4.6 Hz, 2H), 1.35 (d, J = 11.4 Hz, 1H)






065


embedded image


491.30

1H NMR (400 MHz, DMSO) δ 12.85 (s, 1H), 10.92 (s, 1H), 8.18 (s, 1H), 7.46 (s, 1H), 7.09 (dd, J = 36.0, 7.6 Hz, 2H), 6.62 (d, J = 17.0 Hz, 1H), 6.37 (d, J = 18.7 Hz, 1H), 5.89 (s, 1H), 4.84 (s, 1H), 4.13 (d, J = 7.3 Hz, 2H), 3.72 (d, J = 15.1 Hz, 1H), 3.54 (d, J = 7.2 Hz, 1H), 2.68 (d, J = 12.7 Hz, 4H), 2.09 (s, 3H), 1.99 (t, J = 14.1 Hz, 4H), 1.39 (s, 1H).






066


embedded image


562.30

1H NMR (400 MHz, DMSO) δ 12.66 (s, 1H), 8.31 (s, 1H), 7.91 (s, 1H), 7.40 (d, J = 8.0 Hz, 1H), 7.09 (t, J = 7.6 Hz 1H), 6.99 (d, J = 7.5 Hz, 1H), 6.37- 6.21 (m, 2H), 5.85-5.65 (m, 1H), 4.81 (s, 1H), 4.28 (dd, J = 13.0, 10.3 Hz, 1H), 4.20-4.05 (m, 1H), 4.03-3.88 (m, 1H), 3.75 (dt, J = 14.7, 7.7 Hz, 1H), 3.60 (dd, J = 8.7, 3.2 Hz, 2H), 2.09 (s, 3H), 1.97 (d, J = 11.6 Hz, 3H), 1.37 (dd, J = 12.4, 6.9 Hz, 1H).






067


embedded image


531.30

1H NMR (400 MHz, DMSO) δ 12.68 (s, 1H), 11.29 (s, 1H), 9.75 (s, 1H), 7.92 (s, 1H), 7.41 (d, J = 8.0 Hz, 1H), 7.10 (s, 1H), 7.00 (d, J = 7.5 Hz, 1H), 6.93 (s, 1H), 6.55 (d, J = 26.3 Hz, 1H), 6.40 (s, 1H), 6.22 (d, J = 17.1 Hz, 2H), 5.83- 5.65 (m, 1H), 4.81 (s, 1H), 4.28 (t, J = 10.1 Hz, 1H), 4.12 (d, J = 11.6 Hz, 1H), 3.75 (s, 1H), 3.56 (s, 1H), 2.65 (s, 3H), 2.09 (s, 3H), 1.45 (s, 1H).






068


embedded image


523.3

1H NMR (400 MHz, DMSO) δ 12.70 (s, 1H), 10.50 (s, 1H), 8.21 (s, 1H), 7.78 (d, J = 1.6 Hz, 1H), 7.68 (d, J = 1.7 Hz, 1H), 7.41 (dd, J = 8.0, 1.2 Hz, 1H), 7.10 (t, J = 7.7 Hz, 1H), 7.00 (d, J = 7.6 Hz, 1H), 6.75 (d, J = 15.4 Hz, 1H), 6.20 (dt, J = 15.3, 1.6 Hz, 1H), 4.78 (d, J = 12.5 Hz, 1H), 4.21 (d, J = 10.2 Hz, 1H), 4.11 (s, 1H), 3.96 (d, J = 13.6 Hz, 1H), 3.82 (d, J = 8.4 Hz, 1H), 3.08 (dd, J = 6.0, 1.6 Hz, 3H), 2.68 (d, J = 3.9 Hz, 1H), 2.64 (s, 3H), 2.19 (s, 6H), 2.08 (d, J = 7.1 Hz, 3H), 2.03-1.91 (m, 3H), 1.36 (d, J = 11.9 Hz, 1H).






069


embedded image


478.25

1H NMR (400 MHz, DMSO) δ 12.78 (s, 1H), 10.59 (s, 1H), 7.80 (d, J = 1.6 Hz 1H), 7.70 (d, J = 1.7 Hz, 1H), 7.41 (d, J = 7.8 Hz, 1H), 7.09 (t, J = 7.7 Hz, 1H), 8.99 (d, J = 7.6 Hz, 1H), 6.45-6.19 (m, 2H), 5.77 (dd, J = 9.9, 2.3 Hz, 1H), 4.58 (t, J = 7.8 Hz, 2H), 4.36 (s, 1H), 2.82 (t, J = 12.3 Hz, 1H), 2.67 (d, J = 12.5 Hz, 1H), 2.67 (s, 3H), 2.39 (s, 1H), 2.16 (d, J = 6.3 Hz, 2H), 2.10 (s, 3H), 2.07-1.95 (m, 2H).






070


embedded image


538.2
δ: 7.85-8.05 (m, 1H), 7.60 (br d, 1H), 7.36 (br d, 1H), 6.94-7.09 (m, 1H), 6.66- 6.93 (m, 1H), 6.31-6.47 (m, 1H), 6.13- 6.30 (m, 1H), 5.71 (br d, 1H), 5.50-5.67 (m, 1H), 4.56-4.75 (m, 2H), 3.90 (br s, 1H), 2.89-3.21 (m, 8H), 2.63 (br s, 3H), 2.59-2.59 (m, 1H), 1.73 (br d, 8H), 1.24- 1.55 (m, 4H)





071


embedded image


523.2
δ 9.57 (s, 1H), 8.05 (br s, 1H), 7.94 (s, 1H), 7.36 (d, J = 7.9 Hz, 1H), 7.09- 7.01 (m, 1H), 6.99-6.90 (m, 1H), 6.59 (dd, J = 10.1, 17.1 Hz, 1H), 6.23 (dd, J = 1.9, 17.0 Hz, 1H), 5.77-5.65 (m, 1H), 4.69-4.57 (m, 2H), 3.66 (br d, J = 4.2 Hz, 1H), 3.53 (s, 2H), 3.33 (br s, 2H), 3.28 (s, 1H), 3.22-3.10 (m, 2H), 3.03-2.86 (m, 2H), 2.71-2.60 (m, 4H), 2.35-2.26 (m, 1H), 1.99 (br d, J = 10.1 Hz, 2H), 1.83-1.70 (m, 2H), 1.42- 1.25 (m, 2H)





072


embedded image


546.2
δ: 9.76 (br s, 1H), 8.82 (br d, 1H), 7.58- 7.73 (m, 1H), 7.42 (br d, 1H), 7.30 (br d, 1H), 6.95-7.05 (m, 1H), 6.91 (br d, 1H), 6.79 (br d, 1H), 6.41 (br m, 1H), 6.23 (br d, 1H), 5.73 (br d, 1H), 4,66 (br s, 1H), 4.42-4.58 (m, 2H), 3.65 (s, 2H), 2.56-2.70 (m, 3H), 2.06-2.35 (m, 3H), 1.81-2.04 (m, 3H), 1.61-1.79 (m, 3H), 1.55 (br d, 3H), 1.32 (br s, 2H), 1.21 (br s, 2H)





073


embedded image


450.1

1H NMR (400 MHz, CD3OD) δ 8.20 (s, 1H), 7.35 (d, J = 7.0 Hz, 1H), 7.20-7.00 (m, 2H), 6.58-6.41 (m, 1H), 5.90-5.83 (m, 1H), 2.75 (s, 3H), 2.25-2.14 (m, 2H), 1.95 (br d, J = 10.5 Hz, 2H), 0.89 (br d, J = 7.2 Hz, 7H)






074


embedded image


488.1

1H NMR (400 MHz, ACETONITRILE- d3) δ 12.66-12.20 (m, 1H), 9.79-9.61 (m, 1H), 7.72 (s, 1H), 7.39-7.23 (m, 2H), 7.15-7.08 (m, 1H), 7.05-6.98 (m, 1H), 4.95-4.83 (m, 1H), 4.36- 4.11 (m, 3H), 3.75-3.64 (m, 1H), 3.60- 3.51 (m, 1H), 2.83-2.59 (m, 4H), 2.09 (s, 3H), 2.07-1.96 (m, 4H), 1.54- 1.38 (m, 1H)






075


embedded image


506.0

1H NMR (ACETONITRILE-d3) δ 12.33- 12.65 (m, 1H), 10.04 (s, 1H), 7.81 (d, 1H), 7.39-7.46 (m, 1H), 7.39-7.46 (m, 1H), 7.35 (d, 1H), 7.15 (m, 1H), 6.99- 7.10 (m, 1H), 6.59-6.82 (m, 1H), 4.93 (br m, 1H), 4.17-4.39 (m, 2H), 3.69- 3.84 (m, 1H), 3.45-3.66 (m, 1H), 2.76- 2.88 (m, 1H), 2.68-2.75 (m, 3H), 2.12 (s, 3H), 2.02-2.09 (m, 4H), 1.41-1.58 (m, 2H)






076


embedded image


484.0

1H NMR (400 MHz, ACETONITRILE- d3) δ ppm 0.81-1.07 (m, 2 H) 1.26- 1.59 (m, 2 H) 2.01-2.05 (m, 2 H) 2.09 (s, 3 H) 2.68-2.72 (m, 3 H) 3.50-3.62 (m, 1 H) 3.65-3.77 (m, 1 H) 4.16- 4.35 (m, 2 H) 4.82-4.94 (m, 1 H) 5.33 (dd, J = 15.34, 3.61 Hz, 1 H) 5.65-5.84 (m, 1 H) 6.98-7.05 (m, 1 H) 7.08- 7.14 (m, 1 H) 7.31 (d, J = 8.31 Hz, 1 H) 7.34-7.40 (m, 1 H) 7.76 (s, 1 H) 9.84- 10.01 (m, 1 H)






077


embedded image


564.3

1H NMR (400 MHz, CHLOROFORM-d) δ ppm 1.90-2.02 (m, 1 H) 2.05-2.15 (m, 4 H) 2.19 (s, 3 H) 2.23 (s, 1 H) 2.30- 2.34 (m, 3 H) 2.52 (br s, 7 H) 2.61- 2.77 (m, 2 H) 3.51-3.62 (m, 2 H) 3.62- 3.79 (m, 2 H) 4.07-4.32 (m, 2 H) 4.47-4.72 (m, 1 H) 5.74 (br d, J = 10.27 Hz, 1 H) 6.21-6.36 (m, 1 H) 6.38- 6.48 (m, 1 H) 7.12-7.25 (m, 2 H) 7.32 (br s, 1 H) 7.65 (br s, 1 H) 7.91 (br s, 1 H) 8.21-8.55 (m, 1 H) 11.92-12.31 (m, 1 H)










Example 2: HTRF-Based EGFR Biochemical Assays

EGFR biochemical activity measurements were carried out using the homogeneous time-resolved fluorescence (HTRF) assay (Cisbio). Inhibitors and DMSO normalizations were first dispensed to empty black low-volume 384-well plates (Corning) with D300 digital liquid dispenser (HP). All reactions were carried out at room temperature and solutions were added to plates with a Multidrop Combi Reagent Dispenser (ThermoFisher). The reaction mixture (10 μL final volume) contained 1 μM tyrosine kinase peptide-biotin substrate and mutant EGFR in a reaction buffer (50 mM HEPES pH 7.0, 5 mM MgCl2, 1 mM MnCl2, 0.01% BSA, 2 mM TCEP, 0.1 mM NaVO4). Enzyme concentrations were adjusted to accommodate varying kinase activities (L858R 0.1 nM, L858R/T790M 0.02 nM). Enzyme reaction solution (2× concentrations, 5 μL) was added to 384-well plates containing compounds and incubated for 30 mins. Enzyme reactions were initiated with the addition of 5 μL of ATP to a final concentration of 100 μM and reacted for 20 mins. Reactions were quenched with the addition of 10 μL of phospho-tyrosine antibody-Europium(III) cryptate (1-to-180 volume ratio) and Streptavidin-XL665 (46.7 nM) in EDTA-containing detection buffer, then incubated at room temperature for 1 hour, and read with a PHERAstar plate reader (excitation=337 nm, emission=620 nm and 665 nm). IC50 values were determined by inhibition curves (11-point curves from 1.0 μM to 0.130 nM or 23-point curves from 1.0 μM to 0.130 μM) in triplicate with non-linear least squares fit in GraphPad Prism 7.0d.


Example 3: Ba/F3 Cell Proliferation Models

The EGFR mutant L858R Ba/F3 cells have been previously described (Zhou, W., et al. Nature 482, 2009, 1070-1074). The EGFR C797S and C775S mutations were introduced via site directed mutagenesis using the Quick Change Site-Directed Mutagenesis kit into a vector containing EGFR L858R mutation (Stratagene; La Jolla, CA) according to the manufacturer's instructions. All constructs were confirmed by DNA sequencing. The constructs were then shuttled into the retroviral vector JP1540 by either using the Cre-recombination system (Agilent Technologies, Santa Clara, CA) or the in-fusion HD Cloning kit (Takers Bio USA, Inc.; Mountain view, CA). Ba/F3 cells were then infected with retrovirus per standard protocols, as described previously (Zhou, et al, Nature 2009). Stable clones were obtained by selection in puromycin (2 μg/ml). All BaF/3 mutant cells were maintained in RPM 1640 (Cellgro; Mediatech Inc., Herndon, CA) supplemented with 10% FBS, 100 units/mL penicillin, 100 units/mL streptomycin.


Growth and inhibition of growth was assessed by the Cell Titer Glo assay (Promega, Madison, WI) and was performed according to the manufacturer's instructions. The Cell Titer Glo assay is a luminescence-based method used to determine the number of viable cells based on quantitation of the ATP present, which is directly proportional to the amount of metabolically active cells present. Ba/F3 cells of different EGFR genotypes were exposed to compounds for 72 hours and the number of cells used per experiment was determined empirically as has been previously established (Zhou, et al., Nature 2009). All experimental points were set up in triplicates in 384-well plates. The luminescent signal was detected using a spectrometer and the data was graphically displayed using GraphPad Prism version 5.0 for Windows, (GraphPad Software; www.graphpad.com). The curves were fitted using a non-linear regression model with a sigmoidal dose response. The results of this assay for the compounds disclosed herein are shown in Table 5.













TABLE 5







Compound No.
002
009









L BaF3 (uM)
0.11
0.062



L/C797S (uM)
0.43
0.173










The disclosed subject matter is not to be limited in scope by the specific embodiments and examples described herein. Indeed, various modifications of the disclosure in addition to those described will become apparent to those skilled in the art from the foregoing description and accompanying figures. Such modifications are intended to fall within the scope of the appended claims.


All references (e.g., publications or patents or patent applications) cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each individual reference (e.g., publication or patent or patent application) was specifically and individually indicated to be incorporated by reference in its entirety for all purposes. Other embodiments are within the following claims.

Claims
  • 1. A compound of Formula I:
  • 2. The compound of claim 1, wherein: A is selected from the group consisting of phenyl, 5-6 membered heteroaryl, C3-C8 cycloalkyl, and 3-8 membered heterocycloalkyl;R1 is selected from the group consisting of H, C1-C6 alkyl, and 5-6 membered heteroaryl;R3 is selected from the group consisting of C1-C3 alkyl, C2-C4 alkenyl, SO2(C1-C3 alkyl), phenyl, 5-6 membered heteroaryl, C5-C6 cycloalkyl, and 4-8 membered heterocycloalkyl all of which are optionally substituted with one or two R5;R4 is selected from the group consisting of H, halo, and C1-C3 alkyl;R5 is independently selected from the group consisting of C1-C4 alkyl, OH, NH2, N(C1-C3 alkyl)2, C(O)—C1-C3 alkyl, C(O)NH2, C(O)NH—C1-C4 alkyl, C1-C4 alkyl-OH, P(O)(C1-C3 alkyl)2, phenyl, 5-6 membered heteroaryl, 4-8 membered heterocycloalkyl, C5-C6 cycloalkyl; wherein phenyl, heteroaryl, heterocycloalkyl, and cycloalkyl are each optionally substituted with one or two R6;R6 is independently selected from the group consisting of halo, OH, C(O)—C1-C3 alkyl, NH2, N(C1-C3 alkyl)2, and P(O)(C1-C3 alkyl)2;R2 is selected from the group consisting of
  • 3. The compound of claim 1, wherein: A is selected from the group consisting of phenyl, piperidine, thiophene, pyrrolidine, isoxazole, pyrrole, pyridine, isothiazole, pyrazole, imidazole, thiazole, and oxazole;R1 is selected from the group consisting of H, C1-C3 alkyl, and 5-6 membered heteroaryl;R3 is selected from the group consisting of C1-C4 alkyl, C2-C4 alkenyl, SO2(C1-C3 alkyl), azetidine, azepane, diazepane, oxazepane, oxepane, piperidine, pyrrolidine, cycloheptane, cyclohexane, phenyl, pyridizine, pyridine, and pyrazine, all of which are optionally substituted with R5;R4 is selected from the group consisting of H, halo, and C1-C3 alkyl;R5 is independently selected from the group consisting of C1-C4 alkyl, OH, NH2, N(C1-C3 alkyl)2, C(O)—C1-C3 alkyl, C(O)NH2, C(O)NH—C1-C4 alkyl, C1-C4 alkyl-OH, P(O)(C1-C3 alkyl)2, phenyl, 5-6 membered heteroaryl, 4-8 membered heterocycloalkyl, C5-C6 cycloalkyl; wherein phenyl, heteroaryl, heterocycloalkyl, and cycloalkyl are each optionally substituted with one or two R6;R6 is independently selected from the group consisting of halo, OH, C(O)—C1-C3 alkyl, NH2, N(C1-C3 alkyl)2, and P(O)(C1-C3 alkyl)2; andR2 is selected from the group consisting of
  • 4. The compound of claim 1, wherein A is selected from the group consisting of phenyl, 5-6 membered heteroaryl, C4-C7 cycloalkyl, and 4-10 membered heterocycloalkyl.
  • 5. The compound of claim 1, wherein A is selected from the group consisting of phenyl, piperidine, thiophene, pyrrolidine, isoxazole, pyrrole, pyridine, isothiazole, pyrazole, imidazole, thiazole, and oxazole.
  • 6-10. (canceled)
  • 11. The compound of claim 1, wherein R1 is selected from the group consisting of H, 5-6 membered heteroaryl, and C1-C3 alkyl.
  • 12. The compound of claim 1, wherein R3 is selected from the group consisting of C1-C3 alkyl, C2-C4 alkenyl, SO2(C1-C6 alkyl), phenyl, 5-6 membered heteroaryl, C4-C8 cycloalkyl, and 4-8 membered heterocycloalkyl all of which are optionally substituted with one or two R5.
  • 13. The compound of claim 1, wherein R3 is selected from the group consisting of C1-C4 alkyl, C2-C4 alkenyl, SO2(C1-C3 alkyl), azetidine, azepane, diazepane, oxazepane, oxepane, piperidine, pyrrolidine, cycloheptane, cyclohexane, phenyl, pyridizine, pyridine, and pyrazine, all of which are optionally substituted with R5.
  • 14. The compound of claim 1, wherein R4 is selected from the group consisting of H, halo, and C1-C3 alkyl.
  • 15. The compound of claim 1, wherein each R5 is independently selected from the group consisting of C1-C4 alkyl, OH, NH2, N(C1-C6 alkyl)2, C(O)—C1-C3 alkyl, C(O)NH2, C(O)NH—C1-C4 alkyl, C1-C4 alkyl-OH, P(O)(C1-C3 alkyl)2, phenyl, 5-6 membered heteroaryl, 4-8 membered heterocycloalkyl, C4-C8 cycloalkyl; wherein phenyl, heteroaryl, heterocycloalkyl, and cycloalkyl are each optionally substituted with one or two R6; alternatively, R5 is
  • 16. (canceled)
  • 17. The compound of claim 1, wherein each R6 is independently selected from the group consisting of halo, OH, C(O)—C1-C3 alkyl, NH2, N(C1-C3 alkyl)2, and P(O)(C1-C3 alkyl)2.
  • 18. (canceled)
  • 19. The compound of claim 1, wherein R2 is selected from the group consisting of
  • 20. The compound of claim 1, wherein the compound of Formula I is a compound of Formula II:
  • 21. (canceled)
  • 22. The compound of claim 20, wherein R1 is H or 5-6 membered heteroaryl;R3 is selected from the group consisting of C1-C4 alkyl, C2-C4 alkenyl, SO2(C1-C3 alkyl), azetidine, azepane, diazepane, oxazepane, oxepane, piperidine, pyrrolidine, cycloheptane, cyclohexane, phenyl, pyridizine, pyridine, and pyrazine, all of which are optionally substituted with R5;R4 is selected from the group consisting of H, halo, and C1-C3 alkyl;R5 is independently selected from the group consisting of C1-C4 alkyl, OH, NH2, N(C1-C3 alkyl)2, C(O)—C1-C3 alkyl, C(O)NH2, C(O)NH—C1-C4 alkyl, C1-C4 alkyl-OH, P(O)(C1-C3 alkyl)2, phenyl, 5-6 membered heteroaryl, 4-8 membered heterocycloalkyl, C5-C6 cycloalkyl; wherein phenyl, heteroaryl, heterocycloalkyl, and cycloalkyl are each optionally substituted with one or two R6;R6 is independently selected from the group consisting of halo, OH, C(O)—C1-C3 alkyl, NH2, N(C1-C3 alkyl)2, and P(O)(C1-C3 alkyl)2; andR2 is selected from the group consisting of
  • 23. (canceled)
  • 24. The compound of claim 1, wherein the compound of Formula I is selected from the group consisting of
  • 25-26. (canceled)
  • 27. A pharmaceutical composition comprising a compound of claim 1, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • 28. A method of inhibiting the activity of EGFR in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of claim 1.
  • 29. A method of treating cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of claim 1.
  • 30. The method of claim 29, wherein the cancer is selected from the group consisting of lung cancer, colon cancer, breast cancer, endometrial cancer, thyroid cancer, glioma, squamous cell carcinoma, and prostate cancer.
  • 31. The method according to claim 29, wherein the cancer is non-small cell lung cancer (NSCLC).
RELATED APPLICATIONS

This application claims priority to U.S. Provisional Application No. 63/088,764 filed on Oct. 7, 2020 and U.S. Provisional Application No. 63/246,054 filed on Sep. 20, 2021, the entire contents of which are hereby incorporated by reference in their entirety.

Provisional Applications (2)
Number Date Country
63246054 Sep 2021 US
63088764 Oct 2020 US
Continuations (1)
Number Date Country
Parent PCT/US2021/053809 Oct 2021 US
Child 18191574 US