2,4-disubstituted pyrimidine compounds useful as kinase inhibtors

Information

  • Patent Grant
  • 9375431
  • Patent Number
    9,375,431
  • Date Filed
    Tuesday, March 3, 2015
    9 years ago
  • Date Issued
    Tuesday, June 28, 2016
    8 years ago
Abstract
The present invention provides 2,4-disubstituted pyrimidine compounds useful as kinase inhibitors, pharmaceutically acceptable compositions thereof, and methods of using the same.
Description
TECHNICAL FIELD OF THE INVENTION

The present invention relates to compounds useful as mutant-selective epidermal growth factor receptor (EGFR) kinase inhibitors. The invention also provides pharmaceutically acceptable compositions comprising compounds of the present invention and methods of using said compositions in the treatment of various disorders.


BACKGROUND OF THE INVENTION

Protein tyrosine kinases are a class of enzymes that catalyze the transfer of a phosphate group from ATP or GTP to a tyrosine residue located on a protein substrate. Receptor tyrosine kinases act to transmit signals from the outside of a cell to the inside by activating secondary messaging effectors via a phosphorylation event. A variety of cellular processes are promoted by these signals, including proliferation, carbohydrate utilization, protein synthesis, angiogenesis, cell growth, and cell survival.


There is strong precedent for involvement of the EGFR in human cancer because over 60% of all solid tumors overexpress at least one of these proteins or their ligands. Overexpression of EGFR is commonly found in breast, lung, head and neck, bladder tumors.


Activating mutations in the tyrosine kinase domain of EGFR have been identified in patients with non-small cell lung cancer (Lin, N. U.; Winer, E. P., Breast Cancer Res 6: 204-210, 2004). The reversible inhibitors Tarceva (erlotinib) and Iressa (gefitinib) currently are first-line therapy for non-small cell lung cancer patients with activating mutations. The most common activating mutations are L858R and delE746-A750.


Additionally, in the majority of patients that relapse, acquired drug resistance, such as by mutation of gatekeeper residue T790M, has been detected in at least half of such clinically resistant patients. Moreover, T790M may also be pre-existing, there may be an independent, oncogenic role for the T790M mutation. For example, there are patients with the L858R/T790M mutation who never received gefitinib treatment. In addition, germline EGFR T790M mutations are linked with certain familial lung cancers.


Current drugs in development, including second generation covalent inhibitors, such as BIBW2992, HKI-272 and PF-0299804, are effective against the T790M resistance mutation but exhibit dose-limiting toxicities due to concurrent inhibition of WT EGFR. Accordingly, there remains a need to find mutant-selective EGFR kinase inhibitors useful as therapeutic agents.


SUMMARY OF THE INVENTION

It has now been found that compounds of this invention, and pharmaceutically acceptable compositions thereof, are effective as mutant-selective EGFR kinase inhibitors. Such compounds have general formula I:




embedded image



or a pharmaceutically acceptable salt thereof, wherein each of n, m, W, G, R1 R2, and R5 is as defined and described herein.


Compounds of the present invention, and pharmaceutically acceptable compositions thereof, are useful for treating cancers associated with one or more EGFR mutations. Such diseases, disorders, or conditions include those described herein.


Compounds provided by this invention are also useful for the study of kinases in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated by such kinases; and the comparative evaluation of new kinase inhibitors.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 depicts MS analysis confirming covalent modification of EGFR T790M/L858R by compound I-4.





DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS

1. General Description of Compounds of the Invention


In certain embodiments, the present invention provides a compound of formula I:




embedded image



or a pharmaceutically acceptable salt thereof, wherein:

  • n is 0, 1, or 2;
  • m is 0, 1, or 2, wherein m and n are not simultaneously 0;
  • W is —O— or —NH—;
  • R1 is —OR;
  • each R is independently C1-4 alkyl or C1-4 fluoroalkyl;
  • R2 is —CF3, Cl, or Br;
  • G is —O—, —NR3—, —S(O)2—, or —CH(OR4)—;
  • R3 is —C(O)—R, —C(O)OR, —C(O)NHR, —SO2—R, —SO2NH2, —C(O)—C1-4 alkylene-OH or —SO2—C1-4 alkylene-OH;
  • R4 is hydrogen, C1-4 alkyl, or C1-4 fluoroalkyl; and
  • R5 is hydrogen or —C(O)OR.


In certain embodiments, the present invention provides a compound of formula I:




embedded image



or a pharmaceutically acceptable salt thereof, wherein:

  • n is 0, 1, or 2;
  • m is 0, 1, or 2, wherein m and n are not simultaneously 0;
  • W is —O— or —NH—;
  • R1 is —OR;
  • each R is independently C1-4 alkyl or C1-4 fluoroalkyl;
  • R2 is —CF3, Cl, or Br;
  • G is —O—, —NR3—, or —CH(OR4)—;
  • R3 is —C(O)—R, —C(O)OR, —C(O)NHR, —SO2—R, —SO2NH2, —C(O)—C1-4 alkylene-OH or —SO2—C1-4 alkylene-OH; and
  • R4 is hydrogen, C1-4 alkyl, or C1-4 fluoroalkyl.


As used herein, the term “C1-4 alkylene” refers to a bivalent, straight or branched saturated hydrocarbon chain having 1-4 carbon atoms.


In some embodiments, n is 0 and G is —CH(OR4)—.


In some embodiments, m is 0 and G is —CH(OR4)—.


In some embodiments, the present invention provides a compound of formula I or I-a, wherein W is —NH—.


In certain embodiments, the present invention provides a compound of formula I or I-a, wherein W is —NH— and R2 is —CF3.


In certain embodiments, the present invention provides a compound of formula I or I-a, wherein W is —O— and R2 is —Cl.


In certain embodiments, the present invention provides a compound of formula I-a wherein G is —O— thereby forming a compound of formula II:




embedded image



or a pharmaceutically acceptable salt thereof, wherein W and R2 are as defined above for formula I and I-a.


In some embodiments, the present invention provides a compound of formula II, wherein W is —NH—.


In certain embodiments, the present invention provides a compound of formula II, wherein W is —NH— and R2 is —CF3.


In some embodiments, the present invention provides a compound of formula I, I-a, or II wherein at least one; or both of the following characteristics apply:

    • (a) W is —O— or —NH—; and
    • (b) R2 is —CF3 or Cl.


In some embodiments, the present invention provides a compound of formula I, I-a, or II wherein at least one; or both of the following characteristics apply:

    • (a) W is —O—; and
    • (b) R2 is —CF3 or Cl.


In some embodiments, the present invention provides a compound of formula I, I-a, or II wherein at least one; or both of the following characteristics apply:

    • (a) W is —NH—; and
    • (b) R2 is —CF3 or Cl.


In certain embodiments, the present invention provides a compound of formula I wherein G is —NR3— thereby forming a compound of formula III:




embedded image



or a pharmaceutically acceptable salt thereof, wherein W, R2, and R3 are as defined above for formula I.


In certain embodiments, the present invention provides a compound of formula I-a wherein G is —NR3— thereby forming a compound of formula III-a:




embedded image



or a pharmaceutically acceptable salt thereof, wherein W, R2, and R3 are as defined above for formula I.


As defined above, the R3 group of formula III or III-a is —C(O)—C1-4 alkyl, —SO2—C1-4 alkyl, —C(O)—C1-4 alkylene-OH or —SO2—C1-4 alkylene-OH. One of ordinary skill in the art will appreciate that the R3 substituent on the piperazine nitrogen renders that nitrogen “non-basic.” It will be appreciated that such a non-basic nitrogen moiety is not amenable to acting as a proton-acceptor as compared, for example, to the corresponding secondary amine or alkyl substituted derivative thereof.


In some embodiments, the present invention provides a compound of formula III or III-a, wherein W is —NH—.


In certain embodiments, the present invention provides a compound of formula III or III-a, wherein W is —NH— and R2 is —CF3.


In certain embodiments, the present invention provides a compound of formula III or III-a, wherein W is —O— and R2 is —Cl.


In some embodiments, the present invention provides a compound of formula III or III-a wherein at least one; at least two; or all three of the following characteristics apply:

    • (a) W is —O— or —NH—;
    • (b) R2 is —CF3 or Cl; and
    • (c) R3 is —C(O)CH3 or —SO2CH3.


In some embodiments, the present invention provides a compound of formula III or III-a wherein at least one; at least two; or all three of the following characteristics apply:

    • (a) W is —NH—;
    • (b) R2 is —CF3 or Cl; and
    • (c) R3 is —C(O)CH3.


In some embodiments, the present invention provides a compound of formula III or III-a wherein at least one; at least two; or all three of the following characteristics apply:

    • (a) W is —NH—;
    • (b) R2 is —CF3 or Cl; and
    • (c) R3 is —SO2CH3.


In some embodiments, the present invention provides a compound of formula III or III-a wherein at least one; at least two; or all three of the following characteristics apply:

    • (a) W is —O—;
    • (b) R2 is —CF3 or Cl; and
    • (c) R3 is —C(O)CH3.


In some embodiments, the present invention provides a compound of formula III or III-a wherein at least one; at least two; or all three of the following characteristics apply:

    • (a) W is —O—;
    • (b) R2 is Cl; and
    • (c) R3 is —C(O)CH3.


In some embodiments, the present invention provides a compound of formula III or III-a wherein at least one; at least two; or all three of the following characteristics apply:

    • (a) W is —O—;
    • (b) R2 is —CF3 or Cl; and
    • (c) R3 is —SO2CH3.


In some embodiments, the present invention provides a compound of formula III or III-a wherein at least one; at least two; or all three of the following characteristics apply:

    • (a) W is —O—;
    • (b) R2 is Cl; and
    • (c) R3 is —SO2CH3.


Exemplary compounds of formula I are set forth in Table 1, below.









TABLE 1





Exemplary Compounds


















embedded image


I-1







embedded image


I-2







embedded image


I-3







embedded image


I-4







embedded image


I-5







embedded image


I-6







embedded image


I-7







embedded image


I-8







embedded image


I-9







embedded image


I-10







embedded image


I-11







embedded image


I-12







embedded image


I-13







embedded image


I-14







embedded image


I-15







embedded image


I-16







embedded image


I-17







embedded image


I-18







embedded image


I-19







embedded image


I-20







embedded image


I-21







embedded image


I-22







embedded image


I-23







embedded image


I-24







embedded image


I-25







embedded image


I-26







embedded image


I-27







embedded image


I-28







embedded image


I-29







embedded image


I-30







embedded image


I-31







embedded image


I-32







embedded image


I-33







embedded image


I-34







embedded image


I-35







embedded image


I-36







embedded image


I-37







embedded image


I-38







embedded image


I-39







embedded image


I-40







embedded image


I-41







embedded image


I-42







embedded image


I-43







embedded image


I-44







embedded image


I-45







embedded image


I-46







embedded image


I-47







embedded image


I-48







embedded image


I-49







embedded image


I-50









In certain embodiments, the present invention provides a compound set forth in Table 1, above, or a pharmaceutically acceptable salt thereof.


In certain embodiments, a provided compound does not have the structure




embedded image


As used herein, the term “pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like.


Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(C1-4alkyl)4 salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.


2. Description of Exemplary Embodiments


As described in detail herein, infra, provided compounds are selective inhibitors of at least one mutation of EGFR. It has been surprisingly found that provided compounds are selective inhibitors of at least one mutation of EGFR as compared to wild-type (“WT”) EGFR. In certain embodiments, an at least one mutation of EGFR is T790M. In certain embodiments, the at least one mutation of EGFR is a deletion mutation. In some embodiments, the at least one mutation of EGFR is an activating mutation. In certain embodiments, a provided compound selectively inhibits at least one resistant mutation and at least one activating mutation as compared to WT EGFR. In some embodiments, a provided compound selectively inhibits at least one deletion mutation and/or at least one point mutation, and is sparing as to WT EGFR inhibition.


A mutation of EGFR can be selected from T790M (resistant or oncogenic), L858R (activating), delE746-A750 (activating), G719S (activating), or a combination thereof.


As used herein, the term “selectively inhibits,” as used in comparison to inhibition of WT EGFR, means that a provided compound inhibits at least one mutation of EGFR (i.e., at least one deletion mutation, at least one activating mutation, at least one restistant mutation, or a combination of at least one deletion mutation and at least one point mutation) in at least one assay described herein (e.g., biochemical or cellular). In some embodiments, the term “selectively inhibits,” as used in comparison to WT EGFR inhibition means that a provided compound is at least 50 times more potent, at least 45 times, at least 40, at least 35, at least 30, at least 25, or at least 20 times more potent as an inhibitor of at least one mutation of EGFR, as defined and described herein, as compared to WT EGFR.


As used herein, the term “sparing as to WT EGFR” means that a selective inhibitor of at least one mutation of EGFR, as defined and described above and herein, inhibits EGFR at the upper limit of detection of at least one assay as described herein (e.g., biochemical or cellular as described in detail in Examples 56-58). In some embodiments, the term “sparing as to WT EGFR” means that a provided compound inhibits WT EGFR with an IC50 of at least 10 μM, at least 9 μM, at least 8 μM, at least 7 μM, at least 6 μM, at least 5 μM, at least 3 μM, at least 2 μM, or at least 1 μM.


In certain embodiments, a provided compound selectively inhibits (a) at least one activating mutation; and (b) T790M; and (c) is sparing as to WT. In some embodiments, an at least one activating mutation is a deletion mutation. In some embodiments, an at least one activating mutation is a point mutation. In some embodiments, an activating mutation is delE746-A750. In some embodiments, an activating mutation is L858R. In some embodiments, an activating mutation is G719S.


In some embodiments, the at least one mutation of EGFR is L858R and/or T790M.


Without wishing to be bound by any particular theory, it is believed that administration of a provided compound to a patient having at least one activating mutation may preempt formation of the T790M resistance mutation. Thus, in certain embodiments, the present invention provides a method for inhibiting an activating mutation in a patient comprising administering to the patient a provided compound or composition thereof, as described herein.


One of ordinary skill in the art will appreciate that certain patients have an oncogenic form of the T790M mutation, i.e., the T790M mutation is present prior to administration to the patient any EGFR inhibitor and is therefore oncogenic. Accordingly, in some embodiments, the present invention provides a method for inhibiting oncogenic T790M in a patient comprising administering to the patient a provided compound or composition thereof, as described herein.


Tarceva (erlotinib) and Iressa (gefitinib) are first-line therapies for patients with activating mutations but exhibit dose-limiting toxicities due to concurrent inhibition of WT EGFR. In addition, drugs currently in development, including second generation covalent inhibitors, such as BIBW2992, HKI-272 and PF-0299804, are effective against the T790M resistance mutation but exhibit dose-limiting toxicities due to concurrent inhibition of WT EGFR.


It has been surprisingly found that provided compounds selectively inhibit each of the EGFR activating and deletion mutations. Moreover, provided compounds are sparing for WT EGFR and associated dose-limiting toxicities.


This stands in contrast to other known EGFR inhibitors (e.g., BIBW2992 and HKI-272) which are only somewhat effective against mutants but retain activity against WT EGFR and are therefore limited by toxicities associated with inhibition of WT EGFR. Table 2, below, sets forth GI50 values of Tarceva, BIBW2992 and HKI-272 as compared with provided compounds I-2 and I-4 (where compound numbers correspond to compound numbers in Table 1, supra). The data shown in Table 2 correspond to GI50 values obtained in the cellular proliferation assay described in detail in Example 58, where A431 cells express WT EGFR, HCC827 express EGFR having the deletion mutation delE746-A750, and H1975 cells express EGFR having a double mutation L858R/T790M.









TABLE 2







Comparative GI50 Values (nM)












Cell Line
Tarceva
BIBW2992
HKI-272
I-2
I-4















A431
298
20
4
>1000
500-1000


HCC827
12
<5
78
10-100
10-100


H1975
>5000
196
13
10-100
10-100









In some embodiments, a provided compound is at least 50, at least 45 times, at least 40, at least 35, at least 30, at least 25, or at least 20 times more potent for at least one mutation of EGFR as compared to WT EGFR, as determined by the biochemical assay described in detail in Example 56, infra. In certain embodiments, a provided compound is at least 20, at least 15, or at least 10 times more potent for at least one mutation of EGFR as compared to WT EGFR, as determined by the cellular assay described in detail in Example 58, infra.


In some embodiments, a provided compound is at least 50, at least 45 times, at least 40, at least 35, at least 30, at least 25, or at least 20 times more potent for at least one deletion mutation of EGFR as compared to WT EGFR, as determined by the biochemical assay described in detail in Example 56, infra. In certain embodiments, a provided compound is at least 20, at least 15, or at least 10 times more potent for at least one deletion mutation of EGFR as compared to WT EGFR, as determined by the cellular assay described in detail in Example 58, infra.


In some embodiments, a provided compound is at least 50, at least 45 times, at least 40, at least 35, at least 30, at least 25, or at least 20 times more potent for L858R and/or T790M mutation of EGFR as compared to WT EGFR, as determined by the biochemical assay described in detail in Example 56, infra. In certain embodiments, a provided compound is at least 20, at least 15, or at least 10 times more potent for L858R and/or T790M mutation of EGFR as compared to WT EGFR, as determined by the cellular assay described in detail in Example 58, infra.


In some embodiments, a provided compound is at least 20, at least 15, or at least 10 times more potent for double mutant in H1975 cells, as compared to WT EGFR, in the signaling assay described in detail in Example 57.


In certain embodiments, a provided compound inhibits at least one mutation of EGFR selectively as compared to WT EGFR and as compared to other protein kinases (e.g., ErbB2, ErbB4, a TEC-kinase, and/or JAK3). It will be appreciated that the acrylamide moiety, depicted in formula I, is a warhead group for covalently binding to a key cysteine residue in the binding domain of at least one mutation of EGFR selectively as compared to WT EGFR and other protein kinases. Protein kinases having a cysteine residue in the binding domain are known to one of ordinary skill in the art. Such protein kinases having a cysteine residue in the binding domain include the TEC-family of protein kinases (including TEC, BTK, ITK, BMX, JAK3, and RLK). In certain embodiments, the cysteine residue is conserved across a protein kinase sub-family, such as ErbB1 (commonly referred to as EGFR), ErbB2, and ErbB4.


Without wishing to be bound by any particular theory, it is believed that provided compounds irreversibly inhibit (i.e., covalently modify) at least one mutation of EGFR selectively as compared to WT EGFR and other protein kinases. In some embodiments, a provided compound irreversibly inhibits at least one mutation of EGFR selectively as compared to at least one protein kinase selected from ErbB1, ErbB2, ErbB4, TEC, BTK, ITK, BMX, JAK3, or RLK.


Notwithstanding, in certain embodiments, provided compounds do not appreciably inhibit, either reversibly or irreversibly, other protein kinases. In some embodiments, a provided compound is selective for inhibiting at least one mutatnt of EGFR as compared to off-target protein kinases thereby avoiding effects and toxicities associated with inhibition thereof.


3. Synthesis and Intermediates


In certain embodiments, a provided compound is synthesized using one or more of the following steps and intermediates.




embedded image



wherein R2 and W are as defined and described in classes and subclasses herein.


An R2-substituted 2,4-dichloropyrimidine is allowed to react with, for example, a Boc-protected 3-aminophenol in step 1 to form Intermediate S1. In certain embodiments, step 1 is performed under basic conditions. In some embodiments, step 1 is performed in the presence of a tertiary amine. In certain embodiments, step 1 is performed in the presence of Hunig's base. In some embodiments, step 1 is performed in a protic solvent. In some embodiments, step 1 is performed in an alcohol solvent. In certain embodiments, step 1 is performed in n-butanol.


In step 2, Intermediate S1 is deprotected to form Intermediate S2. In some embodiments, Intermediate S1 is deprotected using acid. In certain embodiments, Intermediate S1 is deprotected in the presence of trifluoroacetic acid.


In step 3, Intermediate S2 is acylated with an acryloyl group to form Intermediate S3. In certain embodiments, acryoyl chloride is the acylating agent. In certain embodiments, step 3 is performed in a halogenated solvent. In certain embodiments, step 3 is performed in dichloromethane.


Intermediate S3 can be reacted with various anilines to form compounds as described herein.


4. Uses, Formulation and Administration


Pharmaceutically Acceptable Compositions


According to another embodiment, the invention provides a composition comprising a compound of this invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle. An amount of compound in a composition of this invention is such that is effective to measurably inhibit a protein kinase, particularly to inhibit at least one mutant of EGFR selectively as compared to WT EGFR, in a biological sample or in a patient. In certain embodiments, an at least one mutant of EGFR is T790M. In certain embodiments, the at least one mutant of EGFR is a deletion mutation of EGFR. In some embodiments, the at least one mutation of EGFR is L858R and/or T790M.


In certain embodiments, an amount of compound in a provided composition is such that is effective to measurably inhibit at least one mutation of EGFR selectively as compared to WT EGFR.


In certain embodiments, an amount of compound in a provided composition is such that is effective to measurably inhibit at least one mutation of EGFR selectively as compared to WT EGFR and other protein kinases (e.g., ErbB2, ErbB4, a TEC-kinase, and/or JAK3).


In certain embodiments, the amount of compound in a provided composition is such that is effective to measurably inhibit at least one mutant of EGFR selectively as compared to WT EGFR, in a biological sample or in a patient. In certain embodiments, a composition of this invention is formulated for administration to a patient in need of such composition. In some embodiments, a composition of this invention is formulated for oral administration to a patient.


In certain embodiments, the amount of compound in a provided composition is such that is effective to measurably inhibit at least one mutant of EGFR selectively as compared to WT EGFR, in a biological sample or in a patient. In certain embodiments, a composition of this invention is formulated for administration to a patient in need of such composition. In some embodiments, a composition of this invention is formulated for oral administration to a patient.


In certain embodiments, the amount of compound in a provided composition is such that is effective to measurably inhibit at least one mutant of EGFR selectively as compared to WT EGFR and other protein kinases (e.g., ErbB2, ErbB4, a TEC-kinase, and/or JAK3), in a biological sample or in a patient. In certain embodiments, a composition of this invention is formulated for administration to a patient in need of such composition. In some embodiments, a composition of this invention is formulated for oral administration to a patient.


The term “patient”, as used herein, means an animal, preferably a mammal, and most preferably a human.


The term “pharmaceutically acceptable carrier, adjuvant, or vehicle” refers to a nontoxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of this invention 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, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.


Compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term “parenteral” as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension 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 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 may be employed including synthetic mono- or di-glycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used surfactants, such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.


Pharmaceutically acceptable compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.


Alternatively, pharmaceutically acceptable compositions of this invention may be administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols.


Pharmaceutically acceptable compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.


Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches may also be used.


For topical applications, provided pharmaceutically acceptable compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers. Carriers for topical administration of compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, provided pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.


For ophthalmic use, provided pharmaceutically acceptable compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride. Alternatively, for ophthalmic uses, the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum.


Pharmaceutically acceptable compositions of this invention may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.


In certain embodiments, pharmaceutically acceptable compositions of this invention are formulated for oral administration.


The amount of compounds of the present invention that may be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration. Preferably, provided compositions should be formulated so that a dosage of between 0.01-100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions.


It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated. The amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.


Uses of Compounds and Pharmaceutically Acceptable Compositions


Compounds and compositions described herein are generally useful for the selective inhibition of at least one mutant of EGFR as compared to WT EGFR. In certain embodiments, an at least one mutant of EGFR is T790M. In certain embodiments, the at least one mutant of EGFR is a deletion mutation of EGFR, an activating mutation of EGFR, or a combination thereof. In some embodiments, the at least one mutation of EGFR is L858R and/or T790M.


In certain embodiments, a provided compound selectively inhibits (a) at least one activating mutation, (b) T790M, and (c) is sparing as to WT. In some embodiments, an at least one activating mutation is a deletion mutation. In some embodiments, an at least one activating mutation is a point mutation. In some embodiments, an activating mutation is delE746-A750. In some embodiments, an activating mutation is L858R. In some embodiments, an activating mutation is G719S.


In some embodiments, the at least one mutation of EGFR is L858R and/or T790M.


The activity of a compound utilized in this invention as a selective inhibitor of at least one mutant of EGFR as compared to WT EGFR, may be assayed in vitro, in vivo or in a cell line. In vitro assays include assays that determine inhibition of the phosphorylation activity and/or the subsequent functional consequences, or ATPase activity of activated EGFR (WT or mutant). Alternate in vitro assays quantitate the ability of the inhibitor to bind to EGFR (WT or mutant). Inhibitor binding may be measured by radiolabeling the inhibitor prior to binding, isolating the inhibitor/EGFR (WT or mutant) complex and determining the amount of radiolabel bound. Alternatively, inhibitor binding may be determined by running a competition experiment where new inhibitors are incubated with EGFR (WT or mutant) bound to known radioligands. Detailed conditions for assaying a compound utilized in this invention as an inhibitor of EGFR (WT or mutant), are set forth in the Examples below.


Protein tyrosine kinases are a class of enzymes that catalyze the transfer of a phosphate group from ATP or GTP to a tyrosine residue located on a protein substrate. Receptor tyrosine kinases act to transmit signals from the outside of a cell to the inside by activating secondary messaging effectors via a phosphorylation event. A variety of cellular processes are promoted by these signals, including proliferation, carbohydrate utilization, protein synthesis, angiogenesis, cell growth, and cell survival.


As used herein, the terms “treatment,” “treat,” and “treating” refer to reversing, alleviating, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more symptoms thereof, as described herein. In some embodiments, treatment may be administered after one or more symptoms have developed. In other embodiments, treatment may be administered in the absence of symptoms. For example, treatment may be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment may also be continued after symptoms have resolved, for example to prevent or delay their recurrence.


Provided compounds are inhibitors of at least one mutant of EGFR and are therefore useful for treating one or more disorders associated with activity of one of more EGFR mutants (e.g., a deletion mutation, an activating mutation, a resistant mutation, or combination thereof). Thus, in certain embodiments, the present invention provides a method for treating a mutant EGFR-mediated disorder comprising the step of administering to a patient in need thereof a compound of the present invention, or pharmaceutically acceptable composition thereof.


As used herein, the term “mutant EGFR-mediated” disorders or conditions as used herein means any disease or other deleterious condition in which at least one mutant of EGFR is known to play a role. In certain embodiments, an at least one mutant of EGFR is T790M. In some embodiments, the at least one mutant of EGFR is a deletion mutation. In certain embodiments, the at least one mutant of EGFR is an activating mutation. In some embodiments, the at least one mutant of EGFR is L858R and/or T790M. In certain embodiments, a provided compound selectively inhibits (a) at least one activating mutation, (b) T790M, and (c) is sparing as to WT. In some embodiments, an at least one activating mutation is a deletion mutation. In some embodiments, an at least one activating mutation is a point mutation. In some embodiments, an activating mutation is delE746-A750. In some embodiments, an activating mutation is L858R. In some embodiments, an activating mutation is G719S.


Accordingly, another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which at least one mutant of EGFR is known to play a role. Specifically, the present invention relates to a method of treating or lessening the severity of a disease or condition selected from a proliferative disorder, wherein said method comprises administering to a patient in need thereof a compound or composition according to the present invention.


In some embodiments, the present invention provides a method for treating or lessening the severity of one or more disorders selected from a cancer. In some embodiments, the cancer is associated with a solid tumor. In certain embodiments, the cancer is breast cancer, glioblastoma, lung cancer, cancer of the head and neck, colorectal cancer, bladder cancer, or non-small cell lung cancer. In some embodiments, the present invention provides a method for treating or lessening the severity of one or more disorders selected from squamous cell carcinoma, salivary gland carcinoma, ovarian carcinoma, or pancreatic cancer.


In certain embodiments, the present invention provides a method for treating or lessening the severity of neurofibromatosis type I (NF1), neurofibromatosis type II (NF2) Schwann cell neoplasms (e.g. MPNST's), or Schwannomas.


The compounds and compositions, according to the method of the present invention, may be administered using any amount and any route of administration effective for treating or lessening the severity of a cancer. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like. Compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage. The expression “dosage unit form” as used herein refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular patient or organism 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 like factors well known in the medical arts. The term “patient”, as used herein, means an animal, preferably a mammal, and most preferably a human.


Pharmaceutically acceptable compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated. In certain embodiments, the compounds of the invention may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg or from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.


Liquid dosage forms for oral administration include, but are not limited to, 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.


Injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, terminal (heat) sterilization, or sterilization via ionizing radiationor by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.


In order to prolong the effect of a compound of the present invention, it is often desirable to slow the absorption of the compound 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 compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.


Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention 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 dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.


Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. 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 polethylene glycols and the like. In some embodiments, a solid composition is a liquid filled hard gelatin capsule or solid dispersion.


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. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.


Dosage forms for topical or transdermal administration of a compound of this invention 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, and eye drops are also contemplated as being within the scope of this invention. Additionally, the present invention contemplates the use of transdermal patches, which 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 another embodiment, the invention relates to a method of inhibiting at least one mutant of EGFR (e.g., a deletion mutation, an activating mutation, a resistant mutations, or combination thereof) activity in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound. In certain embodiments, the invention relates to a method of irreversibly inhibiting at least one mutant of EGFR (e.g., a deletion mutation, an activating mutation, a resistant mutation, or combination thereof) activity in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.


In certain embodiments, a provided compound selectively inhibits in a biological sample (a) at least one activating mutation, (b) T790M, and (c) is sparing as to WT. In some embodiments, an at least one activating mutation is a deletion mutation. In some embodiments, an at least one activating mutation is a point mutation. In some embodiments, an activating mutation is delE746-A750. In some embodiments, an activating mutation is L858R. In some embodiments, an activating mutation is G719S.


The term “biological sample”, as used herein, includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.


Inhibition of at least one mutant of EGFR (e.g., a deletion mutation, an activating mutation, a resistant mutation, or combination thereof) activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ transplantation, biological specimen storage, and biological assays.


Another embodiment of the present invention relates to a method of inhibiting at least one mutant of EGFR (e.g., a deletion mutation, an activating mutation, a resistant mutation, or combination thereof) activity in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound. In certain embodiments, the present invention provides a method for inhibiting (a) at least one activating mutation, and (b) T790M in a patient, and (c) is sparing as to WT, wherein said method comprises administering to the patient a provided compound, or composition thereof. In some embodiments, an at least one activating mutation is a deletion mutation. In some embodiments, an at least one activating mutation is a point mutation. In some embodiments, the present invention provides a method for inhibiting at least one mutant of EGFR in a patient, wherein an activating mutation is delE746-A750. In some embodiments, the present invention provides a method for inhibiting at least one mutant of EGFR in a patient, wherein an activating mutation is L858R. In some embodiments, the present invention provides a method for inhibiting at least one mutant of EGFR in a patient, wherein an activating mutation is G719S.


According to another embodiment, the invention relates to a method of inhibiting at least one mutant of EGFR (e.g., a deletion mutation, an activating mutation, a resistant mutation, or combination thereof) activity in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound. According to certain embodiments, the invention relates to a method of irreversibly inhibiting at least one mutant of EGFR activity (e.g., a deletion mutation, an activating mutation, a resistant mutation, or combination thereof) in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound. In certain embodiments, the present invention provides a method for irreversibly inhibiting (a) at least one activating mutation, and (b) T790M in a patient, and (c) is sparing as to WT, wherein said method comprises administering to the patient a provided compound, or composition thereof. In some embodiments, an irreversibly inhibited at least one activating mutation is a deletion mutation. In some embodiments, an irreversibly inhibited at least one activating mutation is a point mutation. In some embodiments, the present invention provides a method for irreversibly inhibiting at least one mutant of EGFR in a patient, wherein an activating mutation is delE746-A750. In some embodiments, the present invention provides a method for irreversibly inhibiting at least one mutant of EGFR in a patient, wherein an activating mutation is L858R. In some embodiments, the present invention provides a method for irreversibly inhibiting at least one mutant of EGFR in a patient, wherein an activating mutation is G719S.


In other embodiments, the present invention provides a method for treating a disorder mediated by one or more of at least one mutant of EGFR (e.g., a deletion mutation, an activating mutation, a resistant mutation, or combination thereof) in a patient in need thereof, comprising the step of administering to said patient a compound according to the present invention or pharmaceutically acceptable composition thereof. Such disorders are described in detail herein.


Depending upon the particular condition, or disease, to be treated, additional therapeutic agents, which are normally administered to treat that condition, may also be present in the compositions of this invention. As used herein, additional therapeutic agents that are normally administered to treat a particular disease, or condition, are known as “appropriate for the disease, or condition, being treated.”


For example, compounds of the present invention, or a pharmaceutically acceptable composition thereof, are administered in combination with chemotherapeutic agents to treat proliferative diseases and cancer. Examples of known chemotherapeutic agents include, but are not limited to, Adriamycin, dexamethasone, vincristine, cyclophosphamide, fluorouracil, topotecan, taxol, interferons, platinum derivatives, taxane (e.g., paclitaxel), vinca alkaloids (e.g., vinblastine), anthracyclines (e.g., doxorubicin), epipodophyllotoxins (e.g., etoposide), cisplatin, an mTOR inhibitor (e.g., a rapamycin), methotrexate, actinomycin D, dolastatin 10, colchicine, emetine, trimetrexate, metoprine, cyclosporine, daunorubicin, teniposide, amphotericin, alkylating agents (e.g., chlorambucil), 5-fluorouracil, campthothecin, cisplatin, metronidazole, and Gleevec™, among others. In other embodiments, a compound of the present invention is administered in combination with a biologic agent, such as Avastin or VECTIBIX.


In certain embodiments, compounds of the present invention, or a pharmaceutically acceptable composition thereof, are administered in combination with an antiproliferative or chemotherapeutic agent selected from any one or more of abarelix, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, amifostine, anastrozole, arsenic trioxide, asparaginase, azacitidine, BCG Live, bevacuzimab, fluorouracil, bexarotene, bleomycin, bortezomib, busulfan, calusterone, capecitabine, camptothecin, carboplatin, carmustine, celecoxib, cetuximab, chlorambucil, cladribine, clofarabine, cyclophosphamide, cytarabine, dactinomycin, darbepoetin alfa, daunorubicin, denileukin, dexrazoxane, docetaxel, doxorubicin (neutral), doxorubicin hydrochloride, dromostanolone propionate, epirubicin, epoetin alfa, erlotinib, estramustine, etoposide phosphate, etoposide, exemestane, filgrastim, floxuridine fludarabine, fulvestrant, gefitinib, gemcitabine, gemtuzumab, goserelin acetate, histrelin acetate, hydroxyurea, ibritumomab, idarubicin, ifosfamide, imatinib mesylate, interferon alfa-2a, interferon alfa-2b, irinotecan, lenalidomide, letrozole, leucovorin, leuprolide acetate, levamisole, lomustine, megestrol acetate, melphalan, mercaptopurine, 6-MP, mesna, methotrexate, methoxsalen, mitomycin C, mitotane, mitoxantrone, nandrolone, nelarabine, nofetumomab, oprelvekin, oxaliplatin, paclitaxel, palifermin, pamidronate, pegademase, pegaspargase, pegfilgrastim, pemetrexed disodium, pentostatin, pipobroman, plicamycin, porfimer sodium, procarbazine, quinacrine, rasburicase, rituximab, sargramostim, sorafenib, streptozocin, sunitinib maleate, talc, tamoxifen, temozolomide, teniposide, VM-26, testolactone, thioguanine, 6-TG, thiotepa, topotecan, toremifene, tositumomab, trastuzumab, tretinoin, ATRA, uracil mustard, valrubicin, vinblastine, vincristine, vinorelbine, zoledronate, or zoledronic acid.


Other examples of agents the inhibitors of this invention may also be combined with include, without limitation: treatments for Alzheimer's Disease such as donepezil hydrochloride (Aricept®) and rivastigmine (Exelon®); treatments for Parkinson's Disease such as L-DOPA/carbidopa, entacapone, ropinrole, pramipexole, bromocriptine, pergolide, trihexephendyl, and amantadine; agents for treating Multiple Sclerosis (MS) such as beta interferon (e.g., Avonex® and Rebif®), glatiramer acetate)(Copaxone®, and mitoxantrone; treatments for asthma such as albuterol and montelukast (Singulair®); agents for treating schizophrenia such as zyprexa, risperdal, seroquel, and haloperidol; anti-inflammatory agents such as corticosteroids, TNF blockers, IL-1 RA, azathioprine, cyclophosphamide, and sulfasalazine; immunomodulatory and immunosuppressive agents such as cyclosporin, tacrolimus, rapamycin, mycophenolate mofetil, interferons, corticosteroids, cyclophophamide, azathioprine, and sulfasalazine; neurotrophic factors such as acetylcholinesterase inhibitors, MAO inhibitors, interferons, anti-convulsants, ion channel blockers, riluzole, and anti-Parkinsonian agents; agents for treating cardiovascular disease such as beta-blockers, ACE inhibitors, diuretics, nitrates, calcium channel blockers, and statins; agents for treating liver disease such as corticosteroids, cholestyramine, interferons, and anti-viral agents; agents for treating blood disorders such as corticosteroids, anti-leukemic agents, and growth factors; and agents for treating immunodeficiency disorders such as gamma globulin.


In certain embodiments, compounds of the present invention, or a pharmaceutically acceptable composition thereof, are administered in combination with a monoclonal antibody or an siRNA therapeutic.


Those additional agents may be administered separately from an inventive compound-containing composition, as part of a multiple dosage regimen. Alternatively, those agents may be part of a single dosage form, mixed together with a compound of this invention in a single composition. If administered as part of a multiple dosage regime, the two active agents may be submitted simultaneously, sequentially or within a period of time from one another normally within five hours from one another.


As used herein, the term “combination,” “combined,” and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this invention. For example, a compound of the present invention may be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form. Accordingly, the present invention provides a single unit dosage form comprising a provided compound, an additional therapeutic agent, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.


The amount of both, an inventive compound and additional therapeutic agent (in those compositions which comprise an additional therapeutic agent as described above)) that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. Preferably, compositions of this invention should be formulated so that a dosage of between 0.01-100 mg/kg body weight/day of an inventive can be administered.


In those compositions which comprise an additional therapeutic agent, that additional therapeutic agent and the compound of this invention may act synergistically. Therefore, the amount of additional therapeutic agent in such compositions will be less than that required in a monotherapy utilizing only that therapeutic agent. In such compositions a dosage of between 0.01-1,000 μg/kg body weight/day of the additional therapeutic agent can be administered.


The amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent. Preferably the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent.


The compounds of this invention, or pharmaceutical compositions thereof, may also be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents and catheters. Vascular stents, for example, have been used to overcome restenosis (re-narrowing of the vessel wall after injury). However, patients using stents or other implantable devices risk clot formation or platelet activation. These unwanted effects may be prevented or mitigated by pre-coating the device with a pharmaceutically acceptable composition comprising a kinase inhibitor. Implantable devices coated with a compound of this invention are another embodiment of the present invention.


EXEMPLIFICATION

As depicted in the Examples below, in certain exemplary embodiments, compounds are prepared according to the following general procedures. It will be appreciated that, although the general methods depict the synthesis of certain compounds of the present invention, the following general methods, and other methods known to one of ordinary skill in the art, can be applied to all compounds and subclasses and species of each of these compounds, as described herein.


Compound numbers utilized in the Examples below correspond to compound numbers set forth in Table 1, supra.


Provided compounds are prepared according to methods known to one of ordinary skill in the art and include methods described in detail in US 20100029610, published Feb. 4, 2010, the entirety of which is hereby incorporated herein by reference.


Example 1
Intermediate 1



embedded image



Step 1:


In a 25 mL 3-neck RBF previously equipped with a magnetic stirrer, Thermo pocket and CaCl2 guard tube, N-Boc-1,3-diaminobenzene (0.96 g) and n-butanol (9.00 mL) were charged. Reaction mixture was cooled to 0° C. 2,4-Dichloro-5-trifluoromethylpyrimidine (1.0 g) was added dropwise to the above reaction mixture at 0° C. The DIPEA (0.96 mL) was dropwise added to the above reaction mixture at 0° C. and the reaction mixture was stirred for 1 hr at 0° C. to 5° C. Finally the reaction mixture was allowed to warm to room temperature. Reaction mixture was stirred for another 4 hrs at room temperature. Completion of reaction was monitored by TLC using hexane:ethyl acetate (7:3). The solid precipitated out was filtered off and washed with 1-butanol (2 mL). Solid was dried under reduced pressure at 40° C. for 1 hr. 1H-NMR (DMSO-d6, 400 MHz) δ 1.48 (S, 9 H), 7.02 (m, 1 H), 7.26 (m, 2 H), 7.58 (S, 1 H), 8.57 (S, 1 H), 9.48 (S, 1 H), 9.55 (S, 1 H).


Step 2:


To the above crude (3.1 g) in DCM (25 mL) was added TFA (12.4 mL) slowly at 0° C. The reaction mixture was allowed to warm to room temperature. Reaction mixture was stirred for another 10 min at room temperature. The crude was concentrated under reduced pressure.


Step 3:


The concentrated crude was dissolved in DIPEA (2.0 mL) and DCM (25 mL), and then cooled to −30° C. To the reaction mixture was slowly added acryloyl chloride (0.76 g) at −30° C. The reaction mass was warmed to room temperature stirred at room temperature for 1.0 hr. The reaction was monitored on TLC using hexane:ethyl acetate (7:3) as mobile phase. Reaction got completed after 1 hr. 1H-NMR (DMSO-d6, 400 MHz) δ 5.76 (dd, J=2.0, 10.0 Hz, 1 H), 6.24 (dd, J=2.0, 17.2 Hz, 1 H), 6.48 (m, 1 H), 7.14 (d, J=8.8 Hz, 1 H), 7.37 (t, J=8.0 Hz, 1 H), 7.94 (S, 1 H), 8.59 (S, 1 H), 9.60 (S, 1 H), 10.26 (S, 1 H).


Example 2
Compound I-2 N-(3-(2-(2-methoxy-4-morpholinophenylamino)-5-(trifluoromethyl)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


To obtain the title compound I-2, a mixture of intermediate 1 in Example 1 (16 mg) and 2-methoxy-4-morpholinoaniline in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the title compound as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.4 (S, 1 H), 9.72 (br, 1 H), 9.18 (br, 1 H), 8.49 (br, 1 H), 7.83 (S, 1 H), 7.59 (d, J=8.4 Hz, 1 H), 7.31-7.48 (m, 2 H), 7.41 (t, J=15.2 Hz, 1 H), 7.12 (br, 1 H), 6.67 (S, 1 H), 6.49 (dd, J=10.0, 16.8 Hz, 1 H), 6.25 (dd, J=2.0, 16.8 Hz, 1 H), 5.77 (dd, J=2.0, 10.0 Hz, 1 H), 3.7-3.9 (m, 7 H), 3.1 (br, 4 H); calculated mass for C25H25F3N6O3: 514.2. found: 515.5 (M+H+).


Example 3
Compound I-4 N-(3-(2-(4-(4-acetylpiperazin-1-yl)-2-methoxyphenylamino)-5-(trifluoromethyl)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


Using 2-methoxy-4-(4-acteylpiperazinyl)aniline and intermediate 1 in Example 1, the title compound I-4 was prepared as described in Example 2. 1H-NMR (DMSO-d6, 400 MHz) δ 10.2 (S, 1 H), 8.2 (br, 1 H), 8.30 (S, 1 H), 7.73 (br, 1 H), 7.52 (d, J=7.8 Hz, 1 H), 7.45 (d, J=7.8 Hz, 1 H), 7.26 (J=8.2 Hz, 1 H), 7.14 (be, 1 H), 6.60 (S, 1 H), 6.42 (dd, J=11.4, 16.9 Hz, 1 H), 6.24 (d, J=16.9 Hz, 1 H), 5.75 (d, J=11.4 Hz, 1 H), 3.76 (S, 3 H), 3.04 (br, 4 H), 2.04 (S, 3 H); calculated mass for C27H28F3N7O3: 555.2. found: 556.2 (M+H+).


Example 4
Intermediate 2



embedded image



Step 1:


The title step was executed according to Step 1 in Scheme 1 of Example 1. 1H-NMR (DMSO-d6, 400 MHz) δ 1.48 (S, 9 H), 7.16 (d, 1 H), 7.25 (m, 2 H), 7.70 (S, 1 H), 8.37 (S, 1 H), 9.47 (S, 1 H), 9.55 (S, 1 H).


Step 2:


The title step was executed according to Step 2 in Scheme 1 of Example 1.


Step 3:


The title step was executed according to Step 3 in Scheme 1 of Example 1. 1H-NMR (DMSO-d6, 400 MHz) δ 5.76 (dd, J=1.6, 10.8, Hz 1 H), 6.25 (dd, J=1.6, 16.8 Hz, 1 H), 6.46 (m, 1 H), 7.30 (m, 2 H), 7.46 (d, J=8.0 Hz, 1 H), 7.91 (S, 1 H), 8.38 (S, 1 H), 9.60 (S, 1 H), 10.23 (S, 1 H).


Example 5
Intermediate 3



embedded image



Step 1:


The title step was executed according to Step 1 in Scheme 1 of Example 1. 1H-NMR (DMSO-d6, 400 MHz) δ 1.47 (S, 9 H), 6.89 (d, J=7.6 Hz, 1 H), 7.35 (m, 2 H), 7.45 (S, 1 H), 8.89 (S, 1 H), 9.64 (S, 1 H).


Step 2:


The title step was executed according to Step 2 in Scheme 1 of Example 1.


Step 3:


The title step was executed according to Step 3 in Scheme 1 of Example 1. 1H-NMR (DMSO-d6, 400 MHz) δ 5.77 (d, J=10.0 Hz, 1 H), 6.25 (d, J=17.2 Hz, 1 H), 6.45 (m, 1H). 7.01 (d, J=7.2 Hz, 1 H), 7.53 (m, 2 H), 7.73 (S, 1 H), 8.98 (S, 1 H), 10.40 (S, 1 H).


Example 6
Intermediate 4



embedded image



Step 1:


The title step was executed according to Step 1 in Scheme 1 of Example 1. 1H-NMR (DMSO-d6, 400 MHz) δ 1.50 (S, 9 H), 7.10 (d, J=6.0 Hz, 1 H), 7.25 (m, 2 H), 8.44 (S, 1 H), 9.32 (S, 1 H), 9.47 (S, 1 H).


Step 2:


The title step was executed according to Step 2 in Scheme 1 of Example 1.


Step 3:


The title step was executed according to Step 3 in Scheme 1 of Example 1. 1H-NMR (DMSO-d6, 400 MHz) δ 5.76 (dd, J=1.6, 10.0 Hz, 1 H), 6.25 (dd, J=1.6, 16.8 Hz, 1 H), 6.43 (m, 1 H), 7.23 (d, J=8.0 Hz, 1 H), 7.35 (t, J=8.0 Hz, 1 H), 7.48 (d, J=8.0 Hz, 1 H), 7.80 (S, 1 H), 8.38 (S, 1 H), 9.36 (S, 1 H), 10.23 (S, 1 H).


Example 7
Intermediate 5



embedded image



Step 1:


The title step was executed according to Step 1 in Scheme 1 of Example 1. 1H-NMR (DMSO-d6, 400 MHz) δ 1.47 (S, 9 H), 6.90 (d, J=6.0 Hz, 1 H), 7.35 (m, 2 H), 7.50 (S, 1 H), 9.05 (S, 1 H), 9.65 (S, 1 H).


Step 2:


The title step was executed according to Step 2 in Scheme 1 of Example 1.


Step 3:


The title step was executed according to Step 3 in Scheme 1 of Example 1. 1H-NMR (DMSO-d6, 400 MHz) δ 5.76 (d, J=10.0 Hz, 1 H), 6.25 (dd, J=1.6, 16.8 Hz, 1 H), 6.46 (m, 2 H), 7.01 (d, J=8.0 Hz, 1 H), 7.08 (t, J=8.4 Hz, 1H) 7.25 (S, 1 H), 9.44 (S, 1 H), 10.02 (S, 1 H).


Example 8
Intermediate 6



embedded image



Step 1:


The title step was executed according to Step 1 in Scheme 1 of Example 1. 1H-NMR (DMSO-d6, 400 MHz) δ 1.47 (S, 9 H), 6.60 (S, 1 H), 6.86 (d, J=8.4 Hz, 1 H), 7.13 (t, J=7.6 Hz, 1 H), 7.36 (m, 1 H), 7.50 (S, 1 H), 8.40 (S, 1 H).


Step 2:


The title step was executed according to Step 2 in Scheme 1 of Example 1.


Step 3:


The title step was executed according to Step 3 in Scheme 1 of Example 1. 1H-NMR (DMSO-d6, 400 MHz) δ 5.78 (dd, J=2.0, 10.0 Hz, 1 H), 6.25 (dd, J=2.0, 17.2 Hz, 1 H), 6.40 (m, 1 H), 7.02 (d, 1 H), 7.50 (m, 2 H), 7.71 (S, 1 H), 8.40 (S, 1 H), 10.35 (S, 1 H).


Example 9
Compound I-5 N-(3-(5-chloro-2-(2-methoxy-4-morpholinophenylamino)pyrimidin-4-yloxy)phenyl)acrylamide)



embedded image


To obtain the title compound, a mixture of intermediate 6 in Example 8 and 2-methoxy-4-morpholinoaniline in n-butanol with catalytic HCl was microwaved for 20 min at 150° C. The crude was concentrated under reduced pressure and purified to give the title compound. 1H-NMR (chloroform-d, 400 MHz) δ 8.23 (S, 1 H), 7.6-7.8 (br, 2 H), 7.4-7.5 (m, 3 H), 7.00 (dd, J=1.4, 8.2 Hz, 1 H), 6.41 (m, 2 H), 6.23 (m, 2 H), 5.77 (dd, J=1.4, 10.1 Hz, 1 H), 3.84 (m, 4 H), 3.81 (S, 3 H), 3.04 (m, 4 H); calculated mass for C24H24ClN5O4: 481.2. found: 482.2 (M+H+).


Example 10
Compound I-6 N-(3-(2-(4-(4-(2-hydroxyacetyl)piperazin-1-yl)-2-methoxyphenylamino)-5-(trifluoromethyl)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


Using 2-methoxy-4-(4-(2-hydroxyacteyl)piperazinyl)aniline and intermediate 1 of Example 1, the title compound I-6 was prepared as described in Example 2. 1H-NMR (CDCl3, 400 MHz) δ 8.33 (S, 1 H), 8.08 (br, 1 H), 7.86 (br, 1 H), 7.60 (br, 1 H), 7.39 (m, 1 H), 6.89 (S, 1H), 6.22-6.55 (m, 3 H), 5.80 (d, J=10.0 Hz), 4.24 (S, 2 H), 3.90 (S, 2 H), 3.85 (S, 2 H), 3.64 (S, 1 H), 3.45 (S, 2 H), 3.13 (S, 3 H); calculated mass for C27H28F3N7O4: 571.2. found: 572.4 (M+H+).


Example 11
Compound I-7 N-(3-(2-(4-(4-acetyl-1,4-diazepan-1-yl)-2-methoxyphenylamino)-5-(trifluoromethyl)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


Using 1-(4-(4-amino-3-methoxyphenyl)-1,4-diazepan-1-yl)ethanone and intermediate 1 of Example 1, the title compound I-7 was prepared as described in Example 2. 1H-NMR (DMSO-d6, 400 MHz) δ 10.2 (S, 1 H), 9.2 (br, 1 H), 8.7 (br, 1 H), 8.4 (br, 1 H), 7.76 (br, 1 H), 7.49 (d, J=8.2 Hz, 1 H), 7.27 (br, 2 H), 7.1 (br, 1 H), 6.42 (dd, J=11.0, 16.5 Hz, 1 H), 6.30 (br, 1 H), 6.24 (d, J=16.5 Hz, 1 H), 5.9 (br, 1 H), 5.74 (d, J=11.0 Hz, 1 H), 3.3-3.7 (m, 4 H), 1.7-1.95 (m, 5 H); calculated mass for C28H30F3N7O3: 569.2. found: 570.2 (M+H+).


Example 12
Compound I-10 N-(3-(2-(4-(4-acetylpiperazin-1-yl)-2-methoxyphenylamino)-5-(trifluoromethyl)pyrimidin-4-yloxy)phenyl)acrylamide)



embedded image


To obtain the title compound, a mixture of intermediate 5 of Example 7 and 1-(4-(4-amino-3-methoxyphenyl)piperazin-1-yl)ethanone (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the title compound as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.32 (S, 1 H), 8.92 (S, 1 H), 8.60 (S, 1 H), 7.72 (t, J=2.3 Hz, 1 H), 7.58 (d, J=8.7 Hz, 1 H), 7.43 (m, 2 H), 6.98 (m, 1 H), 6.61 (d, J=2.3, 1 H), 6.42 (m, 2 H), 6.25 (dd, J=1.8, 16.9 Hz, 1 H), 5.77 (d, J=1.8, 10.1 Hz, 1 H), 3.7-4.0 (m, 4 H), 3.77 (S, 3 H), 3.1 (m, 4 H), 1.99 (S, 3 H); calculated mass for C27H27F3N6O4: 556.2. found: 557.1 (M+H+).


Example 13
Compound I-9 N-(3-(2-(4-(4-acetylpiperazin-1-yl)-2-methoxyphenylamino)-5-chloropyrimidin-4-yloxy)phenyl)acrylamide)



embedded image


Using 1-(4-(4-amino-3-methoxyphenyl)piperazin-1-yl)ethanone and intermediate 6 of Example 8, the title compound was prepared as described in Example 9. 1H-NMR (chloroform-d, 400 MHz) δ 8.26 (S, 1 H), 8.08 (br, 1 H), 7.93 (br, 1 H), 7.68 (S, 1 H), 7.57 (m, 1 H), 7.45 (m, 2 H), 6.96 (d, J=7.8 Hz, 1 H), 6.69 (S, 1 H), 6.60 (d, J=7.4 Hz, 1 H), 6.41 (d, J=1.4, 17.0 Hz, 1 H), 6.30 (dd, J=10.1, 16.5 Hz, 1 H), 5.75 (d, J=1.4, 10.1 Hz, 1 H), 3.97 (m, 2 H), 3.85 (S, 3 H), 3.82 (m, 2 H), 3.29 (m, 2 H), 3.24 (m, 2 H), 2.19 (S, 3 H); calculated mass for C26H27ClN6O4: 522.2. found: 523.2 (M+H+).


Example 14
Compound I-3 N-(3-(5-chloro-2-(4-(4-(2-hydroxyacetyl)piperazin-1-yl)-2-methoxyphenylamino)pyrimidin-4-yloxy)phenyl)acrylamide)



embedded image


Using 1-(4-(4-amino-3-methoxyphenyl)piperazin-1-yl)-2-hydroxyethanone and intermediate 6 of Example 8, the title compound was prepared as described in Example 9. 1H-NMR (chloroform-d, 400 MHz) δ 8.24 (S, 1 H), 7.71 (br, 1 H), 7.63 (m, 1 H), 7.49 (m, 1 H), 7.44 (t, J=8.2 Hz, 1 H), 7.39 (d, J=6.9 Hz, 2 H), 7.00 (dd, J=1.8, 7.8 Hz, 1 H), 6.45 (d, J=2.8 Hz, 1 H), 6.44 (dd, J=1.4, 16.9 Hz, 1 H), 6.23 (dd, J=10.1, 16.9 Hz, 1 H), 5.79 (dd, J=1.4, 10.1 Hz, 1 H), 4.22 (s, 2 H), 3.82 (S, 3 H), 3.80 (S, 2 H), 3.42 (m, 2 H), 3.06 (m, 4 H); calculated mass for C26H27ClN6O5: 538.2. found: 539.1 (M+H+).


Example 15
Compound I-8 N-(3-(2-(4-(4-acetylpiperazin-1-yl)-2-methoxyphenylamino)-5-chloropyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


To obtain the title compound, a mixture of intermediate 2 of Example 4 and 1-(4-(4-amino-3-methoxyphenyl)piperazin-1-yl)ethanone in n-butanol with catalytic HCl was microwaved for 20 min at 150° C. The crude was concentrated under reduced pressure and purified to give the title compound. 1H-NMR (DMSO-d6, 400 MHz) δ 10.2 (S, 1 H), 8.86 (S, 1 H), 8.07 (S, 1 H), 7.94 (br, 1 H), 7.70 (S, 1 H), 7.68 (S, 1 H), 7.46 (d, J=7.6 Hz, 1 H), 7.27 (m, 2H), 6.63 (d, J=2.4 Hz, 1 H), 6.46 (dd, J=10.0, 16.8 Hz, 1 H), 6.31 (br, 1 H), 6.29 (dd, J=2.0, 16.8 Hz, 1 H), 5.76 (dd, J=2.0, 10.0 Hz, 1 H), 3.79 (S, 3 H), 3.56 (m, 4 H), 3.0-3.2 (m, 4 H), 2.04 (S, 3 H); calculated mass for C26H28ClN7O3: 521.2. found: 522.4 (M+H+).


Example 16
Compound I-12 N-(3-(5-chloro-2-(4-(4-(2-hydroxyacetyl)piperazin-1-yl)-2-methoxyphenylamino)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


Using 1-(4-(4-amino-3-methoxyphenyl)piperazin-1-yl)-2-hydroxyethanone and intermediate 2 of Example 4, the title compound was prepared as described in Example 15. 1H-NMR (DMSO-d6, 400 MHz) δ 10.2 (S, 1 H), 8.86 (S, 1 H), 8.07 (S, 1 H), 7.94 (br, 1 H), 7.70 (m, 2 H), 7.46 (d, J=7.6 Hz, 1 H), 7.27 (m, 2 H), 6.63 (d, J=2.4 Hz, 1 H), 6.46 (dd, J=10.0, 16.8 Hz, 1 H), 6.31 (br, 1 H), 6.29 (dd, J=2.0, 16.8 Hz, 1 H), 5.76 (dd, J=2.0, 10.0 Hz, 1 H), 4.66 (t, J=5.6 Hz, 1 H), 4.14 (t, J=5.6 Hz, 2 H), 3.79 (S, 3 H), 3.61 (br, 2 H), 3.48 (br, 2 H), 3.05 (m, 4 H), 2.04 (S, 3 H); calculated mass for C26H28ClN7O4: 537.2. found: 538.4 (M+H+).


Example 17
Compound I-11 N-(3-(2-(4-(4-(2-hydroxyacetyl)piperazin-1-yl)-2-methoxyphenylamino)-5-(trifluoromethyl)pyrimidin-4-yloxy)phenyl)acrylamide)



embedded image


Using 1-(4-(4-amino-3-methoxyphenyl)piperazin-1-yl)-2-hydroxyethanone and intermediate 5 of Example 7, the title compound was prepared as described in Example 12. 1H-NMR (DMSO-d6, 400 MHz) δ 10.34 (S, 1 H), 8.84 (S, 1 H), 8.56 (br, 1 H), 7.63 (S, 1 H), 7.53 (d, J=8.0 Hz, 1 H), 7.41 (m, 1 H), 7.16 (m, 1 H), 6.96 (br, 1 H), 6.57 (br, 1 H), 6.45 (br, 1 H), 6.44 (dd, J=10.0, 16.8 Hz, 1 H), 6.29 (dd, J=1.6, 16.8 Hz, 1 H), 5.79 (dd, J=1.6, 10.0 Hz, 1 H), 4.66 (t, J=5.6 Hz, 1 H), 4.14 (d, J=5.6 Hz, 2 H), 3.73 (S, 3 H), 3.60 (br, 2 H), 3.47 (br, 2 H), 3.08 (br, 4 H); calculated mass for C27H27F3N6O5: 572.2. found: 573.6 (M+H+).


Example 18
Compound I-27, N-(3-(2-(2-methoxy-4-(4-sulfamoylpiperazin-1-yl)phenylamino)-5-(trifluoromethyl)pyrimidin-4-yloxy)phenyl)acrylamide)



embedded image


A mixture of the intermediate 5 (20 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N-methylmorpholine (20 uL), dioxane (0.5 mL), and sulfamide (50 mg). The reaction mixture was microwaved at 90° C. for 30 minutes. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.30 (s, 1 H), 8.81 (s, 1 H), 8.54 (br, 1 H), 7.62 (s, 1 H), 7.52 (d, J=8.2 Hz, 1 H), 7.39 (t, J=8.2 Hz, 1 H), 7.16 (d, J=8.7 Hz, 1 H), 6.95 (m, 1 H), 6.85 (s, 2 H), 6.58 (m, 1 H), 6.43 (dd, J=11.4, 17.0 Hz, 1 H), 6.26 (d, J=17.0 Hz, 1 H), 5.78 (d, J=11.4 Hz, 1 H), 3.72 (s, 3 H), 3.19 (m, 4 H), 3.06 (m, 4 H); calculated mass for C25H26F3N7O5S: 593.2. found: 594.2 (M+H+).


Example 19
Compound I-28 (4-(4-(4-(3-acrylamidophenylamino)-5-(trifluoromethyl)pyrimidin-2-ylamino)-3-methoxyphenyl)-N-methylpiperazine-1-carboxamide)



embedded image


A mixture of the intermediate 1 (16 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and N-methyl-N-hydroxysuccinyl carbamate (50 mg) at 0° C. The reaction mixture was stirred room temperature overnight. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C27H29F3N8O3: 570.2. found: 571.2 (M+H+).


Example 20
Compound I-29 (methyl 4-(4-(4-(3-acrylamidophenylamino)-5-(trifluoromethyl)pyrimidin-2-ylamino)-3-methoxyphenyl)piperazine-1-carboxylate)



embedded image


A mixture of the intermediate 1 (16 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and methyl chloroformate (20 uL) at 0° C. The reaction mixture was stirred room temperature for 10 min. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C27H28F3N7O4: 571.2. found: 572.2 (M+H+).


Example 21
Compound I-17 (N-(3-(2-(2-methoxy-4-(4-(methylsulfonyl)piperazin-1-yl)phenylamino)-5-(trifluoromethyl)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


A mixture of the intermediate 1 (16 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and methanesulfonyl chloride (20 uL) at 0° C. The reaction mixture was stirred at 0° C. for 10 min. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.22 (s, 1 H), 9.28 (br, 1 H), 8.72 (br, 1 H), 8.37 (br, 1 H), 7.77 (s, 1 H), 7.52 (d, J=8.0 Hz, 1 H), 7.41 (d, J=8.0 Hz, 1 H), 7.30 (t, J=8.0 Hz, 1 H), 7.12 (br, 1 H), 6.62 (s, 1 H), 6.43 (dd, J=10.0, 16.8 Hz, 1 H), 6.24 (d, J=16.8 Hz, 1 H), 6.22 (br, 1 H), 5.76 (d, J=10.0 Hz, 1 H), 3.77 (s, 3 H), 3.21 (m, 4 H), 3.18 (m, 4 H), 2.92 (s, 3 H); calculated mass for C26H28F3N7O4S: 591.2. found: 592.2 (M+H+).


Example 22
Compound I-19 (N-(3-(2-(2-methoxy-4-(4-(2,2,2-trifluoroacetyl)-1,4-diazepan-1-yl)phenylamino)-5-(trifluoromethyl)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


A mixture of the intermediate 1 (16 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)-1,4-diazepane-1-carboxylate (21 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and trifluoroacetic anhydride (10 uL) at 0° C. The reaction mixture was stirred at 0° C. for 10 min. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.2 (S, 1 H), 9.1 (br, 1 H), 8.6 (br, 1 H), 8.3 (br, 1 H), 7.75 (br, 1 H), 7.50 (d, J=8.7 Hz, 1 H), 7.26 (m, 2 H), 7.11 (m, 1 H), 6.42 (dd, J=10.1, 17.0 Hz, 1 H), 6.34 (m, 1 H), 6.23 (d, J=17.0 Hz, 1 H), 5.74 (dd, J=1.8, 10.1 Hz, 1 H), 3.3-3.8 (m, 8 H), 1.88 (m, 2 H); calculated mass for C28H27F6N7O3: 623.2. found: 624.2 (M+H+).


Example 23
Compound I-20 (methyl 4-(4-(4-(3-acrylamidophenoxy)-5-(trifluoromethyl)pyrimidin-2-ylamino)-3-methoxyphenyl)piperazine-1-carboxylate)



embedded image


A mixture of the intermediate 5 (20 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and methyl chloroformate (20 uL) at 0° C. The reaction mixture was stirred room temperature for 10 min. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.30 (s, 1 H), 8.80 (s, 1 H), 8.53 (br, 1 H), 7.60 (s, 1 H), 7.51 (d, J=8.2 Hz, 1H), 7.40 (t, J=8.2 Hz, 1 H), 7.14 (d, J=9.6 Hz, 1 H), 6.93 (m, 1 H), 6.55 (m, 1 H), 6.40 (dd, J=10.0, 16.8 Hz, 1 H), 6.24 (dd, J=1.8, 16.8 Hz, 1 H), 5.76 (dd, J=1.8, 10.0 Hz, 1 H), 3.70 (s, 3H), 3.58 (s, 3 H), 3.49 (m, 4 H), 3.05 (m, 4 H); calculated mass for C27H27F3N6O5: 572.2. found: 573.2 (M+H+).


Example 24
Compound I-21 (4-(4-(4-(3-acrylamidophenoxy)-5-(trifluoromethyl)pyrimidin-2-ylamino)-3-methoxyphenyl)-N-methylpiperazine-1-carboxamide)



embedded image


A mixture of the intermediate 5 (18 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and N-methyl-N-hydroxysuccinyl carbamate (50 mg) at 0° C. The reaction mixture was stirred room temperature overnight. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C27H28F3N7O4: 571.2. found: 572.2 (M+H+).


Example 25
Compound I-22 (N-(3-(2-(2-methoxy-4-(4-(methylsulfonyl)piperazin-1-yl)phenylamino)-5-(trifluoromethyl)pyrimidin-4-yloxy)phenyl)acrylamide)



embedded image


A mixture of the intermediate 5 (16 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and methanesulfonyl chloride (10 uL) at 0° C. The reaction mixture was stirred at 0° C. for 10 min. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C26H27F3N6O5S: 592.2. found: 593.2 (M+H+).


Example 26
Compound I-23 (N-(3-(5-chloro-2-(4-(4-(3,3-dimethylbutanoyl)piperazin-1-yl)-2-methoxyphenylamino)pyrimidin-4-yloxy)phenyl)acrylamide)



embedded image


A mixture of the intermediate 6 (16 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in n-butanol (1.0 mL) with catalytic trifluoroacetic acid was microwaved for 20 min at 100° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and 3,3-dimethylbutyryl chloride at 0° C. The reaction mixture was stirred at 0° C. for 10 min. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.33 (s, 1 H), 8.35 (s, 1 H), 8.15 (s, 1 H), 7.60 (s, 1 H), 7.55 (d, J=8.2 Hz, 1 H), 7.40 (t, J=8.2 Hz, 1 H), 7.24 (d, J=8.7 Hz, 1 H), 6.95 (d, J=7.8 Hz, 1 H), 6.57 (s, 1 H), 6.42 (dd, J=10.0, 16.8 Hz, 1 H), 6.25 (d, J=16.8 Hz, 1 H), 6.18 (m, 1 H), 6.77 (d, J=10.0 Hz, 1 H), 3.93 (s, 3 H), 3.68 (m, 4 H), 2.99 (m, 4 H), 2.26 (s, 2 H), 0.99 (s, 9 H); calculated mass for C30H35ClN6O4: 578.2. found: 579.2 (M+H+).


Example 27
Compound I-24 (N-(3-(5-chloro-2-(2-methoxy-4-(4-(2,2,2-trifluoroacetyl)piperazin-1-yl)phenylamino)pyrimidin-4-yloxy)phenyl)acrylamide)



embedded image


A mixture of the intermediate 6 (20 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in n-butanol (1.0 mL) with catalytic trifluoroacetic acid was microwaved for 20 min at 100° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and trifluoroacetic anhydride (10 uL) at 0° C. The reaction mixture was stirred at 0° C. for 10 min. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.32 (s, 1 H), 8.35 (s, 1 H), 8.16 (s, 1 H), 7.59 (s, 1 H), 7.55 (d, J=8.2 Hz, 1 H), 7.40 (t, J=8.2 Hz, 1 H), 7.24 (d, J=8.7 Hz, 1 H), 6.96 (d, J=7.8 Hz, 1 H), 6.57 (d, J=2.8 Hz, 1 H), 6.41 (dd, J=10.0, 16.8 Hz, 1 H), 6.24 (dd, J=1.8, 16.8 Hz, 1 H), 6.19 (m, 1 H), 5.76 (dd, J=1.8, 10.0 Hz, 1 H), 3.72 (s, 3 H), 3.68 (m, 4 H), 3.13 (m, 4 H); calculated mass for C26H24ClF3N6O4: 576.2. found: 577.0 (M+H+).


Example 28
Compound I-25 (4-(4-(4-(3-acrylamidophenoxy)-5-chloropyrimidin-2-ylamino)-3-methoxyphenyl)-N-tert-butylpiperazine-1-carboxamide)



embedded image


A mixture of the intermediate 6 (20 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in n-butanol (1.0 mL) with catalytic trifluoroacetic acid was microwaved for 20 min at 100° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and tert-butyl isocyanate at 0° C. The reaction mixture was stirred at 0° C. for 10 min. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.35 (s, 1 H), 8.37 (s, 1 H), 8.19 (s, 1 H), 7.62 (s, 1 H), 7.56 (d, J=8.2 Hz, 1 H), 7.41 (t, J=8.2 Hz, 1 H), 7.28 (d, J=8.2 Hz, 1 H), 6.96 (d, J=8.2 Hz, 1 H), 6.65 (s, 1 H), 6.43 (dd, J=10.0, 16.8 Hz, 1 H), 6.29 (s, 1 H), 6.26 (d, J=16.8 Hz, 1 H), 5.92 (s, 1 H), 5.77 (d, J=10.0 Hz, 1 H), 3.74 (s, 3 H), 3.41 (s, 4 H), 3.04 (s, 4 H), 1.26 (s, 9 H); calculated mass for C29H34ClN7O4: 579.2. found: 580.2 (M+H+).


Example 29
Compound I-26 (N-(3-(5-chloro-2-(2-methoxy-4-(4-(methylsulfonyl)piperazin-1-yl)phenylamino)pyrimidin-4-yloxy)phenyl)acrylamide)



embedded image


A mixture of the intermediate 6 (20 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in n-butanol (1.0 mL) with catalytic trifluoroacetic acid was microwaved for 20 min at 100° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and methanesulfonyl chloride at 0° C. The reaction mixture was stirred at 0° C. for 10 min. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C25H27ClN6O5S: 558.2. found: 559.2 (M+H+).


Example 30
Compound I-18 (tert-butyl 4-(4-(4-(3-acrylamidophenoxy)-5-chloropyrimidin-2-ylamino)-3-ethoxyphenyl)piperazine-1-carboxylate)



embedded image


A mixture of the intermediate 6 (16 mg) and tert-butyl 4-(4-amino-3-ethoxyphenyl)piperazine-1-carboxylate (21 mg) in n-butanol (1.0 mL) with catalytic trifluoroacetic acid was microwaved for 20 min at 100° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C30H35ClN6O5: 594.2. found: 595.5 (M+H+).


Example 31
Compound I-30 (N-(3-(2-(4-(4-(2-hydroxyacetyl)piperazin-1-yl)-2-(trifluoromethoxy)phenylamino)-5-(trifluoromethyl)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


A mixture of the intermediate 1 (16 mg) and tert-butyl 4-(4-amino-3-trifluoromethoxyphenyl)piperazine-1-carboxylate (22 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), N,N-dimethylformamide (1.0 mL), HATU, and glycolic acid at 0° C. The reaction mixture was stirred at room temperature for 30 min. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.14 (s, 1 H), 8.99 (s, 1 H), 8.61 (s, 1 H), 8.29 (s, 1 H), 7.71 (s, 1 H), 7.44 (s, 1 H), 7.42 (s, 1 H), 7.18 (m, 2 H), 6.85 (s, 1 H), 6.75 (m, 1 H), 6.45 (dd, J=10.0, 16.8 Hz, 1 H), 6.26 (d, J=16.8 Hz, 1 H), 5.77 (d, J=10.0 Hz, 1 H), 4.66 (t, J=5.6 Hz, 1 H), 4.14 (d, J=5.6 Hz, 2 H), 3.61 (br, 2 H), 3.49 (br, 2 H), 3.11 (br, 4 H); calculated mass for C27H25F6N7O4: 625.2. found: 625.8 (M+H+).


Example 32
Compound I-31 (N-(3-(2-(2-(difluoromethoxy)-4-(4-(2-hydroxyacetyl)piperazin-1-yl)phenylamino)-5-(trifluoromethyl)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


A mixture of the intermediate 1 (16 mg) and tert-butyl 4-(4-amino-3-difluoromethoxyphenyl)piperazine-1-carboxylate (22 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), N,N-dimethylformamide (1.0 mL), HATU, and glycolic acid at 0° C. The reaction mixture was stirred at room temperature for 30 min. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C27H26F5N7O4: 607.2. found: 607.8 (M+H+).


Example 33
Compound I-1 (N-(3-(2-(2-methoxy-4-morpholinophenylamino)-5-(trifluoromethyl)pyrimidin-4-yloxy)phenyl)acrylamide)



embedded image


A mixture of the intermediate 5 (16 mg) and 2-methoxy-4-morpholinoaniline (20 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.35 (S, 1 H), 8.83 (S, 1 H), 8.55 (br, 1 H), 7.63 (S, 1 H), 7.53 (d, J=8.0 Hz, 1 H), 7.40 (m, 1 H), 7.16 (d, J=8.8 Hz, 1 H), 6.96 (br, 1 H), 6.54 (br, 1 H), 6.43 (m, 1 H), 6.27 (dd, J=1.7, 16.8 Hz, 1 H), 5.77 (dd, J=1.7, 10.4 Hz, 1 H), 3.72 (br, 7 H), 3.04 (br, 4 H); calculated mass for C25H24F3N5O4: 515.2. found: 516.7 (M+H+).


Example 34
Compound I-13 (N-(3-(2-(2-(difluoromethoxy)-4-morpholinophenylamino)-5-(trifluoromethyl)pyrimidin-4-yloxy)phenyl)acrylamide)



embedded image


A mixture of the intermediate 5 (16 mg) and 2-difluoromethoxy-4-morpholinoaniline (22 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.33 (s, 1 H), 9.34 (s, 1 H), 8.55 (br, 1 H), 7.63 (s, 1 H), 7.51 (br, 1 H), 7.40 (br, 1 H), 7.19 (d, J=9.2 Hz, 1 H), 6.96 (br, 1H), 6.65 (br, 1 H), 6.43 (dd, J=10.0, 16.8 Hz, 1 H), 6.27 (dd, J=2.0, 16.8 Hz, 1 H), 5.79 (dd, J=2.0, 10.0 Hz, 1 H), 3.73 (t, J=4.4 Hz, 1 H), 3.06 (m, 4 H); calculated mass for C25H22F5N5O4: 551.2. found: 551.7 (M+H+).


Example 35
Compound I-14 (N-(3-(2-(2-(difluoromethoxy)-4-morpholinophenylamino)-5-(trifluoromethyl)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


A mixture of the intermediate 1 (16 mg) and 2-difluoromethoxy-4-morpholinoaniline (22 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.14 (s, 1 H), 8.63 (s, 1 H), 8.28 (s, 1 H), 7.71 (s, 1 H), 7.46 (d, J=7.2 Hz, 1 H), 7.38 (s, 1 H), 7.15 (m, 2 H), 6.91 (m, 1 H), 6.67 (m, 2 H), 6.44 (dd, J=10.0, 16.8 Hz, 1 H), 6.25 (d, J=16.8 Hz, 1 H), 5.76 (d, J=10.0 Hz, 1 H), 3.73 (t, J=4.4 Hz, 1 H), 3.05 (br, 4 H); calculated mass for C25H23F5N6O3: 550.2. found: 550.9 (M+H+).


Example 36
Compound I-15 (N-(3-(5-chloro-2-(2-methoxy-4-morpholinophenylamino)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


A mixture of the intermediate 2 (16 mg) and 2-difluoromethoxy-4-morpholinoaniline (20 mg) in n-butanol (1.0 mL) with catalytic trifluoroacetic acid was microwaved for 20 min at 150° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ C10.16 (s, 1 H), 8.85 (s, 1 H), 8.07 (s, 1 H), 7.95 (br, 1 H), 7.68 (m, 2 H), 7.45 (d, J=7.6 Hz, 1 H), 7.27 (m, 2 H), 6.60 (d, J=2.4 Hz, 1 H), 6.45 (dd, J=10.0, 16.8 Hz, 1 H), 6.26 (m, 2 H), 5.76 (dd, J=2.0, 10.0 Hz, 1 H), 3.79 (s, 3 H), 3.73 (m, 4 H), 3.03 (m, 4 H); calculated mass for C24H25ClN6O3: 480.2. found: 481.4 (M+H).


Example 37
Compound I-32 (N-(3-(2-(4-(4-acetylpiperazin-1-yl)-2-(difluoromethoxy)phenylamino)-5-(trifluoromethyl)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


A mixture of the intermediate 1 (16 mg) and tert-butyl 4-(4-amino-3-difluoromethoxyphenyl)piperazine-1-carboxylate (22 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and acetic anhydride (50 uL). The reaction mixture was stirred room temperature overnight. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.13 (s, 1 H), 8.63 (s, 2 H), 8.29 (s, 1 H), 7.71 (s, 1 H), 7.47 (d, J=7.2 Hz, 1 H), 7.39 (m, 1 H), 7.16 (m, 2 H), 6.91 (s, 1 H), 6.69 (s, 1 H), 9.59 (m, 1 H), 6.44 (dd, J=10.0, 16.8 Hz, 1 H), 6.26 (d, J=16.8 Hz, 1 H), 5.77 (d, J=10.0 Hz, 1 H), 3.57 (s, 4 H), 3.10 (s, 2 H), 3.04 (s, 2 H), 2.05 (s, 3 H); calculated mass for C27H26F5N7O3: 591.2. found: 591.8 (M+H+).


Example 38
Compound I-33 (N-(3-(2-(4-(4-acetylpiperazin-1-yl)-2-(trifluoromethoxy)phenylamino)-5-(trifluoromethyl)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


A mixture of the intermediate 1 (16 mg) and tert-butyl 4-(4-amino-3-trifluoromethoxyphenyl)piperazine-1-carboxylate (22 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and acetic anhydride (30 uL). The reaction mixture was stirred room temperature overnight. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.15 (s, 1 H), 9.00 (s, 1 H), 8.62 (s, 1 H), 8.29 (s, 1 H), 7.71 (s, 1 H), 7.43 (s, 2 H), 7.18 (m, 2 H), 6.84 (s, 1 H), 6.75 (br, 1 H), 6.45 (dd, J=10.0, 16.8 Hz, 1 H), 6.26 (d, J=16.8 Hz, 1 H), 5.77 (d, J=10.0 Hz, 1 H), 3.57 (br, 4 H), 3.13 (br, 2 H), 3.06 (br, 2 H), 2.05 (s, 3 H); calculated mass for C27H25F6N7O3: 609.2. found: 610.0 (M+H+).


Example 39
Compound I-34 (N-(3-(2-(2-methoxy-4-(4-pivaloylpiperazin-1-yl)phenylamino)-5-(trifluoromethyl)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


A mixture of the intermediate 1 (16 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and pivaloyl chloride (20 uL) at 0° C. The reaction mixture was stirred room temperature for 10 min. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C30H34F3N7O3: 597.3. found: 598.3 (M+H+).


Example 40
Compound I-35 (N-(3-(2-(2-methoxy-4-(4-propionylpiperazin-1-yl)phenylamino)-5-(trifluoromethyl)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


A mixture of the intermediate 1 (16 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and propionyl chloride (10 uL) at 0° C. The reaction mixture was stirred room temperature for 10 min. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C27H28F3N7O3: 555.2. found: 556.2 (M+H+).


Example 41
Compound I-36 (N-(3-(5-chloro-2-(2-(difluoromethoxy)-4-morpholinophenylamino)pyrimidin-4-yloxy)phenyl)acrylamide)



embedded image


A mixture of the intermediate 6 (20 mg) and 2-difluoromethoxy-4-morpholinoaniline (22 mg) in n-butanol (1.0 mL) with catalytic trifluoroacetic acid was microwaved for 20 min at 150° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.33 (s, 1 H), 8.71 (s, 1 H), 8.35 (s, 1 H), 7.62 (t, J=2.0 Hz, 1 H), 7.53 (d, J=8.0 Hz, 1 H), 7.40 (t, J=8.0 Hz, 1 H), 7.23 (d, J=8.8 Hz, 1 H), 6.97 (dd, J=64.8, 66.4 Hz, 1 H), 6.95 (s, 1 H), 6.64 (d, J=2.0 Hz, 1 H), 6.59 (br, 1 H), 6.44 (dd, J=10.0, 16.8 Hz, 1 H), 6.27 (dd, J=2.0, 16.8 Hz, 1 H), 5.79 (dd, J=2.0, 10.0 Hz, 1 H), 3.72 (m, 4 H), 3.03 (m, 4 H); calculated mass for C24H22ClF2N5O4: 517.1. found: 517.7 (M+H+).


Example 42
Compound I-37 (N-(3-(5-chloro-2-(2-methoxy-4-(1,4-oxazepan-4-yl)phenylamino)pyrimidin-4-yloxy)phenyl)acrylamide)



embedded image


A mixture of the intermediate 6 (20 mg) and 2-methoxy-4-(1,4-oxazepan-4-yl)aniline (21 mg) in n-butanol (1.0 mL) with catalytic trifluoroacetic acid was microwaved for 20 min at 150° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C25H26ClN5O4: 495.2. found: 495.8 (M+H+).


Example 43
Compound I-38 (N-(3-(2-(2-methoxy-4-(1,4-oxazepan-4-yl)phenylamino)-5-(trifluoromethyl)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


A mixture of the intermediate 1 (16 mg) and 2-methoxy-4-(1,4-oxazepan-4-yl)aniline (21 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C26H27F3N6O3: 528.2. found: 528.8 (M+H+).


Example 44
Compound I-39 (tert-butyl 4-(4-(4-(3-acrylamidophenoxy)-5-(trifluoromethyl)pyrimidin-2-ylamino)-3-methoxyphenyl)-1,4-diazepane-1-carboxylate)



embedded image


A mixture of the intermediate 5 (16 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)-1,4-diazepane-1-carboxylate (21 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C26H27F3N6O3: 528.2. found: 528.8 (M-Boc+H+).


Example 45
Compound I-40 (N-(3-(2-(4-(4-acetylpiperazin-1-yl)-2-methoxyphenylamino)-5-bromopyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


A mixture of intermediate 4 and 1-(4-(4-amino-3-methoxyphenyl)piperazin-1-yl)ethanone (18 mg) in n-butanol (1 mL) with catalytic HCl was microwaved for 20 min at 150° C. The crude was concentrated under reduced pressure and purified to give the title compound. 1H-NMR (DMSO-d6, 400 MHz) δ 10.15 (s, 1 H), 8.60 (br, 1 H), 8.15 (s, 1 H), 7.89 (br, 1 H), 7.67 (m, 2 H), 7.47 (d, J=6.8 Hz, 1 H), 7.26 (m, 2 H), 6.63 (d, J=2.4 Hz, 1 H), 6.45 (dd, J=10.0, 16.8 Hz, 1 H), 6.28 (m, 1 H), 6.26 (dd, J=2.0, 16.8 Hz, 1 H), 5.76 (dd, J=2.0, 10.0 Hz, 1H), 3.79 (s, 3 H), 3.56 (m, 4 H), 3.06 (m, 2 H), 3.03 (m, 2 H), 2.05 (s, 3 H); calculated mass for C26H28BrN7O3: 565.1. found: 566.3 (M+H+).


Example 46
Compound I-41 (N-(3-(5-chloro-2-(2-methoxy-4-(4-(methylsulfonyl)piperazin-1-yl)phenylamino)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


A mixture of the intermediate 2 (16 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in n-butanol (1.0 mL) with catalytic trifluoroacetic acid was microwaved for 20 min at 100° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and methanesulfonyl chloride (10 uL) at 0° C. The reaction mixture was stirred at 0° C. for 10 min. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.16 (s, 1 H), 8.86 (s, 1 H), 8.08 (s, 1 H), 7.95 (s, 1 H), 7.70 (m, 2 H), 7.44 (d, J=7.6 Hz, 1 H), 7.28 (m, 2 H), 6.64 (d, J=2.4 Hz, 1 H), 6.46 (dd, J=10.0, 16.8 Hz, 1 H), 6.32 (m, 1 H), 6.26 (dd, J=2.0, 16.8 Hz, 1 H), 5.77 (dd, J=2.0, 10.0 Hz, 1 H), 3.80 (s, 3 H), 3.30 (m, 4H), 3.25 (m, 2 H), 3.17 (m, 2 H), 2.93 (s, 3 H); calculated mass for C25H28ClN7O4S: 557.2. found: 558.4 (M+H+).


Example 47
Compound I-42 (tert-butyl 4-(4-(4-(3-acrylamidophenoxy)-5-(trifluoromethyl)pyrimidin-2-ylamino)-3-methoxyphenyl)piperazine-1-carboxylate)



embedded image


A mixture of the intermediate 5 (16 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C30H33F3N6O5: 614.3. found: 615.2 (M+H+).


Example 48
Compound I-43 (tert-butyl 4-(4-(4-(3-acrylamidophenoxy)-5-chloropyrimidin-2-ylamino)-3-methoxyphenyl)-1,4-diazepane-1-carboxylate)



embedded image


A mixture of the intermediate 6 (16 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)-1,4-diazepane-1-carboxylate (21 mg) in n-butanol (1 mL) with catalytic HCl was microwaved for 20 min at 100° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C30H35ClN6O5: 594.2. found: 594.8 (M+H+).


Example 49
Compound I-44 (tert-butyl 4-(4-(4-(3-acrylamidophenylamino)-5-(trifluoromethyl)pyrimidin-2-ylamino)-3-methoxyphenyl)-1,4-diazepane-1-carboxylate)



embedded image


A mixture of the intermediate 1 (16 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)-1,4-diazepane-1-carboxylate (21 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C31H36F3N7O4: 627.3. found: 628.0 (M+H+).


Example 50
Compound I-45 (tert-butyl 4-(4-(4-(3-acrylamidophenylamino)-5-(trifluoromethyl)pyrimidin-2-ylamino)-3-methoxyphenyl)piperazine-1-carboxylate)



embedded image


A mixture of the intermediate 1 (16 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C30H34F3N7O4: 613.3. found: 614.1 (M+H+).


Example 51
Compound I-46 (tert-butyl 4-(4-(4-(3-acrylamidophenoxy)-5-chloropyrimidin-2-ylamino)-3-methoxyphenyl)piperazine-1-carboxylate)



embedded image


A mixture of the intermediate 6 (16 mg) and tert-butyl 4-(4-amino-3-methoxyphenyl)piperazine-1-carboxylate (20 mg) in n-butanol (1 mL) with catalytic HCl was microwaved for 20 min at 120° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C29H33ClN6O5: 580.2. found: 581.2 (M+H+).


Example 52
Compound I-47 (tert-butyl 4-(4-(4-(3-acrylamidophenoxy)-5-chloropyrimidin-2-ylamino)-3-(trifluoromethoxy)phenyl)piperazine-1-carboxylate)



embedded image


A mixture of the intermediate 6 (16 mg) and tert-butyl 4-(4-amino-3-trifluoromethoxyphenyl)piperazine-1-carboxylate (22 mg) in n-butanol (1.0 mL) with catalytic HCl was microwaved for 20 min at 120° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C29H30ClF3N6O5: 634.2. found: 635.4 (M+H+).


Example 53
Compound I-48 ((S)-methyl 1-acetyl-4-(4-(4-(3-acrylamidophenylamino)-5-(trifluoromethyl)pyrimidin-2-ylamino)-3-methoxyphenyl)piperazine-2-carboxylate)



embedded image


A mixture of the intermediate 1 (16 mg) and (S)-1-tert-butyl 2-methyl 4-(4-amino-3-methoxyphenyl)piperazine-1,2-dicarboxylate (23 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier). The intermediate was dissolved in dichloromethane (1.0 mL) and treated with TFA (0.3 mL). After 10 minutes, the mixture was concentrated under reduced pressure. To the residue were added N,N-diethylisopropylamine (20 uL), dichloromethane (1.0 mL), and acetic anhydride (20 uL). The reaction mixture was stirred room temperature overnight. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C29H30F3N7O5: 613.2. found: 614.2 (M+H+).


Example 54
Compound I-49 (N-(3-(2-(2-methoxy-4,4-dioxo-4-thiomorpholinophenylamino)-5-(trifluoromethyl)pyrimidin-4-ylamino)phenyl)acrylamide)



embedded image


A mixture of the intermediate 1 (18 mg) and 2-methoxy-4-S,S-dioxothiomorpholino-aniline (24 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. 1H-NMR (DMSO-d6, 400 MHz) δ 10.16 (s, 1 H), 8.66 (br, 1 H), 8.29 (s, 1 H), 8.13 (s, 1 H), 7.77 (br, 1 H), 7.51 (s, 1 H), 7.49 (s, 1 H), 7.26 (t, J=8.0 Hz, 1 H), 7.18 (br, 1 H), 6.64 (d, J=2.0 Hz, 1 H), 6.44 (dd, J=10.0, 16.8 Hz, 1 H), 6.31 (m, 1 H), 6.26 (dd, J=2.0, 16.8 Hz, 1 H), 5.77 (dd, J=2.0, 10.0 Hz, 1 H), 3.79 (s, 3 H), 3.70 (m, 4H), 3.12 (m, 4 H); calculated mass for C25H25F3N6O5S: 562.2. found: 562.8 (M+H+).


Example 55
Compound I-50 ((S)-1-tert-butyl 2-methyl 4-(4-(4-(3-acrylamidophenylamino)-5-(trifluoromethyl)pyrimidin-2-ylamino)-3-methoxyphenyl)piperazine-L2-dicarboxylate)



embedded image


A mixture of the intermediate 1 (20 mg) and (S)-1-tert-butyl 2-methyl 4-(4-amino-3-methoxyphenyl)piperazine-1,2-dicarboxylate (26 mg) in dioxane (1.0 mL) with catalytic trifluoroacetic acid was stirred overnight at 50° C. The crude was concentrated under reduced pressure and purified using HPLC (TFA modifier) to give the desired as a TFA salt. Calculated mass for C32H36F3N7O6: 671.3. found: 672.3 (M+H+).


Biological Examples


Described below are assays used to measure the biological activity of provided compounds as selective inhibitors of mutant EGFR as compared to WT EGFR (and other protein kinases).


Example 56
Omnia Assay Protocol for Potency Assessment Against EGFR (WT) and EGFR (T790M/L858R) Active Enzymes

Below describes the biochemical assay protocol using EGFR-WT and EGFR-T790M/L858R.


The mechanics of the assay platform are best described by the vendor (Invitrogen, Carlsbad, Calif.) on their website at the following URL: www.invitrogen.com/content.cfm?pageid=11338 or www.invitrogen.com/site/us/en/home/Products-and-Services/Applications/Drug-Discovery/Target-and-Lead-Identification-and-Validation/KinaseBiology/KB-Misc/Biochemical-Assays/Omnia-Kinase-Assays.html.


Briefly, 10× stocks of EGFR-WT (PV3872) from Invitrogen and EGFR-T790M/L858R (40350) from BPS Bioscience, San Diego, Calif., 1.13×ATP (AS001A) and appropriate Tyr-Sox conjugated peptide substrates (KCZ1001) were prepared in 1× kinase reaction buffer consisting of 20 mM Tris, pH 7.5, 5 mM MgCl2, 1 mM EGTA, 5 mM β-glycerophosphate, 5% glycerol (10× stock, KB002A) and 0.2 mM DTT (DS001A). 5 μL of each enzyme were pre-incubated in a Corning (#3574) 384-well, white, non-binding surface microtiter plate (Corning, N.Y.) for 30 min. at 25° C. with a 0.5 μL volume of 50% DMSO and serially diluted compounds prepared in 50% DMSO. Kinase reactions were started with the addition of 45 μL of the ATP/Tyr-Sox peptide substrate mix and monitored every 71 seconds for 60 minutes at λex360/λem485 in a Synergy4 plate reader from BioTek (Winooski, Vt.). At the conclusion of each assay, progress curves from each well were examined for linear reaction kinetics and fit statistics (R2, 95% confidence interval, absolute sum of squares). Initial velocity (0 minutes to ˜30 minutes) from each reaction was determined from the slope of a plot of relative fluorescence units vs time (minutes) and then plotted against inhibitor concentration to estimate IC50 from log [Inhibitor] vs Response, Variable Slope model in GraphPad Prism from GraphPad Software (San Diego, Calif.).


[EGFR-WT]=5 nM, [ATP]=15 uM, [Y12-Sox]=5 uM (ATP KMapp˜12 uM); and [EGFR-T790M/L858R]=2.5 nM, [ATP]=20 uM, [Y12-Sox]=5 uM (ATP KMapp˜20 uM).


Table 3 shows the activity of selected compounds of this invention in the EGFR inhibition assay described above. Table 3 shows mutant EGFR data as compared to WT EGFR and provides the selectivity ratio of WT to mutant for each test compound. The compound numbers correspond to the compound numbers in Table 1.









TABLE 3







EGFR (Mutant and Wild Type) Biochemical Inhibition Data














EGFR





WT EGFR
(T790M/L858R)
Ratio



Compound #
IC50 (nM)
IC50 (nM)
WT/mutant
















I-1
30-100
1-10
>40



I-2
10-30 
<1
>20



I-3
1-10
<1
>5



I-4
1-10
<1
>10



I-5
10-30 
1-10
>25



I-6
1-10
<1
>5



I-7
10-30 
<1
>15



I-8
10-30 
1-10
>10



I-9
1-10
<1
>5



I-10
1-10
1-10
>1



I-11
10-30 
<1
>25



I-12
10-30 
<1
>15



I-13
30-100
<1
>35



I-14
10-30 
<1
>25



I-15
30-100
1-10
>15



I-17
10-30 
<1
>30



I-18
>1000
10-30 
>50



I-19
100-300
1-10
>50



I-20
10-30 
1-10
>5



I-21
10-30 
<1
>35



I-22
30-100
<1
>50



I-23
100-300 
1-10
>25



I-24
30-100
1-10
>15



I-26
10-30 
<1
>25



I-27
1-10
1-10
>5



I-28
1-10
<1
>10



I-29
10-30 
<1
>30



I-30
30-100
<1
>50



I-31
  <1
<1
1



I-32
  <1
<1
>1



I-33
1-10
<1
>10



I-34
30-100
<1
>40



I-35
1-10
<1
>10



I-36
10-30 
<1
>25



I-37
30-100
1-10
>25



I-38
10-30 
<1
>50



I-39
>1000
10-30 
>50



I-40
10-30 
1-10
>20



I-41
30-100
1-10
>20



I-42
300-1000
300-1000
>1



I-43
300-1000
1-10
>50



I-44
300-1000
1-10
>50



I-45
30-100
1-10
>20



I-46
100-300 
<1
>50



I-47
>1000
10-30 
>50



I-48
10-30 
<1
>25



I-49
1-10
<1
>1










Example 57

Cell Culture and Antibodies


A431 human epidermoid carcinoma, H1975 human NSCLC and HCC827 human NSCLC adenocarcinoma cells were obtained from the American Type Culture Center (Manassas, Va.). A431 cells were grown in DMEM (Invitrogen, Carlsbad, Calif.) supplemented with 10% FBS (HyClone, South Logan, Utah) and 1% Penicillin-Streptomycin (P/S, Lonza, Walkersville, Md.). H1975 and HCC827 cells were grown in complete RPMI 1640 (Invitrogen) supplemented with 10% FBS and 1% P/S. All cells were maintained and propagated as monolayer cultures at 37° C. in a humidified 5% CO2 incubator.


All primary antibodies were obtained from Cell Signaling (Danvers, Mass.) and used at 1:1000. Secondary antibodies were used at 1:10,000. Goat anti-mouse IgG IRDye 800CW antibody was obtained from LiCor Biosciences (Lincoln, Nebr.) and goat anti-rabbit IgG Alexa Fluor 680 was obtained from Invitrogen.


Immunoblotting


Cells were grown in 12-well plates (Corning, Coring, N.Y.) to 90% confluence and then incubated in low-serum (0.1% FBS) media for 16-18 hr. Cells were then treated with 5, 1.25, 0.31, 0.078, 0.020 or 0.005 μM test compound in low-serum (0.1% FBS) media for 1 hr. A431 cells were then stimulated with 50 ng/ml EGF (Peprotech, Rocky Hill, N.J.) for 15 min. After treatment, cell monolayers were washed with cold PBS (Invitrogen) and immediately lysed by scrapping into 60 uL cold Cell Extraction Buffer (Invitrogen) supplemented with Complete Protease inhibitors (Roche, Indianapolis, Ind.) and PhosphoSTOP (Roche) phosphatase inhibitors.


Lysate protein concentrations were determined by BCA Assay (Pierce, Rockford, Ill.) and 50 ug of each lysate was separated by 4-12% gradient SDS-PAGE (Invitrogen), transferred to nitrocellulose membrane (Biorad, Hercules, Calif.) and probed with specific antibodies. Phospho-protein signals were quantitated using Odyssey Infrared Imaging (Li-Cor Biosciences).


To assess phosphor-EGFR signaling, blots were probed with rabbit anti-Phospho-EGFR (Y1068) and mouse total anti-EGFR antibodies. Phospho-EGFR signal was normalized to total EGFR expression for each sample. Results are indicated as % DMSO control. Normalized data was fitted using a sigmoidal curve analysis program (Graph Pad Prism version 5) with variable Hill slope to determine the EC50 values.


Table 4 shows mutant EGFR data in H1975 (double mutation L858R/T790M) and HCC827 (delE746-A750 deletion mutation) cells as compared to WT EGFR (A431 cells). The compound numbers recited in Table 4 correspond to the compound numbers in Table 1.









TABLE 4







EGFR (Mutant and Wild Type) Signaling (1 hr)













WT EGFR
H1975
Ratio WT/
HCC827
Ratio WT/


Compound #
EC50 (nM)
EC50 (nM)
mutant
EC50 (nM)
mutant















I-1
>1000
10-100
>50




I-2
>1000
10-100
>50




I-3
>1000
500-1000
>5




I-4
>1000
10-100
>50
100-500
>25


I-5
>1000
10-100
>30




I-6
>1000
10-100
>20




I-7
>1000
100-500 
>5




I-8
>1000
>1000
>1




I-9
>1000






I-11
>1000
100-500 
>15




I-12
>1000
>1000
>1




I-13

10-100





I-14
500-1000
10-100
>15
 10-100
>15


I-17
>1000
10-100
>30
100-500
 >5


I-21
>1000
100-500 
>5




I-22
>1000
100-500 
>35




I-26
>1000
100-500 
>40




I-27
>1000






I-29
>1000






I-33
>1000
10-100
>50




I-34
>1000
100-500 
>35




I-35
>1000
 <10
>50




I-40
>1000
>1000
>1




I-41
>1000
>1000
>1




I-44
>1000
500-1000
>5




I-45
>1000
100-500 
>10




I-46
>1000
100-500 
>10
<10
>50


I-48
>1000
10-100
>50




I-49
>1000
10-100
>20











Example 58

Cell Proliferation


Cells were plated in Growth Media supplemented with 5% FBS and 1% P/S at a density of 3,000 cells per well in 96 well tissue culture plates (Corning). Cells were allowed to settle down for 4 hr and then treated with 5, 1.25, 0.31, 0.078, 0.020 or 0.005 μM test compound for 72 hr. Cell viability was determined by CellTiter Glo (Promega, Madison, Wis.) and results were converted to cell numbers using a standard curve. Growth inhibition (GI50) values were determined by Graph Pad Prism.


The result of this experiment is depicted in Table 5, where it shows mutant selective inhibition in H1975 (double mutation L858R/T790M) and HCC827 (delE746-A750 deletion mutation) cells but not in WT-EGFR A431 cells.









TABLE 5







EGFR (Mutant and Wild Type) Cell Proliferation













WT EGFR
H1975
Ratio WT/
HCC827
Ratio WT/


Compound #
GI50 (nM)
GI50 (nM)
mutant
GI50 (nM)
mutant















I-1
>1000
 10-100
>15
10-100
>15


I-2
>1000
 10-100
>20
10-100
>40


I-3
>1000
100-500
>5
10-100
>45


I-4
500-1000
 10-100
>10
10-100
>35


I-5
>1000
100-500
>5
10-100
>20


I-6
500-1000
 10-100
>10
10-100
>40


I-7
>1000
100-500
>1
100-500 
>10


I-8
500-1000
100-500
>1
10-100
>20


I-9
>1000
100-500
>5
10-100
>50


I-10
>1000
 500-1000
>1
100-500 
>20


I-11
>1000
100-500
>5
10-100
>45


I-12
500-1000
100-500
>1
10-100
>30


I-13
>1000
100-500
>10
10-100
>50


I-14
>1000
100-500
>5
10-100
>10


I-15
>1000
 500-1000
>1
100-500 
>10


I-17
>1000
100-500
>10
10-100
>15


I-18
>1000
 500-1000
>1
100-500 
>5


I-19
>1000
100-500
>5
100-500 
>10


I-20
>1000
100-500
>5
100-500 
>15


I-21
500-1000
100-500
>1
10-100
>20


I-22
>1000
100-500
>10
10-100
>35


I-23
>1000
 500-1000
>1
100-500 
>20


I-24
500-1000
100-500
>1
10-100
>40


I-26
>1000
>1000
>1
10-100
>35


I-27
>1000
100-500
>1
10-100
>10


I-28
>1000
 500-1000
>5
10-100
>50


I-29
>1000
 10-100
>10
10-100
>15


I-30
>1000
100-500
>1
10-100
>25


I-33
>1000
 10-100
>30
100-500 
>15


I-34
>1000
100-500
>5
10-100
>50


I-35
>1000
 10-100
>10
10-100
>25


I-36
>1000
100-500
>25
10-100
>50


I-37
>1000
 500-1000
>1
100-500 
>10


I-38
>1000
>1000
>1
500-1000
>5


I-39
>1000
>1000
<1
>1000
>1


I-40
500-1000
100-500
>5
10-100
>10


I-41
>1000
100-500
>1
10-100
>20


I-44
>1000
 500-1000
>5
500-1000
>5


I-45
>1000
100-500
>1
100-500 
>10


I-46
>1000
 500-1000
>1
100-500 
>10


I-48
>1000
 10-100
>10
10-100
>15


I-49
>1000
100-500
>10
10-100
>25


I-50
>1000
100-500
>10
10-100
>50









Example 59

Washout Experiment in H1975 Cells Containing EGFR Deletion/T790M Mutation


Cells were plated in Growth Media supplemented with 10% FBS and 1% P/S at a density of 2.0×10 5 cells per well in 12 well tissue culture plates. Cells were allowed to settle down for 4 hrs and then maintained in low-serum (0.1% FBS) media overnight.


The following morning the media was removed and the cells were treated with 500 nM test compound in low-serum media for 1 hr. The cells were washed free of compound 2× with PBS (Invitrogen). One set of cells were immediately lysed as indicated above as the 0 hr time point. The remaining cells were incubated with complete RPMI-1640 growth media (10% FBS) for 1, 3, 6 and 24 hr. For the first 1 hr, cells were washed 2× with PBS every 30 min. DMSO (0.5%) controls were collected at the 0, 3, 6 and 24 hr time points.


Compounds I-2 and I-4 demonstrate prolonged duration of action after compound removal. pEGFR phosphorylation is inhibited by 80-100% 1 hr after compound removal. pEGFR remained 60-90% inhibited for at least 8 hours after compound was removed, but activity was restored to 40-60% by new protein synthesis at 24 h.


Example 60

Mass Spectrometry for Mutant EGFR


Compound I-4 modifies EGFR T790M/L858R singly and completely, as confirmed by whole protein MS analysis. Intact EGFR T790M/L858R (BPS, 40350) was incubated for 60 min. at a 10-fold excess of Compound I-4 to protein. 5 μL aliquots of the samples were diluted with 15 μL of 0.2% TFA prior to micro C4 ZipTipping directly onto the MALDI target using sinapinic acid as the desorption matrix (10 mg/ml in 0.1% TFA:Acetonitrile 50:50). Panel A shows the mass spec trace of the intact EGFR T790M/L858R protein (m/z=88,389 Da). Panel B shows the mass spec trace of EGFR T790M/L858R incubated with Compound I-4 (mw=555.56) for 30 min. The centroid mass (m/z=88,820 Da) shows a mass shift of 431 Da (78%), indicating complete modification of EGFR T790M/L858R by Compound I-4.


Compounds I-1 and I-3 were similarly tested and found to covalently modify the protein.


Example 61

H1975 Tumor In Vivo Study


Female nu/nu mice were implanted with 1×107 H1975 tumor cells in 50% Matrigel subcutaneously (0.2 ml injection volume) in the flank. Tumor measurements were recorded three times per week. Tumors were pair matched when they reached an average size of 100-150 mg. Group size was 10 mice. Test compound was administered intraperitoneal, 25 mg/kg daily for 21 days. % Tumor inhibition values were determined at 15 days, the time at which the control group reached a maximum tumor volume. Tumor volume was followed until tumors reached 1500 mm3 or 60 days.


Tumor inhibition values for provided compounds are shown in Table 6, below.












TABLE 6







Compound #
% Tumor inhibition









I-1
>66



I-2
>66



I-4
>66



I-5
>33










While a number of embodiments of this invention are described herein, it is apparent that the basic examples may be altered to provide other embodiments that utilize the compounds and methods of this invention. Therefore, it will be appreciated that the scope of this invention is to be defined by the appended claims rather than by the specific embodiments that have been represented by way of example.

Claims
  • 1. A method for selectively inhibiting at least one mutant of epidermal growth factor receptor (EGFR) as compared to wild-type epidermal growth factor receptor (WT EGFR), in a biological sample or in a patient, comprising contacting the biological sample with, or administering to the patient, a compound of formula I-a
  • 2. The method according to claim 1, wherein said compound is sparing for WT EGFR.
  • 3. The method according to claim 1, wherein the at least one mutant is T790M.
  • 4. The method according to claim 2, wherein the at least one mutant is T790M.
  • 5. The method according to claim 1, wherein the at least one mutant of EGFR is an activating mutant.
  • 6. The method according to claim 5, wherein the activating mutant is a deletion mutant.
  • 7. The method according to claim 5, wherein the activating mutant is a point mutation.
  • 8. The method according to claim 6, wherein the deletion mutant is delE746-A750.
  • 9. The method according to claim 7, wherein the point mutation is L858R.
  • 10. The method according to claim 7, wherein the point mutation is G719S.
  • 11. The method according to claim 1, wherein the compound selectively inhibits at least one activating mutant and T790M.
  • 12. The method according to claim 11, wherein the at least one activating mutant is a deletion mutant.
  • 13. The method according to claim 11, wherein the at least one activating mutant is a point mutation.
  • 14. The method according to claim 12, wherein the deletion mutant is delE746-A750.
  • 15. The method according to claim 13, wherein the point mutation is L858R.
  • 16. The method according to claim 13, wherein the point mutation is G719S.
  • 17. A method for modulating mutant epidermal growth factor receptor in a patient, comprising administering to the patient a compound of formula I-a:
  • 18. The method according to claim 17, wherein the patient is suffering from a cancer.
  • 19. The method according to claim 18, wherein the cancer is non-small cell lung cancer.
  • 20. The method according to claim 1, wherein the compound is of formula III-a:
  • 21. The method according to claim 20, wherein W is —NH—.
  • 22. The method according to claim 20, wherein R2 is —CF3.
  • 23. The method according to claim 20, wherein R2 is —CF3 or Cl; andR3 is —C(O)CH3 or —SO2CH3.
  • 24. The method according to claim 21, wherein: R2 is —CF3 or Cl; andR3 is —C(O)CH3.
  • 25. The method according to claim 21, wherein: R2 is —CF3 or Cl; andR3 is —SO2CH3.
  • 26. The method according to claim 20, wherein: W is —O—;R2 is —CF3 or Cl; andR3 is —C(O)CH3.
  • 27. The method according to claim 20, wherein: W is —O—;R2 is —CF3 or Cl; andR3 is —SO2CH3.
  • 28. The method according to claim 1, wherein the compound is selected from: selected from:
  • 29. The method according to claim 1 wherein the compound is:
CROSS REFERENCE TO RELATED APPLICATIONS

The present application is a divisional of U.S. application Ser. No. 13/286,061 filed Oct. 31, 2011, now issued as U.S. Pat. No. 8,975,249 on Mar. 10, 2015, which claims priority to U.S. Provisional Application Nos. 61/409,080, filed Nov. 1, 2010, 61/411,829, filed Nov. 9, 2010, 61/412,330, filed Nov. 10, 2010, and 61/534,323, filed Sep. 13, 2011, the entirety of each of which is hereby incorporated by reference.

US Referenced Citations (183)
Number Name Date Kind
4879303 Davison et al. Nov 1989 A
5958935 Davis et al. Sep 1999 A
6093716 Davis et al. Jul 2000 A
6114333 Davis et al. Sep 2000 A
6127376 Davey et al. Oct 2000 A
6160010 Uckun et al. Dec 2000 A
6262088 Phillips Jul 2001 B1
6469168 Ratzne Simonek et al. Oct 2002 B1
6579983 Batchelor et al. Jun 2003 B1
6593326 Bradbury et al. Jul 2003 B1
6838464 Pease et al. Jan 2005 B2
6939874 Harmange et al. Sep 2005 B2
7060827 Singh et al. Jun 2006 B2
7122542 Singh et al. Oct 2006 B2
7125879 Guillemont et al. Oct 2006 B2
7176212 Breault et al. Feb 2007 B2
7202033 Prescott et al. Apr 2007 B2
7241769 Stadtmueller et al. Jul 2007 B2
7282504 Armistead et al. Oct 2007 B2
7329671 Singh et al. Feb 2008 B2
7329672 Singh et al. Feb 2008 B2
7332484 Singh et al. Feb 2008 B2
7435814 Singh et al. Oct 2008 B2
7452879 Singh et al. Nov 2008 B2
7485724 Singh et al. Feb 2009 B2
7491732 Li et al. Feb 2009 B2
7498435 Singh et al. Mar 2009 B2
7504396 Nunes et al. Mar 2009 B2
7514444 Honigberg et al. Apr 2009 B2
7514445 Freyne et al. Apr 2009 B2
7514446 Davis-Ward et al. Apr 2009 B2
7517886 Singh et al. Apr 2009 B2
7528143 Noronha et al. May 2009 B2
7531548 Guillemont et al. May 2009 B2
7550460 Singh et al. Jun 2009 B2
7557210 Singh et al. Jul 2009 B2
7582648 Singh et al. Sep 2009 B2
7589200 Singh et al. Sep 2009 B2
7642351 Singh et al. Jan 2010 B2
7655797 Singh et al. Feb 2010 B2
7718662 Chen et al. May 2010 B1
7741330 Chen et al. Jun 2010 B1
7803939 Singh et al. Sep 2010 B2
7812029 Singh et al. Oct 2010 B1
7820819 Singh et al. Oct 2010 B2
7825116 Singh et al. Nov 2010 B2
7858633 Li et al. Dec 2010 B2
7884111 Argade et al. Feb 2011 B2
7906644 Singh et al. Mar 2011 B2
8088781 Honigberg et al. Jan 2012 B2
8148525 Singh et al. Apr 2012 B2
8158621 Singh et al. Apr 2012 B2
8188276 Singh et al. May 2012 B2
8334296 Singh et al. Dec 2012 B2
8338439 Singh et al. Dec 2012 B2
8450335 Singh et al. May 2013 B2
8501751 Honigberg et al. Aug 2013 B2
8557806 Singh et al. Oct 2013 B2
8563568 Witowski et al. Oct 2013 B2
8609679 Singh et al. Dec 2013 B2
8710222 Singh et al. Apr 2014 B2
8735404 Honigberg et al. May 2014 B2
8748438 Honigberg et al. Jun 2014 B2
8748597 Singh et al. Jun 2014 B2
8796255 Lee et al. Aug 2014 B2
8822685 Singh et al. Sep 2014 B2
8835430 Singh et al. Sep 2014 B2
8853397 Singh et al. Oct 2014 B2
8883435 Honigberg et al. Nov 2014 B2
8883803 Honigberg et al. Nov 2014 B2
8975249 Lee et al. Mar 2015 B2
20040019067 Armistead et al. Jan 2004 A1
20040023957 Wang et al. Feb 2004 A1
20040077661 Arbiser Apr 2004 A1
20050004125 Freyne et al. Jan 2005 A1
20050014753 Ding et al. Jan 2005 A1
20050085637 Cheung et al. Apr 2005 A1
20050209221 Nunes et al. Sep 2005 A1
20050272083 Seshagiri Dec 2005 A1
20060030018 Zuccola et al. Feb 2006 A1
20060079543 Sum et al. Apr 2006 A1
20060084644 Pal et al. Apr 2006 A1
20060084645 Pal et al. Apr 2006 A1
20060100227 Baenteli et al. May 2006 A1
20060148800 Stadtmueller et al. Jul 2006 A1
20060160803 Adams et al. Jul 2006 A1
20060247241 Garcia-Echeverria et al. Nov 2006 A1
20060247262 Baenteli et al. Nov 2006 A1
20060270694 Wong Nov 2006 A1
20060293311 Li et al. Dec 2006 A1
20070010668 Davis-Ward et al. Jan 2007 A1
20070032493 Foley et al. Feb 2007 A1
20070066658 Chappell Mar 2007 A1
20070203161 Argade et al. Aug 2007 A1
20070203162 Li et al. Aug 2007 A1
20070259904 Noronha et al. Nov 2007 A1
20080009484 Argade et al. Jan 2008 A1
20080009494 Li et al. Jan 2008 A1
20080021020 Argade et al. Jan 2008 A1
20080027045 Argade et al. Jan 2008 A1
20080039622 Singh et al. Feb 2008 A1
20080058358 Luecking et al. Mar 2008 A1
20080076921 Honigberg et al. Mar 2008 A1
20080132504 Garcia-Echeverria et al. Jun 2008 A1
20080139582 Honigberg et al. Jun 2008 A1
20080182852 Johnson et al. Jul 2008 A1
20080194603 Li et al. Aug 2008 A1
20080207613 Styles et al. Aug 2008 A1
20080207913 Breitenkamp et al. Aug 2008 A1
20080214501 Pan et al. Sep 2008 A1
20080260754 Li et al. Oct 2008 A1
20080279867 Atuegbu et al. Nov 2008 A1
20080300268 Singh et al. Dec 2008 A1
20080312438 Singh et al. Dec 2008 A1
20090131436 Imbach et al. May 2009 A1
20090137588 Singh et al. May 2009 A1
20090156622 Singh et al. Jun 2009 A1
20090171086 Singh et al. Jul 2009 A1
20090181987 Honigberg et al. Jul 2009 A1
20090215803 Rice et al. Aug 2009 A1
20090286778 Combs et al. Nov 2009 A1
20090298830 Mann et al. Dec 2009 A1
20090318407 Bauer et al. Dec 2009 A1
20100004270 Honigberg et al. Jan 2010 A1
20100016296 Singh et al. Jan 2010 A1
20100022561 Honigberg et al. Jan 2010 A1
20100029610 Singh et al. Feb 2010 A1
20100041677 Honigberg et al. Feb 2010 A1
20100081679 Greul et al. Apr 2010 A1
20100088912 Higgs et al. Apr 2010 A1
20100173285 Varmus et al. Jul 2010 A1
20100197918 Singh et al. Aug 2010 A1
20100249092 Singh et al. Sep 2010 A1
20100254905 Honigberg et al. Oct 2010 A1
20110039868 Honigberg et al. Feb 2011 A1
20110105472 Greul et al. May 2011 A1
20110144330 Singh et al. Jun 2011 A1
20110207736 Gray et al. Aug 2011 A1
20110224235 Honigberg et al. Sep 2011 A1
20110245156 Sielecki-Dzurdz Oct 2011 A1
20110245284 Greul et al. Oct 2011 A1
20110281322 Honigberg et al. Nov 2011 A1
20110281850 Flynn et al. Nov 2011 A1
20120040968 Shimada et al. Feb 2012 A1
20120065201 Honigberg et al. Mar 2012 A1
20120071497 Buggy et al. Mar 2012 A1
20120083006 Ramsden et al. Apr 2012 A1
20120087915 Buggy et al. Apr 2012 A1
20120088912 Honigberg et al. Apr 2012 A1
20120094999 Gray et al. Apr 2012 A1
20120101113 Honigberg et al. Apr 2012 A1
20120101114 Honigberg et al. Apr 2012 A1
20120149722 Lee et al. Jun 2012 A1
20120165328 Honigberg et al. Jun 2012 A1
20120165332 Major et al. Jun 2012 A1
20120184013 Honigberg et al. Jul 2012 A1
20120184567 Honigberg et al. Jul 2012 A1
20120202264 Honigberg et al. Aug 2012 A1
20120270237 Ramsden et al. Oct 2012 A9
20120296089 Honigberg et al. Nov 2012 A1
20120316135 Dalgarno et al. Dec 2012 A1
20120329130 Honigberg et al. Dec 2012 A1
20130035334 Honigberg et al. Feb 2013 A1
20130065879 Singh et al. Mar 2013 A1
20130072469 Singh et al. Mar 2013 A1
20130137708 Garske et al. May 2013 A1
20140057929 Witowski et al. Feb 2014 A1
20140134265 Buggy et al. May 2014 A1
20140142123 Honigberg et al. May 2014 A1
20140163027 Verner et al. Jun 2014 A1
20140163046 Honigberg et al. Jun 2014 A1
20140187564 Honigberg et al. Jul 2014 A1
20140187565 Honigberg et al. Jul 2014 A1
20140213574 Singh et al. Jul 2014 A1
20140303154 Singh et al. Oct 2014 A1
20140303191 Buggy et al. Oct 2014 A1
20140330007 Singh et al. Nov 2014 A1
20140371241 Buggy et al. Dec 2014 A1
20150005297 Singh et al. Jan 2015 A1
20150025055 Lee et al. Jan 2015 A1
20150038518 Balasubramanian Feb 2015 A1
20150126504 Singh et al. May 2015 A1
20150158823 Singh et al. Jun 2015 A1
Foreign Referenced Citations (102)
Number Date Country
102558149 Jul 2012 CN
103159742 Jun 2013 CN
1 054 004 Nov 2000 EP
H0741461 Feb 1995 JP
WO-9628427 Sep 1996 WO
WO-9719065 May 1997 WO
WO-0027825 May 2000 WO
WO-0046203 Aug 2000 WO
WO-0078731 Dec 2000 WO
WO-0147897 Jul 2001 WO
WO-0160816 Aug 2001 WO
WO-0164654 Sep 2001 WO
WO-0164655 Sep 2001 WO
WO-0185699 Nov 2001 WO
WO-02083653 Oct 2002 WO
WO-03016306 Feb 2003 WO
WO-03030909 Apr 2003 WO
WO-03063794 Aug 2003 WO
WO-03066601 Aug 2003 WO
WO-2004014382 Feb 2004 WO
WO-2004031232 Apr 2004 WO
WO-2004056786 Jul 2004 WO
WO-2004069812 Aug 2004 WO
WO-2004074244 Sep 2004 WO
WO-2004080980 Sep 2004 WO
WO-2004096224 Nov 2004 WO
WO-2005013996 Feb 2005 WO
WO-2005016893 Feb 2005 WO
WO-2005016894 Feb 2005 WO
WO-2005026130 Mar 2005 WO
WO-2005026158 Mar 2005 WO
WO-2005063722 Jul 2005 WO
WO-2005070890 Aug 2005 WO
WO-2006021544 Mar 2006 WO
WO-2006045066 Apr 2006 WO
WO-2006053109 May 2006 WO
WO-2006055561 May 2006 WO
WO-2006068770 Jun 2006 WO
WO-2006074057 Jul 2006 WO
WO-2006078846 Jul 2006 WO
WO-2006101977 Sep 2006 WO
WO-2006108487 Oct 2006 WO
WO-2006124874 Nov 2006 WO
WO-2006128129 Nov 2006 WO
WO-2006129100 Dec 2006 WO
WO-2006133426 Dec 2006 WO
WO-2007027238 Mar 2007 WO
WO-2007048064 Apr 2007 WO
WO-2007053452 May 2007 WO
WO-2007056151 May 2007 WO
WO-2007085833 Aug 2007 WO
WO-2007089768 Aug 2007 WO
WO-2007113254 Oct 2007 WO
WO-2007113256 Oct 2007 WO
WO-2007120339 Oct 2007 WO
WO-2007120980 Oct 2007 WO
WO-2007125351 Nov 2007 WO
WO-2008005538 Jan 2008 WO
WO-2008009458 Jan 2008 WO
WO-2008025556 Mar 2008 WO
WO-2008049123 Apr 2008 WO
WO-2008064274 May 2008 WO
WO-2008073687 Jun 2008 WO
WO-2008074515 Jun 2008 WO
WO-2008079719 Jul 2008 WO
WO-2008079907 Jul 2008 WO
WO-2008080964 Jul 2008 WO
WO-2008080965 Jul 2008 WO
WO-2008088303 Jul 2008 WO
WO-2008092199 Aug 2008 WO
WO-2008107096 Sep 2008 WO
WO-2008115738 Sep 2008 WO
WO-2008115742 Sep 2008 WO
WO-2008118822 Oct 2008 WO
WO-2008118823 Oct 2008 WO
WO-2009012421 Jan 2009 WO
WO-2009017838 Feb 2009 WO
WO-2009029682 Mar 2009 WO
WO-2009032668 Mar 2009 WO
WO-2009032694 Mar 2009 WO
WO-2009032703 Mar 2009 WO
WO-2009080638 Jul 2009 WO
WO-2009105675 Aug 2009 WO
WO-2009112490 Sep 2009 WO
WO-2009115267 Sep 2009 WO
WO-2009127642 Oct 2009 WO
WO-2009136995 Nov 2009 WO
WO-2009143389 Nov 2009 WO
WO-2009158571 Dec 2009 WO
WO-2010025833 Mar 2010 WO
WO-2010081679 Jul 2010 WO
WO-2010129053 Nov 2010 WO
WO-2011079231 Jun 2011 WO
WO-2011090760 Jul 2011 WO
WO-2011140338 Nov 2011 WO
WO-2011153514 Dec 2011 WO
WO-2012021444 Feb 2012 WO
WO 2012061299 May 2012 WO
WO-2012061303 May 2012 WO
WO-2012061415 May 2012 WO
WO-2012064706 May 2012 WO
WO-2012158843 Nov 2012 WO
Non-Patent Literature Citations (63)
Entry
Jordan, V. C. Nature Reviews: Drug Discovery, 2, 2003, 205.
Hackam, et al. JAMA, 296(14), 2006, 1731-1732.
U.S. Appl. No. 13/518,833, Jun. 22, 2012, Gray et al.
Aliagas-Martin, I. et al., A class of 2,4-bisanilinopyrimidine Aurora A inhibitors with unusually high selectivity against Aurora B, J. Med. Chem. 52:3300-3307 (2009).
Andrulis, I. et al., Neu/ErbB-2 amplification identifies a poor-prognosis group of women with node-negative breast cancer, J Clin Oncol 16:1340-9 (1998).
Bamborough, P. et al., N-4-Pyrimidinyl-1H-indazol-4-amine inhibitors of Lck: Indazoles as phenol isosteres with improved pharmacokinetics, Bioorg. & Med. Chem. Lett. 17:4363-4368 (2007).
Calvo, E. et al., Administration of CI-1033, an Irreversible Pan-erbB Tyrosine Kinase Inhibitor, is Feasible on a 7-Day Off Schedule: A Phase I Pharmacokinetic and Food Effect Study, Clinical Cancer Research, 10: 7112-7120 (2004).
Carter, T. et al, Inhibition of drug-resistant mutants of ABL, KIT, and EGF receptor kinases, Proc. Natl. Acad. Sci. USA 102(31):11011-11016 (2005).
Cohen, M. et al., Structural bioinformatics-based design of selective, irreversible inhibitors, Science 308:1318-1321 (2005).
Curto, M. et al., Contact-dependent inhibition of EGFR signaling by Nf2/Merlin, J Cell Biol 177:893-903 (2007).
Ding, K. et al., Design, Synthesis and Biological Evaluation of Novel Conformationally Constrained Inhibitors Targeting Epidermal Growth Factor Receptor T790M mutant, J. Med. Chem., Just Accepted Manuscript, 1-36 (2012).
European Search Report for EP10844293.0, 8 pages (Jun. 27, 2013).
European Search Report for EP11816874.9, 5 pages (Dec. 12, 2014).
European Search Report for EP11838624.2, 5 pages (Jun. 6, 2014).
European Search Report for EP11838628.3, 7 pages (Jun. 20, 2014).
European Search Report for EP11839800.7, 8 pages (Jun. 24, 2014).
Fabian, M. et al., A small molecule-kinase interaction map for clinical kinase inhibitors, Nature Biotechnology, 23(3): 329 (2005).
Fallon, K. et al., Constitutive activation of the neuregulin-1/erbB signaling pathway promotes the proliferation of a human peripheral neuroepithelioma cell line, J Neuro Oncol 66:273-84 (2004).
Frank, D., STAT signaling in the pathogenesis and treatment of cancer, Mol. Med. 5 :432-456 (1999).
Fry, D. et al., Specific, irreversible inactivation of the epidermal growth factor receptor and erbB2, by a new class of tyrosine kinase inhibitor, Proc. Natl. Acad. Sci. USA 95:12022-12027 (1998).
Ghoneim, K., Synthesis and evaluation of some 2-, 4-, di- substituted -6-methylpyrimidine derivatives for antimicrobial activity, J. Indian Chem. Soc. 63(10):914-917 (1986).
Ghosh, D., 2-4-bis (arylamino) -5- methylpyrimidines as antimicrobial agents, J. Med. Chem. 10(5):974 (1967).
Ghosh, D., 2-4-bis (arylamino) -6- methylpyrimidines as antimicrobial agents, J. Indian Chem. Soc. 58(5):512-573 (1981).
Gonzales, A. et al, Antitumor activity and pharmacokinetic properties of PF-00299804, a second-generation, irreversible pan-erbB receptor tyrosine kinase inhibitor, Mol. Cancer Ther. 7(7):1880-1889 (2008).
Hur, W. et al., Clinical stage EGFR inhibitors irreversibly alkylate Bmx kinase, Bioorg. Med. Chem. Lett. 18:5916-5919 (2008).
International Search Report for PCT/US10/31714 dated Aug. 13, 2010.
International Search Report for PCT/US11/46926, 2 pages (Dec. 22, 2011).
International Search Report for PCT/US11/58610, 4 pages (Mar. 27, 2012).
International Search Report for PCT/US11/58616, 3 pages (Mar. 27, 2012).
International Search Report for PCT/US11/59726, 3 pages (Mar. 20, 2012).
International Search Report for PCT/US2009/048784, 8 pages (Nov. 16, 2009).
International Search Report for PCT/US2010/062432, 4 pages (May 26, 2011).
Kirken, R., Targeting Jak3 for immune suppression and allograft acceptance, Transplant. Proc. 33 :3268-3270 (2001).
Kumar, A., et al, Structure and Clinical Relevance of the Epidermal Growth Factor Receptor in Human Cancer, Journal of Clinical Oncology 26(10):1742-1751 (2008).
Kwak, E. et al., Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib, Proc. Natl. Acad. Sci. USA 102:7665-7670 (2005).
Lajeunesse, D. et al., A systematic screen for dominant second-site modifiers of Merlin/NF2 phenotypes reveals an interaction with blistered/DSRF and scribbler, Genetics 158:667-79 (2001).
Li, D. et al., BIBW2992, an irreversible EGFR/HER2 inhibitor highly effective in preclinical lung cancer models, Oncogene 27:4702-4711 (2008).
Lin, N. and Winer, E., New targets for therapy in breast cancer: Small molecule tyrosine kinase inhibitors, Breast Cancer Res 6:204-210 (2004).
Malaviya, R. et al., Targeting Janus Kinase 3 in Mast Cells Prevents Immediate Hypersensitivity Reactions and Anaphylaxis, J. Biol. Chem. 274 :27028-27038 (1999).
McClatchey, A. and Giovannini, M., Membrane organization and tumorigenesis—the NF2 tumor suppressor, Merlin, Genes Dev 19:2265-77 (2005).
Minkovsky, N. and Berezov, A., BIBW-2992, a dual receptor tyrosine kinase inhibitor for the treatment of solid tumors, Curr Opin Invest Drugs 9:1336-1346 (2008).
Pearlstein, R. et al., Understanding the structure-activity relationship of the human ether-a-go-go-related gene cardiac K+ channel. A model for bad behavior, Journal of Medicinal Chemistry, 46(11):2017-2022 (2003).
Pelton, P. et al., Ruffling membrane, stress fiber, cell spreading and proliferation abnormalities in human Schwann cells, Oncogene 17:2195-2209 (1998).
PubChem CID 44594695. Feb. 1, 2010. [Retrieved from the Internet May 15, 2011: http://pubchem.ncbi.nlm.nih.gov/summary.cgi?cid=44594695&loc=ec—rcs].
Readinger, J. et al., Selective Targeting of ITK Blocks Multiple Steps of HIV Replication, Proc. Natl. Acad. Sci. USA 105: 6684-6689 (2008).
Seidel, H. et al., Pharmaceutical intervention in the JAK/STAT signaling pathway, Oncogene 19: 2645-2656 (2000).
Sequist, L., Second-Generation Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Non-Small Cell Lung Cancer, The Oncologist 12(3):325-330 (2007).
Singh, J. et al, Structure-based design of a potent, selective, and irreversible inhibitor of the catalytic domain of the erbB receptor subfamily of protein tyrosine kinases, J. Med. Chem. 40:1130-1135 (1997).
Sjin, R. et al., In vitro and in vivo characterization of irreversible mutant-selective EGFR inhibitors that are wild-type sparing, Molecular Cancer Therapeutics, 13(6):1468-1479 (2014).
Stonecypher, M. et al., Activation of the neuregulin-1/ErbB signaling pathway promotes the proliferation of neoplastic Schwann cells in human malignant peripheral nerve sheath tumors, Oncogene 24:5589-5605 (2005).
Sudbeck, E. et al., Structure-based Design of Specific Inhibitors of Janus Kinase 3 as Apoptosis-inducing Antileukemic Agents, Clin. Cancer Res. 5: 1569-1582 (1999).
Trieu, V. et al., A specific inhibitor of janus kinase-3 increases survival in a transgenic mouse model of amyotrophic lateral sclerosis, Biochem. Biophys. Res. Commun. 267 :22-25 (2000).
Wong, K. et al, A phase I study with neratinib (HKI-272), an irreversible pan Erb B receptor tyrosine kinase inhibitor, in patients with solid tumors, Clin. Cancer Res. 15(7):2552-2558 (2009).
Written Opinion for PCT/US10/31714, 7 pages (Aug. 13, 2010).
Written Opinion for PCT/US11/46926, 9 pages (Dec. 22, 2011).
Written Opinion for PCT/US11/58610, 8 pages (Mar. 27, 2012).
Written Opinion for PCT/US11/58616, 9 pages (Mar. 27, 2012).
Written Opinion for PCT/US11/59726, 7 pages (Mar. 20, 2012).
Written Opinion for PCT/US2009/048784, 9 pages (Nov. 16, 2009).
Written Opinion for PCT/US2010/062432, 6 pages (May 26, 2011).
Zhang, J. et al., Targeting Cancer with Small Molecule Kinase Inhibitors, Nature Rev. Cancer 9:28-39 (2009).
Zhang, Y. et al., Antitumor Activity of Epidermal Growth Factor Receptor-Related Protein Is Mediated by Inactivation of ErbB Receptors and Nuclear Factor-kB in Pancreatic Cancer, Cancer Res 66:1025-1032 (2006).
Zhou, W. et al. Novel mutant-selective EGFR kinase inhibitors against EGFR T790M, Nature, 462(7276): 1070-1074 (2009).
Related Publications (1)
Number Date Country
20150246040 A1 Sep 2015 US
Provisional Applications (4)
Number Date Country
61534323 Sep 2011 US
61412330 Nov 2010 US
61411829 Nov 2010 US
61409080 Nov 2010 US
Divisions (1)
Number Date Country
Parent 13286061 Oct 2011 US
Child 14636905 US