OA18657A - Substituted 2-anilinopyrimidine derivatives as EGFR modulators. - Google Patents

Substituted 2-anilinopyrimidine derivatives as EGFR modulators. Download PDF

Info

Publication number
OA18657A
OA18657A OA1201700215 OA18657A OA 18657 A OA18657 A OA 18657A OA 1201700215 OA1201700215 OA 1201700215 OA 18657 A OA18657 A OA 18657A
Authority
OA
OAPI
Prior art keywords
cancer
compound
pharmaceutically acceptable
prodrug
solvaté
Prior art date
Application number
OA1201700215
Inventor
Jirong Peng
Michael John Costanzo
Michael Nicholas Greco
Michael Alan Green
Victoria Lynn Wilde
Don Zhang
Original Assignee
Bata Pharma, Inc.
Beta Pharma (Shanghai) Co., Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bata Pharma, Inc., Beta Pharma (Shanghai) Co., Ltd. filed Critical Bata Pharma, Inc.
Publication of OA18657A publication Critical patent/OA18657A/en

Links

Abstract

This application discloses novel substituted 2anilinopyrimidine derivatives, and pharmaceutically acceptable salts, solvates, prodrugs, and compositions thereof, which are useful for the treatment or prevention of diseases or medical conditions mediated by epidermal growth factor receptors (EGFRs), including but not limited to a variety of cancers.

Description

BETA PHARMA, INC and
BETA PHARMA (SHANGHAI) CO., LTD
SUBSTITUTED 2-ANILINOPYRIMIDINE DERIVATIVES AS EGFR MODULATORS j
CROSS REFERENCE TO RELATED APPLICATIONS
This application daims priority under 35 U.S.C, § 119(e) to U.S. Provisional Application No. 62/090,869, filed on December 11, 2014, and No. 62/166,883, filed on May 27, 2015, both of which are incorporated herein by reference in their entireties.
FIELD OF THE INVENTION
The présent invention relates to substituted 2-anilinopyrimidine dérivatives and pharmaceutically acceptable salts and compositions thereof useful for the treatment or prévention of diseases or medical conditions mediated through mutated forms of epidermal growth factor receptor (EGFR), such as varions cancers.
BACKGROUND OF THE INVENTION
The epidermal growth factor receptor (EGFR, Herl, ErbBl) is a principal member of the ErbB family of four structurally-related cell surface receptors with the other members being Her2 (Neu, ErbB2), Her3 (ErbB3) and Her4 (ErbB4). EGFR exerts its primary cellular functions though its intrinsic catalytic tyrosine protein kinase activity. The receptor is activated by binding with growth factor ligands, such as epidermal growth factor (EGF) and transforming growth factor-alpha (TGF-α), which transform the catalytically inactive EGFR monomer into catalytically active homo- and hetero- dimers. These catalytically active dimers then initiate intracellular tyrosine kinase activity, which leads to the autophosphorylation of spécifie EGFR tyrosine residues and elicits the downstream activation of signaling proteins. Subsequently, the signaling proteins initiate multiple signal transduction cascades (MAPK, Akt and INK), which ultimately médiate the essential biological processes of cell growth, prolifération, motility and survival. ,
EGFR is found at abnormally high levels on the surface of many types of cancer cells and increased levels of EGFR .hâve been associated with advanced disease, cancer spread and poor clinical prognosis. . Mutations in EGFR can lead to receptor overexpression, perpétuai activation or sustained hyperactivity and resuit in uncontrolled cell growth, i.e. cancer. Consequently, EGFR mutations hâve been identified in several types of malignant tumors, including metastatic lung, head and neck, colorectal and pancreatic cancers. In lung cancer, mutations mainly occur in exons 18 to 21, which encode the adenosine triphosphate (ATP)-binding pocket of the kinase domain. The most clinically relevant drug-sensitive EGFR mutations are délétions in exon 19 that eliminate a common amino acid motif (LREA) and point mutations in exon 21, which lead to a substitution of arginine for leucine at position 858 (L858R). Together, these two mutations account for nearly 85% of the EGFR mutations observed in lung cancer, Both mutations hâve perpétuai tyrosine kinase activity and as a resuit they are oncogenic. Biochemical studies hâve demonstrated that these mutated EGFRs bind preferentially to tyrosine kinase inhibitor drugs such as erlotinib and gefitinib over adenosine triphosphate (ATP).
Erlotinib and gefitinib are oral EGFR tyrosine kinase inhibitors that are first line monotherapies for non-small cell lung cancer (NSCLC) patients having activating mutations in EGFR. Around 70% of these patients respond initially, but unfortunately they develop résistance with a médian time to progression of 10-16 months. In at least 50% of these initially responsive patients, disease progression is associated with the development of a secondary mutation, T790M in exon 20 of EGFR (referred to as the gatekeeper mutation). The additional T790M mutation increases the affmity of the EGFR kinase domain for ATP, thereby reducing the inhibitory activity of ATP-competitive inhibitors like gefitinib and erlotinib.
Recently, irréversible EGFR tyrosine kinase inhibitors hâve been developed that effectively inhibit the kinase domain of the T790M double mutant and therefore overcome the résistance observed with réversible inhibitors in the clinic. These inhibitors possess reactive electrophilic functional groups that react with the nucleophilic thiol of an active-site cysteine. Highly sélective irréversible inhibitors can be achieved by exploiting the inhérent non-covalent selectivity of a given scaffold along with the location of a particular cysteine residue within the ATP binding site. The acrylamide moieties of these inhibitors both undergo a Michael reaction with Cys797 in the ATP binding site of EGFRT79OM to form a covalent bond. This covalent mechanism is thought to overcome the increase in ATP affmity of the T790M EGRF double mutant and give rise to effective inhibition. However, these inhibitors may cause varions undesired toxicities. Therefore, development of new inhibitors for treatment of varions EGFR-related cancers is still in high demand.
SUMMARY OF THE INVENTION
The présent invention provides novel compounds as EGFR tyrosine kinase inhibitors that are therapeutically useful in the treatment or prévention of a number of EGFR-related diseases or disorders, such as varions cancers.
In one aspect, the présent invention provides compounds of formula I:
R3
I or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein:
G is selected from substituted or unsubstituted 177-indol-3-yl, substituted or unsubstituted J/Z-indazol-3-yl, substituted or unsubstituted 2/7-indazoI-3-yl, and substituted or unsubstituted pyrazolo[l,5-a]-pyridin-3-yl, and substituted or unsubstituted 4,5,6,7-tetrahydropyrazolo[l,5a]pyridin-3-yl;
X is selected from oxygen, sulfur, and methylene;
R1 is selected from hydrogen, halogen, methyl, trifluoromethyl, and cyano;
R2, R3, and R4 are the saine or different and are independently selected from hydrogen, halogen and trifluoromethyl;
R5 is selected from lower alkyl, optionally substituted 3- to 6-membered heterocyclyl, R7R8N-(lower alkyl), and R7R8N-(cycloalkylalkyl), wherein R7 and R8 are the same or different and are independently selected from hydrogen and lower alkyl; and
R6 is selected from lower alkoxy and lower alkyl,
Ih some preferred embodiments, in formula I, G is a 77ï-indol-3-yl or 7/7-indazol-3-yl
R9
moiety having a formula i ; and the présent invention provides a compound of formula II:
OCH3 s
II or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein:
X is O, S, or CH2;
Qis C-Rl0orN
R9 is CH3 or CH2CH2F; and
R10 is H or CH3.
In some other preferred embodiments, in formula I, G is pyrazolo[l,5-c]-pyridin-3-yl, and the présent invention provides a compound of formula V:
V or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein X is O, S, or CH2.
In another aspect the présent invention provides pharmaceutical compositions comprising any of the compounds, or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, and a pharmaceutically acceptable carrier.
The compounds and compositions of the présent invention are usefiil for treating diseases, disorders, or conditions associated with one or more EGFR mutations. Such diseases, disorders, or conditions include those described herein, such as varions cancers.
Thus, in another aspect, the présent invention provides methods of treating diseases or disorders associated with EGFR activities, such as varions cancers associated with one or more EGFR mutations, or use of the compounds or compositions in the manufacture of médicaments for treatment of these diseases or disorders.
In another aspect, the compounds of this invention are useful for the study of kinases in biological and pathological phenomena; the study of transduction pathways mediated by such kinases; and the comparative évaluation of new kinase inhibitors.
In another aspect, the présent invention provides methods of synthesizing the compounds disclosed herein.
Other aspects or advantages of the présent invention will be better appreciated in view of the detailed description and daims.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 illustrâtes the H1975 tumor growth inhibition assay results for Example 1 in mice.
FIG. 2 illustrâtes the H1975 tumor growth inhibition assay results for Example 2 in mice.
FIG. 3 illustrâtes the HCC827 tumor growth inhibition assay results for Example 1 in mice.
FIG. 4 illustrâtes the average concentrations of Example 1 in plasma, brain and tumor tissues in mice following oral administration of a 25 mg/kg dose in the HCC827 mouse xenograft model.
DETAILED DESCRIPTION OF THE INVENTION
In one aspect, the présent invention provides a compound of the formula I:
R3 or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein:
G is selected from the group consisting of substituted or unsubstituted 17f-indol-3-yl, substituted or unsubstituted 777-indazol-3-yl, substituted or unsubstituted 2/f-indazol-3-yI, substituted or unsubstituted pyrazolo[l,5-«]-pyridin-3-yl, and substituted or unsubstituted 4,5,6,7tetrahydropyrazolo[l,5-«]pyridin-3-yl;
X is oxygen, sulfur, or methylene;
R* is hydrogen, halogen, methyl, trifluoromethyl, or cyano;
R2, R3, and R4 are the same or different and are. independently selected from the group consisting of hydrogen, halogen, and trifluoromethyl;
R5 is selected from the group consisting of lower alkyl, optionally substituted 3- to 6membered heterocyclyl, R7R8N-(lower alkyl), and R7R8N-(cycloalkylalkyl), wherein R7 and R8 are the same or different and are each independently selected from hydrogen and lower alkyl; and
R6 is lower alkoxy or lower alkyl.
In one embodiment of this aspect, G is selected from the group consisting of 777-indol-3-yl, l-methyl-777-indol-3-yl, l-(2-fluoroethyl)-77/-indol-3-yl, l,2-dimethyl-77ï-indol-3-yl, pyrazolo[l,5-a]-pyridin-3-yl, 4,5,6,7-tetrahydropyrazolo[l,5-n]pyridin-3-yl, 1-methyl-777indazol-3-yl, and 2-methyl-277-indazol-3-yl.
In a preferred embodiment, G is selected from the group consisting of l-methyl-777-indol-3yl, l-(2-fluorocthyl)-777-indol-3-yl, l,2-dimethyl-7/7-indol-3-yl, pyrazolo[l,5-a]-pyridin-3-yl, and l-methyl-77ï-indazol-3-yl.
In a more preferred embodiment, G is 1-methyl-7 77-indol-3-yl, l-(2-fluoroethyl)-777-indol3-yl, or l,2-dimethyl-777-indol-3-yl, and more preferably 1-methyl-777-indol-3-yl.
In another more preferred embodiment, G is pyrazolo[l,5-a]-pyridin-3-yl.
In another more preferred embodiment, G is l-methyl-77ï-indazol-3-yl.
In another embodiment of this aspect, R5 is selected from the group consisting of Ci-Cf, alkyl, substituted or unsubstituted azetidinyl, substituted or unsubstituted pyrrolidinyl, substituted or unsubstituted piperidinyl, R7R8N-(CH2)n- (n is an integer selected from 1 to 5), R7R8N-(C3-C6 cycloalkyl)-(CH2)m- (m = 1, 2, 3), wherein R7 and R8 are the same or different and are independently selected from hydrogen and lower alkyl.
In a preferred embodiment of this aspect, R5 is selected from the group consisting of methyl, l-(dimethylamino)-cyclopropylmethyl, 3-(dimethylamino)cyclobutyl, l-methylazetidin-3-yl, (7?)l-methylpyrrolÎdin-3-yl, (S)-l-methylpyrrolidin-3-yl, and l-methylpiperidin-4-yl, and 2dimethylamino-ethyl.
In a more preferred embodiment, R5 is 2-dimethylamino-ethyl [i.e., (CH3)2NCH2CH2-].
In another embodiment of this aspect, R1 is hydrogen, halogen, or methyl.
In a preferred embodiment of this aspect, R1 is hydrogen.
In another embodiment of this aspect, R2 is hydrogen or halogen, wherein halogen is preferably F or Cl.
In another embodiment of this aspect, R3 is hydrogen, F, Cl, or -CF3.
In another embodiment of this aspect, R4 is hydrogen.
In another embodiment of this aspect, R2 is hydrogen, F, or Cl; R3 is hydrogen, F, Cl, or -CF3; and R4 is hydrogen.
In a preferred embodiment of this aspect, R2, R3, and R4 are ail hydrogen.
In a preferred embodiment of this aspect, R6 is lower alkoxy, preferably methoxy or ethoxy.
fri a more preferred embodiment, R6 is methoxy.
In another embodiment of this aspect, sometimes preferred, X is oxygen.
In another embodiment of this aspect, sometime preferred, X is sulfur.
fri another embodiment of this aspect, sometimes preferred, X is -CH2-.
As would be understood by a person skilled in the art, any plausible and structurally allowable combinations of ail the embodiments or preferred embodiments disclosed herein are encompassed and hereby specifically included in the présent invention.
For example, in some embodiments of this aspect, G is selected from the group consisting of 777-indol-3-yl, l-methyl-777-indol-3-yl, l-(2-fluoroethyl)-77ï-indol-3-yl, l,2-dimethyl-7F7-indol
3-yl, pyrazolo[l,5-a]-pyridin-3-yl, 4,5,6,7-tetrahydropyrazolo[I,5-a]pyridin-3-yl, l-methyl-7/Zindazol-3-yl, and 2-mcthyl-2H-indazol-3-yl;
X is selected from the group consisting of oxygen, sulfur, and methylene;
R1 is selected from the group consisting of hydrogen, halogen, methyl, trifluoromethyl, and cyano;
R2, R3, and R4 are the same or different and are independently selected from the group consisting of hydrogen, halogen, and trifluoromethyl;
R5 is selected from the group consisting of l-(dimethylamino)-cyclopropylmethyl, 3(dimethylamino)cyclobutyl, 1-methylazetidin-3-yl, (R)-l-methylpyrrolidin-3-yl, (5)-1methylpyrrolidin-3-yl, and l-methylpiperidin-4-yl, and 2-dimethylamino-ethyl; and
R6 is lower alkoxy.
In some preferred embodiments, G is a 777-indol-3-yl or 7//-indazol-3-yl moiety having a
R9
formula 1 ; and the présent invention provides a compound of formula II:
or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein:
X is O, S, or CH2;
Q is C-R'° or N
R9 is CHj or CH2CH2F; and
R10 is H or CH3.
In one preferred embodiment, in formula II, Q is C-R10, and the présent invention provides a compound of formula III:
R9
III or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein R9 is CITf or CH2CH2F; and R10 is H or CH3.
In another preferred embodiment, in the compound of formula III, R9 is CH3 and R10 is H.
In another preferred embodiment, in the compound of formula III, R9 is CH3 and R10 is CH3.
In another preferred embodiment, in the compound of formula III, R9 is 2-fluoroethyl (FCH2CH2-), and R10 is H.
In another preferred embodiment, in formula III, R9 is CH3, R10 is H, and X is O, the compound having the structure of formula 1:
In another preferred embodiment, in formula III, R9 is CH3, R10 is CH3, and X is O, the compound having the structure of formula 8:
In another preferred embodiment, in formula III, R9 is CH3, R10 is H, and X is S, the compound having the structure of formula 2:
In another preferred embodiment, in formula III, R9 is CH3, R10 is H, and X is CH2, the compound having the structure of formula 4:
In another preferred embodiment, in formula III, R9 is -CH2CH2F, R10 is H, and X is O, the compound having the structure of formula 11:
F
11
In one preferred embodiment, in formula II, Q is N, and the présent invention provides a compound of formula IV:
R9
IV or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein: R9 is H or CH3; andXisO, S,orCH2.
In a more prefeired embodiment, in formula IV, R9 is H or CH3, and X is O, the compound having the structure of formula 10:
In some other preferred embodiments, in formula I, G is pyrazolo[l,5-a]-pyridin-3-yl, and the présent invention provides a compound of formula V:
V or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein X is O, S, or CH2.
In a more preferred embodiment, in formula V, X is O, the compound having the structure of formula 9:
In some other preferred embodiments, the présent invention provides a compound selected from the group consisting of the Examples listed, or a pharmaceutically acceptable sait, solvaté, or prodrug thereof.
The more preferred compounds are listed below:
la another aspect, the présent invention provides a pharmaceutical composition comprising any one of the compounds of formulas I, II, III, IV, and V, or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, and a pharmaceutically acceptable carrier, adjuvant, diluent, and/or vehicle.
In one embodiment of this aspect, the composition further comprises a second therapeutic agent.
In another embodiment of this aspect, the second therapeutic agent is a different EGFR modulator.
In another embodiment of this aspect, the second therapeutic agent is a chemotherapeutic agent.
In another aspect, the présent invention provides a method of treating a disease or disorder associated with an EGFR activity, comprising administration of a therapeutically effective amount of a compound according to any one of formulas I, II, III, IV, and V, or a pharmaceutically acceptable sait, solvaté, prodrug, or a pharmaceutical composition thereof, to a patient in need of treatment.
In one embodiment of this aspect, the disease or disorder is associated with one or more mutants of EGFR.
In another embodiment of this aspect, the mutant or mutants of EGFR are selected from L858R activating mutants L858R, delE746-A750, G719S; the Exon 19 délétion activating mutant; and the T790M résistance mutant.
In another embodiment of this aspect, the disease or disorder is a cancer.
In another embodiment of this aspect, the cancer is selected from brain cancer, lung cancer, kidney cancer, bone cancer, liver cancer, bladder cancer, head and neck cancer, esophageal cancer, stomach cancer, colon cancer, rectum cancer, breast cancer, ovarian cancer, melanoma, skin cancer, adrenal cancer, cervical cancer, lymphoma, and thyroid tumors and their complications.
In another embodiment of this aspect, the method is used in conjunction with administering to the patient a second therapeutic agent.
In another embodiment of this aspect, the second therapeutic agent is a chemotherapeutic agent.
In another embodiment of this aspect, the second therapeutic agent is a different EGFR modulator.
In another aspect, the présent invention provides a method of inhibiting a mutant of EGFR in a subject, comprising contacting a biological sample of said subject with a compound of any one of formulas I, II, III, IV, and V according to any embodiment disclosed herein, or a pharmaceutically acceptable sait, solvaté, or prodrug thereof. Such inhibition can be in vitro or in vivo. If in vivo, the method may comprise administering to said subject a compound of any one of formulas I, II, III, IV, and V according to any embodiment disclosed herein, or a pharmaceutically acceptable sait, solvaté, or prodrug thereof. If in vitro, such inhibition may be conducted in a medium in any container known to those skilled in the art.
In another aspect, the présent invention provides use of a compound of any one of formulas I, II, III, IV, and V according to any embodiment disclosed herein, or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, or a composition of any one of formulas I, II, III, IV, and V according to any embodiment disclosed herein, in the manufacture of a médicament for treatment of a disease or disorder associated with an EGFR activity.
In one embodiment of this aspect, disease or disorder is a cancer selected from the group consisting of brain cancer, lung cancer, kidney cancer, bone cancer, liver cancer, bladder cancer, head and neck cancer, esophageal cancer, stomach cancer, colon cancer, rectum cancer, breast cancer, ovarian cancer, melanoma, skin cancer, adrenal cancer, cervical cancer, lymphoma, and thyroid tumors and their complications. In a preferred embodiment, the cancer is brain cancer or lung cancer. The lung cancer includes, but is not limited to, non-small cell lung cancer and small cell lung cancer.
The terms in the présent application, if not specifically defined, take their ordinary meanings as would be understood by those skilled in the art.
As used herein, the term “halo” or “halogen” refers to F, Cl, or Br.
The term lower alkyl refers to a branched or straight-chain alkyl group having from one to seven carbon atoms, preferably one to four, and more preferably one to two carbon atoms.
The term lower alkoxy refers to an alkoxy group (-OR) having from one to seven, preferably one to four, and more preferably one to two carbon atoms.
The term “cyano” refers to -CN.
The term “pharmaceutically acceptable,” as used herein, refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of Sound medical judgment, suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergie response, or other problem or complication commensurate with a reasonable benefit/risk ratio, and are effective for their intended use.
As used herein, the term pharmaceutically acceptable sait 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 animais without undue toxicity, irritation, allergie response and the like, and are commensurate with a reasonable benefit/risk ratio. 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, glycérophosphate, 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, stéarate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like.
The terni “solvaté,” as used herein, means a physical association of a compound of this invention with a stoichiometric or non-stoichiometric amount of solvent molécules. For example, one molécule of the compound associâtes with one or more, preferably one to three, solvent molécules. It is also possible that multiple (e.g., two) molécules of the compound share one solvent molécule. This physical association may include hydrogen bonding. In certain instances the solvatés will be capable of isolation as crystalline solid. The solvent molécules in the solvaté may be présent in a regular arrangement and/or a non-ordered arrangement. Exemplary solvatés include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates. Methods of solvation are generally known in the art.
The term “prodrug,” as used herein, refers to a dérivative of a compound that can be transformed in vivo to yield the parent compound, for example, by hydrolysis in blood. Common examples include, but are not limited to, ester and amide forms of an active carboxylic acid compound; or vice versa, an ester from of an active alcohol compound or an amide form of an active amine compound. Such amide or ester prodrug compounds may be prepared according to conventional methods as known in the art. For example, a prodrug of a compound of formula II of the présent invention could be in the form of the following formula VI:
VI wherein Rx and Ry are independently H and -C(O)-R, wherein R is C1-C4 alkyl, preferably methyl or ethyl, and more preferably methyl. Other prodrugs of the présent invention can be prepared similarly from any of formulas I, II, III, IV, and V.
When it is possible that, for use in therapy, therapeutically effective amounts of a compound of the présent invention, or pharmaceutically acceptable salts or solvatés thereof, may be administered as the raw Chemical, it is possible to présent the active ingrédient as a pharmaceutical composition. Accordingly, the disclosure lurther provides pharmaceutical compositions, which include any compounds of the présent invention, or pharmaceutically acceptable salts or solvatés thereof, and one or more, preferably one to three, pharmaceutically acceptable carriers, diluents, or other excipients. The carrier(s), diluent(s), or other excipient(s) must be acceptable in the sense of being compatible with the other ingrédients of the formulation and not deleterious to the subject being treated.
Pharmaceutical formulations may be presented in unit dose forms containing a predetermined amount of active ingrédient per unit dose. Typically, the pharmaceutical compositions of this disclosure will be administered from about 1 to about 5 times per day or altematively, as a continuons infusion. Such administration can be used as a chronic or acute therapy. The amount of active ingrédient that may be combined with the carrier materials to produce a single dosage form will vary depending on the condition being treated, the severity of the condition, the time of administration, the route of administration, the rate of excrétion of the compound employed, the duration of treatment, and the âge, gender, weight, and condition of the patient. Preferred unit dosage formulations are those containing a daily dose or sub-dose, as herein above recited, or an appropriate fraction thereof, of an active ingrédient. Generally, treatment is initiated with small dosages substantially less than the optimum dose of the compound. Thereafter, the dosage is increased by small incréments until the optimum effect under the circumstances is reached. In general, the compound is most desirably administered at a concentration level that will generally afford effective results without causing substantial harmful or deleterious side effects.
When the compositions of this disclosure comprise a combination of a compound of the présent disclosure and one or more, preferably one or two, additional therapeutic or prophylactic agent, both the compound and the additional agent are usually présent at dosage levels of between about 10 to 150%, and more preferably between about 10 and 80% of the dosage normally administered in a monotherapy regimen.
Pharmaceutical formulations may be adapted for administration by any appropriate route, for example, by the oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual, or transdermal), vaginal, or parentéral (including subcutaneous, intracutaneous, intramuscular, intra-articular, intrasynovial, intrastemal, intrathecal, intralesional, intravenous, or intradermal injections or infusions) route. Such formulations may be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingrédient with the carrier(s) or excipient(s). Oral administration or administration by injection is preferred.
Pharmaceutical formulations adapted for oral administration may be presented as discrète units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid émulsions or water-in-oil émulsions.
For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as éthanol, glycerol, water, and the like. Powders are prepared by comminuting the compound to a suitable fine size and mixing with a similarly comminuted pharmaceutîcal carrier such as an edible carbohydrate, as, for example, starch or mannitol, Flavoring, preservative, dispersing, and coloring agent can also be présent.
Capsules are made by preparing a powder mixture, as described above, and fîlling formed gelatin sheaths. Glidants and lubricants such as colloïdal silica, talc, magnésium stéarate, calcium stéarate, or solid polyethylene glycol can be added to the powder mixture before the fîlling operation. A disintegrating or solubilizing agent such as agar-agar, calcium carbonate, or sodium carbonate can also be added to improve the availability of the médicament when the capsule is ingested.
Moreover, when desired or necessary, suitable binders, lubricants, disintegrating agents, and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, com sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, and the like. Lubricants used in these dosage forms include sodium oleate, sodium chloride, and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, betonite, xanthan gum, and the like. Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, addîng a lubricant and disintegrant, and pressing into tablets. A powder mixture is prepared by mixing the compound, suitable comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymethylcellulose, an aliginate, gelating, or polyvînyl pyrrolidone, a solution retardant such as paraffin, a résorption accelerator such as a quatemary sait and/or and absorption agent such as betonite, kaolin, or dicalcium phosphate. The powder mixture can be granulated by wetting with a binder such as syrup, starch paste, acadia mucilage, or solutions of cellulosic or polymètre materials and forcing through a screen. As an alternative to granulating, the powder mixture can be run through the tablet machine and the resuit is imperfectly formed slugs broken into granules. The granules can be lubricated to prevent sticking to the tablet forming dies by means of the addition of stearic acid, a stéarate sait, talc, or minerai oil. The lubricated mixture is then compressed into tablets. The compounds of the présent disclosure can also be combined with a free flowing inert carrier and compressed into tablets directly without going through the granulating or slugging steps. A clear or opaque protective coating consisting of a sealing coat of shellac, a coating of sugar or polymeric material, and a polish coating of wax can be provided. Dyestuffs can be added to these coatings to dîstinguish different unit dosages.
Oral fluids such as solution, syrups, and élixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of the compound. Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while élixirs are prepared through the use of a non-toxic vehicle. Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and polyoxyethylene sorbitol ethers, preservatives, flavor additive such as peppermint oil or natural sweeteners, or saccharin or other artificial sweeteners, and the like can also be added.
Where appropriate, dosage unit formulations for oral administration can be microencapsulated. The formulation can also be prepared to prolong or sustain the release as for example by coating or embedding particulate material in polymers, wax, or the like.
It should be understood that in addition to the ingrédients particularly mentioned above, the formulations may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
The term patient or “subject” includes both human and other mammals.
The term mammal or “mammalian animal” includes, but is not limited to, humans, dogs, cats, horses, pigs, cows, monkeys, rabbits and mice. The preferred mammals are humans.
The term “therapeutically effective amount refers to an amount of a compound or composition that, when administered to a subject for treating a disease, is sufficient to effect such treatment for the disease. A therapeutically effective amount can vary depending on, inter alia, the compound, the disease and its severity, and the âge, weight, or other factors of the subject to be treated. When applied to an individual active ingrédient, administered alone, the term refers to that ingrédient alone. When applied to a combination, the term refers to combined amounts of the active ingrédients that resuit in the therapeutic effect, whether administered in combination, serially, or simultaneously.
The term treating or “treatment” refers to: (i) inhibiting the disease, disorder, or condition, i.e., arresting its development; (ii) relieving the disease, disorder, or condition, i.e., causing régression of the disease, disorder, and/or condition; or (iii) preventing a disease, disorder or condition from occurring in a subject that may be predisposed to the disease, disorder, and/or condition but has not yet been diagnosed as having it. Thus, in one embodiment, “treating or treatment refers to ameliorating a disease or disorder, which may include ameliorating one or more physical parameters, though maybe indiscemible by the subject being treated. In another embodiment, treating or treatment includes modulating the disease or disorder, either physically (e.g., stabilization of a discemible symptom) or physiologically (e.g., stabilization of a physical parameter) or both. In yet another embodiment, treating or treatment includes delaying the onset of the disease or disorder.
When the term “about” is applied to a parameter, such as amount, température, time, or the like, it indicates that the parameter can usually vary by ±10%, preferably within ±5%, and more preferably within ±2%. As would be understood by a person skilled in the art, when a parameter is not critical, a number provided in the Examples is often given only for illustration purpose, instead of being limiting.
The term “a,” “an,” or “the,” as used herein, représenta both singular and plural fonns. In general, when either a singular or a plural form of a noun is used, it dénotés both singular and plural forms of the noun.
The following non-limiting Examples further illustrate certain aspects of the présent invention.
EXAMPLES
Chemical Synthesis
The compounds of the présent invention are prepared generally according to Synthetic Schemes 1 to 8 in the illustrative, non-limiting Examples described below.
Abbreviations
The following abbreviations may be used:
THF = Tetrahydrofuran;
conc. = concentrated
DIEA = DIPEA = Diisopropylethylamine;
sat. = saturated aqueous solution;
FCC - flash column chromatography using silica;
TFA = Trifluoroacetic acid;
r.t. = room température;
DI = deionized;
DME = 1,2-Dimethoxyethane
DMF = AÇV-Dimethylformamidc;
DMSO = Dimethylsulfoxide;
DMA = ΛζΛ'-Dimethylacetamide;
HATU = O-(7-Azabenzotriazol-l -yl)-7V,Ai,jV',A'-tetramethyluronium hexafluoro-phosphate;
EtOAc = Ethyl acetate;
h = hour(s);
NMM = A-Methylmorpholine
Pd2(dba)3 = Trîs(dibenzylideneacetone)dipalladium(0);
P(o-tol)3 = Tri(o-tolyl)phosphine.
Example 1
A-(2-(2-(Dimethylamino)ethoxy)-4-methoxy-5-((4-(l-methyl-lH-indol-3-yl)pyrimidiii2-yl)amino)phenyl)acrylamide (1)
7V-(4-(2-(Dimethylamino)ethoxy)-2-methoxy-5-nitrophenyl)-4-(l-methyl-lH-indol-3yl)pyrimîdin-2-amine (Scheme 1, Intermediate B). To a slurry of NaH (30 mmol, 60% oil dispersion prewashed with hexanes) and 50 mL of 1,4-dioxane was added 2dimethylaminoethanol (27 mmol, 2.7 mL) dropwise with stirring under N2. After stirring for 1 h, a slurry of A (5.4 mmol) in 50 mL of 1,4-dioxane was added portion-wise over 15 min under a stream of N2. The resulting mixture was stirred ovemight, then poured into water and the solid was collected, rinsed with water, and dried under vacuum to yield 2.6 g of product as a yellow solid. A purifïed sample was obtained from chromatography (silica gel; CH2C12-CH3OH gradient). *H NMR (300 MHz, DMSO) δ 2.26 (s, 6H), 2.70 (t, 2H, J= 6 Hz), 3.87 (s, 3H), 4.01 (s, 3H), 4.32 (t, 2H, J= 6 Hz), 7.00-7.53 (m, 5H), 8.18-8.78 (m, 5H); Cj^aN^m/z MH+ 463.
4-(2-(Dimethylamino)ethoxy)-6-methoxy-7Vl-(4-(l-methyl-lH-indol-3-yl)pyrimidin-2- yl)benzene-l,3-diamine (Scheme 1, Intermediate C). A suspension of 2.6 g of Intermediate B, 1.6 g of Fe°, 30 mL of éthanol, 15 mL of water, and 20 mL of conc. HCl was heated to 78 °C for
h. The solution was cooled to room température, adjusted to pH 10 with 10% NaOH (aq) and diluted with CIFCL. The mixture was filtered through Dicalite, and the fïltrate layers were separated. The aqueous phase was extracted with CH2CI2 twice, and the combined organic extracts were dried over Na2SÛ4 and concentrated. Column chromatography (silica gel, CH2C12MeOH gradient) afforded 1.2 g of Intermediate C as a solid. C24H28N6O2 m/z MH+ 433,
Ar-(2-(2-(Dîmethylamino)ethoxy)-4-methoxy-5-((4-(l-methyl-lH-indol-3-yl)pyrimidin2-yl)amino)phcnyl)acrylamide (1). To a solution of Intermediate C (2. S mmol) in 50 mL of THF and 10 mL of water was added 3-chloropropionychloride (2.8 mmol) dropwise with stirring. After 5 h of stirring, NaOH (28 mmol) was added and the mixture was heated at 65°C for 18 h. Aller cooling to room température, THF was partially removed under reduced pressure, and the mixture was extracted with CH2C12, dried over Na2SO4, and concentrated. Chromatography of the crude product (silica gel, CH2Cl2-MeOH) afforded 0.583 g of Example 1 as a beige solid. *H NMR (300 MHz, DMSO) δ 2.28 (s, 6H), 2.50-2.60 (m, 2H), 3.86 (s, 3H), 3.90 (s, 3H), 4.19 (t, 2H, J= 5.5 Hz), 5.73-5.77 (m, 1H), 6.21-6.27 (m, 1H), 6.44-6.50 (m, 1H), 6.95 (s, 1H), 7.11-7.53 (overlapping m, 3H), 7.90 (s, 1H), 8.27-8.30 (overlapping m, 3H), 8.55 (s, 1H), 8.84 (s, 1H), 9.84 (s, 1H) ppm; C27H30N6O3 m/z MH+ 487.
Ëxample 2 A/-(2-((2-(Dimethylamino)ethyl)thio)-4-methoxy-5-((4-(l-methyl-lH-indol“3yl)pyrimidin-2-yl)amino)phenyl)acrylamide (2)
Scheme 2
7V-(4-((2-(Dimethylamino)ethyl)thio)-2-methoxy-5-nitrophenyl)-4-(l-methyI-lH-indol3-yl)pyrimidm-2-amine (Scheme 2, Intermediate D). To a slurry of NaH (54 mmol, 60% oil dispersion prewashed with hexanes) and 25 mL of DMF was added a slurry of 2dimethylaminoethanethiol hydrochloride (27 mmol) in 25 mL of DMF under a stream of N2. After stirring for 45 min, a slurry of A (5.4 mmol) in 25 mL of DMF was added portionwise over 15 min to the mixture under a stream N2. The resulting mixture was stirred ovemight, then poured into water and the solid was collected, rinsed repeatedly with water, and dried under vacuum to yield 2.5 g of product as a yellow solid. C24H26N6O3S m/z MH+ 479.
4-((2-(Dimethylammo)ethyl)thio)-6-methoxy-AT1-(4-(l-methyl-lH-indol-3-yl)pyrimidin2-yl)benzene-l,3-diamine (Scheme 2, Intermediate E). A suspension of 2.5 g of Intermediate D, 3.0 g of Fe°, 50 mL of éthanol, 20 mL of water, and 7 mL of conc. HCl was heated to 78° C for 3 h. The solution was cooled to room température, adjusted to pH 10 with 10% NaOH (aq), and diluted with CH2CI2. The mixture was fïltered through Dicalite, and the fïltrate layers were separated. The aqueous phase was extracted with CH2CI2 twice, and the combined organic extracts were dried over Na2SO4 and concentrated. Column chromatography (silica gel, CH2C12MeOH gradient) afforded 1.2 g of Intermediate E as a solid. C24H28N6OS m/z MH+ 449.
7V-(2-((2-(Dimethylamino)ethyI)thio)-4-methoxy-5-((4-(l-methyl-lH-mdoI-3yl)pyrimidin-2-yl)amino)phenyl)acrylamide (2). To a solution of Intermediate E (2.7 mmol) in 50 mL of THF and 10 mL of water was added 3-chloropropionychloride (4.0 mmol) dropwise with stirring. After 2 h of stirring, NaOH (27 mmol) was added and the mixture was heated at 65°C for 18 h. After cooling to room température, THF was partially removed under reduced pressure, and the mixture was extracted with CH2C12, dried over Na2SO4, and concentrated. Chromatography of the crude product (silica gel, CH2C12-MeOH-NH4OH gradient) afforded 0.622 g of Example 2 as an off-white solid: *H NMR (300 MHz, DMSO) δ 2.19 (s, 6H), 2.34 (t, 2H, 6.5 Hz), 2.98 (t, 2H, J = 6.5 Hz), 3.91 (s, 3H), 3.93 (s, 3H), 5.50-6.57 (overlapping m,
3H), 7.12-9.88 (overlapping m, 10H), 10.17 (s, 1H) ppm. C27H30N6O2S m/z MH+ 503.
Example 3
7V-(2,4-Dimethoxy-5-((4-(l-methyl-lH-indol-3-yl)pyrimidin-2-yl)amino)phenyl)acrylamide (3)
Scheme 3
NaOMe
MeOH, reflux \
F
SnCI2 H2O
EtOAc, relux
2) NaOH (aq.)
THF, 50 °C
2V-(2,4-Dïmethoxy-5-nitrophenyl)-4-(l-methyl-lff-indol-3-yl)pyrimidin-2-amine (Scheme 3, Intermediate F). Sodium methoxide, 25wt.% solution in methanol (40 niL. 175 mmol), was slowly poured into a stirred, ambient température, suspension of 7V-(4-fluoro-2methoxy-5-nitrophenyl)-4-(l-methyl-17/-indol-3-yl)pyrimidin-2-amine (Scheme 1, Intermediate A; 5.8 g, 14.7 mmol) in methanol (125 mL) and heated at reflux for 4 days under nitrogen blanket, during which time the solid did not dissolve. The reaction was cooled, product precipitate isolated by filtration, washed with cold methanol, and dried to yield 5.45 g of JV-(2,4-dimethoxy-
5-nitrophenyl)-4-(l-methyl-li/-indol-3-yl)pyrimidin-2-amine (Intermediate F) as a yellow powder. C2iH19N5O4 m/z MH+ 406.
4,6-Dimethoxy-2V1-(4-(l-methyI-lH-indol“3“yl)pyrimidin-2“yl)benzene-l,3-diamine (Scheme 3, Intermediate G). Stannous chloride dihydrate (8.9 g, 39.4 mmol) was added to a stirred, ambient température suspension of lV-(2,4-dimethoxy-5-nitrophenyl)-4-(l-methyl-l/Zindol-3-yl)pyrimidin-2-amine (Intermediate F; 3.2 g, 7.9 mmol) in ethyl acetate (200 mL) and heated at reflux under nitrogen blanket for 3 h. The reaction was allowed to cool, then poured into a 5% (w/v) solution of sodium bicarbonate in DI water (400 mL) and stirred for 1 h. The multiphase mixture was then filtered through tightly packed Celite, with ethyl acetate rinsing of the filter cake. The filtrate was transferred to a separatory funnel and the liquid phases separated. The retained ethyl acetate solution of product was washed with brine and dried over anhydrous calcium sulfate. Filtration and évaporation yielded 1.6 g of crude product. Purification by gradient flash chromatography (SiO2, 0 to 70% hexanes / ethyl acetate over 20 min.) provided 0.9 g of 4,6-dimethoxy-.Vl-(4-(l-methyl-177-indol-3-yl)pyrimidin-2-yl)benzene-l,3-diaminc (Intermediate G) as a yellow foam. C2iH21N5O2 m/z MH+ 376.
7V-(2,4-Dimethoxy-5-((4-(l-methyl-lff-indol-3-yl)pyrimidin-2yl)amino)phenyl)acrylamide (3). 3-ChIoropropanoyl chloride (90 pL, 0.92 mmol) was rapidly added by syringe to a rapidly stirred, atnbient température, nitrogen blanketed solution of 4,6dimethoxy-jV1 -(4-(1 -methyl- l/7-indol-3 -yl)pyrimidin-2-yl)benzene-1,3 -diamine (Intermediate G; 351 mg, 0.94 mmol) and Λ-methylmorpholine (0.11 mL, 1.0 mmol) in ethyl acetate (9.4 mL), precipitate immediately formed, and reaction was allowed to proceed for 40 min., evaporated to dryness, and dissolved in 10% (v/v) DI water / tetrahydrofuran. Solid sodium hydroxide (3 g, 75 mmol) was added and the stirred mixture heated to 50 °C for 17 h. The reaction solution was cooled, partitioned between brine and ethyl acetate. The ethyl acetate phase was dried over anhydrous calcium sulfate, filtered, and then chilled in an ice bath with stirring while slowly being diluted with hexanes to precipitate the product. This material was isolated by filtration and dried to provide 189 mg of Example 3 as fine light-yellow powder. ‘H NMR (300 MHz, DMSO) Ô 3.88 (s, 6H), 3.90 (s, 3H), 5.70 (dd, 1H, J= 10.15, 1.92 Hz), 6.22 (dd, 1H, J= 16.95, 2.03 Hz), 6.70 (q, 1H, J= 9.06 Hz), 6.85 (s, 1H), 7.11-7.17 (m, 2H), 7.23 (t, 1H, J= 6.96 Hz), 7.50 (d, 1H, J= 8.23 Hz), 7.93 (s, 1H), 8.28 (m, 2H), 8.47 (s, 1H), 8.67 (s, 1H), 9.38 (s, 1H) ppm. 13C NMR (75 MHz, DMSO) δ 33.4, 56.5, 56.7, 97.3, 107.1, 110.8, 113.0, 118.5, 119.5, 121.3, 121.5, 122.3, 122.5, 125.9, 126.4, 132.8, 133.8, 138.1, 147.3, 148.3, 157.8, 160.8, 162.3, 163.5 ppm. C24H23N5Û3 m/z MH+ 430.
Example 4 JV-(2-(3-(Dimethylamino)propyl)-4-methoxy-5-((4-(l-methyl-Lff-indoI-3-yl)pyrimidin-2yl)amïno)phenyl)acrylamîde (4)
Scheme 4
V-(4-(3-(Dimethylammo)prop-l-yn-l-yl)“2-methoxy-5-nitrophenyl)-4-(l-methyl-lHindoI-3-yl)pyrimidin-2-amine (Scheme 4, Intermediate H). A solution of 3-dimethylamino-lpropyne (1.37 mL,12.7 mmol) in 1,4-dioxane (60 mL) was treated with 1 M lithium bis(trimethylsilyl)amide (12.7 mL, 12.7 mmol) and stirred for 30 min at RT under a nitrogen atmosphère. The resulting reaction mixture, which appeared as a white slurry, was treated with A “(4-fluoro-2-methoxy-5 -nitrophenyl)-4-( 1 -methyl-1 H-indol-3 -yl)pyrimidin-2-amine (Intermediate A; 1.00 g, 2.54 mmol) in one portion and heated at 80 °C while vigorously stirring under nitrogen for 5 h, The reaction mixture was cooled to RT, quenched by the addition of 10 mL of water and subsequently concentrated in vacuo. The residue was partitioned between water (100 mL) and CH2C12 (50 mL). The basic aqueous layer was extracted with CH2C12 (2 x 50 mL) and the combined organic extracts were washed with brine (2 x 50 mL), dried (Na2SO4), filtered and concentrated in vacuo to fumish 1.0 g of the crude product as a dark reddish-brown solid. This material was purified by gradient flash chromatography on SiO2 eluting with 0 to 10% methanol (containing 2% NH4OH) in CH2C12 over 60 min to afford 98 mg of A-(4-(3(dimethylamino)prop-1 -yn-1 -yl)-2-methoxy-5 -nitrophenyl)-4-( 1 -methyl- lH-indol-3 yl)pyrimidin-2-amine (Intermediate H) as an orange solid. C25H24N6O3 m/z MH+ 457.
4-(3-(Dimethylamino)propyl)-6“mcthoxy-Ar,-(4-(l-methyl-lZ7-indol-3-yl)pyrimidin-2yl)benzenc-l,3-diamine (Scheme 4, Intermediate I). 10%Pd/C (10 mg) was added under a nitrogen atmosphère to a solution of A'-(4-(3-(dimethylamino)prop-l-yn-l-yl)-2-methoxy-5nitrophenyl)-4-(I-methyl-lH-indol-3-yl)pyrimidin-2-amine (Intermediate H; 50 mg, 0.109 mmol) in 10 mL of THF/methanol (1:1). A hydrogen-filled balloon was connected to the reaction vessel and the reaction was stirred at RT under a hydrogen atmosphère for 6 h. The reaction mixture was filtered through Celite 545 and concentrated in vacuo to give 50 mg of crude product, This material was purifîed by gradient flash chromatography on SiO2 eluting with 0 to 10% methanol (containing 2% NH4OH) in CH2CI2 over 50 min to afford 34 mg of 4-(3-(dimethylamino)propyl)-
6-methoxy-TV1 -(4-(1 -methyl- l//-indol-3-yl)pyrimidin-2-yl)benzene-1,3 -diamine (Intermediate I) as a foam. C25H30N(lO m/z MH+ 431.
/V-(2-(3-(Dimetliylamino)propyl)-4-inethoxy-5-((4-(l-methyl-lZ7-indol-3-yl)pyrimidm2-yl)amino)phenyl)acrylamide (4). 3-Chloropropanoyl chloride (18.2 pL. 0.190 mmol) was rapidly added to a solution of 4-(3-(dimethylamino)propyl)-6-methoxy-7V1 -(4-(1 -methyl- 1Hindol-3-yl)pyrimidin-2-yl)benzene-1,3-diamine (Intermediate I; 34 mg, 0.079 mmol) in 3.2 mL of THF/water (9:1) while stirring under nitrogen at RT. After 3 h, IM aq NaOH (0.79 mL, 0.79 mmol) and the reaction mixture was heated at 65 °C for 17 h. The reaction mixture was cooled to RT, diluted with water (15 mL) and the resulting light gray precipitate was isolated by filtration to give 31 mg of crude product. This material was purifîed by gradient flash chromatography on SiO2 eluting with 0 to 10% methanol (containing 2% NH4OH) in CH2C12 over 35 min to afford 22 mg of Example 4 as an off-white solid. ‘H NMR (300 MHz, CDC13) δ 1.81-1.92 (m, 2H), 2.16 (t, 2H, J= 5.9 Hz), 2.27 (s, 6H), 2.69 (t, 2H, J= 6.3 Hz), 3.89 (s, 3H), 3.98 (s, 3H), 5.71 (dd, 1H, J= 10.1, 1.9 Hz), 6.25 (dd, 1H, J= 16.9, 10.1 Hz), 6.48 (dd, 1H, J= 16.9, 1.9 Hz), 6.66 (s, 1H), 7.17 (d, 1H, J= 5.3 Hz), 7.22-7.43 (m, 3H), 7.72 (s, 1H), 8.05-8.12 (m, 1H), 8.37 (d, 1H, J= 5.3 Hz), 8.85 (s, 1H), 9.33 (s, 1H), 10.95, (br s, 1H); C28H32N6O2 m/z MH+ 485.
Examples 5,6, and 7
Scheme 5
(Me)3Sn
Pd2(dba)3
P(o-tol)3
A-(2-(2-(Dimcthylamino)ethoxy)-4-mcthoxy-5-((4-(2“mcthyl-2//-indazol-3-yl)pyriniidin-2yl)amino)phenyl)acrylamide (5)
5 jV-(2-((2-(Dimethylamino)ethyl)thio)-4-methoxy-5-((4-(2-methyl-2/7-indazol-3-yl)pyrimidin-
2-yl)amino)phenyl)acrylamide (6)
A-(2,4-Dimcthoxy-5-((4-(2-mcthyl-2tf-indazol-3-yl)pyrimidin-2 yl)amino)phenyl)acrylamide (7)
The synthesis of 7V-(4-fluoro-2-methoxy-5-nitrophenyl)-4-(2’methyl-2//-indazol-3yl)pyrimidin-2-amine (Intermediate J) is shown above in Scheme 5. Examples 5, 6, and 7 are 15 prepared as in Schemes 1, 2, and 3, respectively, by substituting Intermediate J for Intermediate A in each of those schemes.
Example $
7V-(5-((4-(l,2-Dimethyl-lH-Îndol-3-yl)pyrimidin-2-yl)amino)-2-(2(dimethylamino)ethoxy)-4-methoxyphenyl)acrylamide (8)
Scheme 6
K
p-TsOH H2O
1,4-dioxane reflux
N 8
3-(2-Chloropyrimidin-4-yl)-l,2-dimethyl-lH-indole (Scheme 6, Intermediate K). Ferrie chloride (5.8 g, 34.7 mmol) was rapidly added to a degassed, clear yellow solution of 1,2dimethyl-177-indole (4.9 g, 33.8 mmol) and 2,4-dichloropyrimidine (5.2 g, 33.9 mmol) dissolved in anhydrous 1,2-dimethoxyethane (100 mL) while stirring at the ambient température. The résultant black, opaque solution was stirred at ambient température for 3 h under dry nitrogen atmosphère, then slowly poured into rapidly stirred 5% (w/v) aqueous NaHCO3 (400mL). Crude product was isolated by filtration, and washed with DI water on the filter. The precipitate was suspended in methanol (200 mL) and evaporated to dryness to remove excess water, then triturated in hot acetonitrile, allowed to cool, and filtered to isolate 6.2 g of 3-(2-chloropyrimidin4-yl)-l,2-dimethyl-127-indole (Intermediate K) as a brown powder. *H NMR (300 MHz, DMSO) Ô 2.77 (s, 3H), 3.79 (s, 3H), 7.23 (quin, 2H, J = 7.53 Hz), 7.57 (d, 1H, J = 7.25 Hz), 7.72 (d, 1H, J = 5.61 Hz), 8.10 (d, 1H, J = 7.46 Hz), 8.61 (d, 1H, J = 5.43 Hz) ppm. 13C NMR (75 MHz, DMSO) δ 12.8, 30.3, 108.8, 110.8, 117.5, 120.0, 121.8, 122.5, 125.8, 137.4, 142.6, 159.8, 160.4, 165.2 ppm. C14H12C1N3 m/z MH+258.
4-(1,2-Dimcthyl-17/-indol-3-yl)-A-(4-fluoro-2-methoxj’-5-nitrophenyl)-pyrimidin-2amine (Scheme 6, Intermediate L). Reagent grade 1,4-dioxane (57 mL) was added to a mixture of 3-(2-chIoropyrimidin-4-yl)-l,2-dimethyl-l/7-indolc (1.47 g, 5.70 mmol), 4-fluoro-2-methoxy5-nitroaniline (1.06 g, 5.69 mmol), and p-toluenesulfonic acid monohydrate (1.31 g, 6.89 mmol) contained in a 100 mL round bottom flask fitted with a reflux condenser and blanketing nitrogen inlet. The magnetically stirred suspension was heated to reflux under nitrogen blanket. While approaching reflux température the suspended solid dissolved. Reflux was continued ovemight, then the reaction was cooled and poured into rapidly stirring DI water (250 mL) to precipitate the product. Crude product was isolated by filtration, washed with water and recrystallized from boiling 2-propanol to yield 2.06 g of 4-(l,2-dimethyl-li/-indol-3-yl)-A-(4-fluoro-2-methoxy-5nitrophenyl)-pyrimidin-2-amine (Intermediate L) as a fine yellow powder. JH NMR (300 MHz, DMSO) δ 2.71 (s, 3H), 3.78 (s, 3H), 4.01 (s, 3H), 7.10-7.20 (m, 3H), 7.41 (d, 1H, J = 13.4 Hz), 7.55 (d, 1H, J = 7.99 Hz), 7.98 (d, 1H, J = 7.90 Hz), 8.44 (d, 1H, J = 5.70 Hz), 8.83 (br s, 1H), 8.93 (d, 1H, J = 8.38 Hz). C2iHi8FN5O3 m/z MH+ 408.
4-(l,2-Dimethyl-lff-indol-3-yl)-7V-(4-(2-(dimethylamino)ethoxy)-2-methoxy-5nitrophenyl) pyrimidin-2-amine (Scheme 6, Intermediate M). 2-Dimethylaminoethanol (0.43 mL, 4.27 mmol) was added, by syringe over 5 min., to a stirred suspension of 60 wt.% sodium (173 mg, 4.33 mmol) in anhydrous 1,4-dioxane at the ambient température. Gas évolution was readily observed. After ten min., with no further observable gas évolution, 4-(l,2-dimethyl-l/7indol-3-yl)-7V-(4-fluoro-2-methoxy-5-nitrophenyl)-pyrimidin-2-amine (Intermediate J) (351 mg, 0.86 mmol) was added, neat, to the rapidly stirred pot as one bolus. The reaction suspension immediately changed to a turbid red-brown color. After 5 min. an aliquot of the reaction was withdrawn, quenched into DI water, and extracted into ethyl acetate. Analysis of this extract by UHPLC-MS revealed the reaction to be complété. The pot contents were then poured into a stirred solution of ammonium chloride (0.23 g, 4.30 mmol) in DI water (150 mL) to precipitate the product. The yellow precipitate was isolated by filtration, washed with DI water, and allowed to dry to afford 386 mg of 4-(l,2-dimethyl-177-indol-3-yl)-7V-(4-(2-(dimethylamino)ethoxy)-2methoxy-5-nitrophenyl) pyrimidin-2-amine (Intermediate Μ). C^NA m/z MH+ = 477.
jV1-(4-(l,2-Dimethyl-lZZ-indol-3-yl)pyrimidin-2-yl)-4-(2-(dimethylamino)-ethoxy)-6methoxybenzene-l,3-diamine (Scheme 6, Intermediate N). Stannous chloride dihydrate (1.73 g, 7.67 mmol) was added to a stirred suspension of 4-(l,2-dimethyl-12f-indol-3-yl)-A'-(4-(2(dimethyl-amino)ethoxy)-2-methoxy-5-nitrophenyl) pyrimidin-2-amine (Intermediate M; 386 mg,
0.8 Immol) in ethyl acetate (40 mL) at the ambient température, and the mixture was heated at reflux under nitrogen blanket for 17h. The reaction was allowed to cool, then poured into a 1% (w/v) solution of sodium hydroxide in DI water (200 mL) and stirred for 1 h, The multiphase mixture was filtered through tightly-packed Celite, with ethyl acetate rinsing of the fil ter cake. The fïltrate was transferred to a separatory funnel and the liquid phases were separated. The retained ethyl acetate solution of product was washed with brine, dried over anhydrous calcium sulfate, filtered and evaporated to provide a brown solid foam which was purified by gradient flash chromatography (SiO2, 2% NH4OH in MeOH / ethyl acetate, 0 to 20% over 40 min.) to provide 186 mg of 2V1-(4-(l,2-dimethyl-l/f-indol-3-yl)pyrimidin-2-yl)-4-(2-(dimethylamino)ethoxy)-6-methoxybenzene-l,3-diamine (Intermediate N) as yellow solid. *H NMR (300 MHz, DMSO) δ 2.34 (s, 6H), 2.70 (t, 2H, J = 6.90 Hz), 2.75 (s, 3H), 3.58 (br s, 2H), 3.74 (s, 3H), 3.83 (s, 3H), 4.07 (t, 2H, J = 5.34 Hz), 6.57 (s, 1H), 6.95 (d, 1H, J = 5.19 Hz), 7.17-7.27 (m, 2H), 7.327.35 (m, 1H), 7.55 (s, 1H), 8.09 (dd, 1H, J = 6.96, 1.77 Hz), 8.18 (s, 1H), 8.38 (d, 1H, J = 5.22 Hz)ppm. C25H30N6O2 m/z MH+= 447.
7V-(5-((4-(l,2-DimethyI-l77-Îndol-3-yl)pyrirnidin-2-yl)amino)-2-(2-(dimethylammo)ethoxy)-4-methoxyphenyl)acrylamide (8). .V1 -(4-( 1,2-Dimethyl -177-indol-3 yl)pyrimidin-2-yl)-4-(2-(dimethylamino)ethoxy)-6-methoxy-benzene-l ,3-diamine (Scheme 6, Intermediate N) is converted into Example 8 by reaction with 3-chloropropionychloride followed by treatment with NaOH by using the procedures described in the préparation of Example 1.
Example 9 7V-(2-(2-(Dimethylammo)ethoxy)-4-methoxy-5-((4-(pyrazolo[l,5-û!]pyridin-3yl)pyrimidin-2-yl)amino)phenyl)acrylamïde (9)
Scheme 7
NaH 1,4-dioxane
Q
2) NaOH
2V-(4-(2-(Dimethylamino)ethoxy)-2-methoxy-5-nitrophenyl)-4-(pyrazolo[l,5-a]pyridin-
3-yl) pyrimidin-2-amine (Scheme 7, Intermediate P), To a slurry of NaH (21 mmol, 60% oil dispersion prewashed with hexanes) and 20 mL of 1,4-dioxane was added 2dimethylaminoethanol (20 mmol, 2.4 mL) dropwîse with stirring under N2. After stirring for 45 min, a slurry of compound O (7.9 mmol) was added portion-wise, with stirring and under a stream of N2. The resulting mixture was stirred ovemight, then poured into water and the solid was collected, rinsed with water, and dried under vacuum to yield 1.7 g of Intermediate P as a yellow solid, which was used in the next step without further purification: C22H23N7O4m/z MH+ 450.
4-(2-(Dimethylamino)ethoxy)“6-methoxy-7Vl-(4-(pyrazoIo[l,5-a]pyridin-3-yl)benzene)1,3-diamine (Scheme 9, Intermediate Q). A suspension of 0.7 g of Intermediate P, 0.9gofFe°, 7 mL of éthanol, 3 mL of water, and 2 mL of glacial acetic acid was heated to 78 °C for 1 h. The solution was cooled to room température, filtered through Dicalite, adjusted to pH 10 with 1 N NaOH (aq) and diluted with CH2C12. The filtrate layers were separated, and the aqueous phase was extracted with CH2C12 twice, and the combined organic extracts were dried over Na2SQ4 and concentrated. Column chromatography (silica gel, CH2Cl2-MeOH gradient) afforded 0.28 g of Intermediate Q as tan solid. C22H25N7O2 m/z MH+ 420.
7V-(2-(2-(Dimethylamino)ethoxy)-4-methoxy-5-(4-(pyrazolo[l,5-«]pyridin-3yl)benzene)-acrylamide (9). To a solution of Intermediate Q (0.6 g, 1.4 mmol) in 10 mL of THF and 4 mL of water was added 3-cbloropropionychloride (0.15 mL, 1.6 mmol) dropwîse with stirring. After 22 h of stirring, NaOH (0.7 g, 17 mmol) was added and the mixture was heated at 65°C for 5 h. After cooling to room température, THF was removed under reduced pressure, and the mixture was extracted with CH2C12, dried over Na2SO4, and concentrated. Chromatography of the crude product (silica gel, CH2Cl2-MeOH) afforded 0.294 g of Example 9 as a beige solid. C25H27N7O3 m/z MH+ 474. *H NMR (300 MHz, DMSO) δ 2.28 (s, 6H), 2.61-2.62 (m, 2H), 3.82 (s, 3H), 4.20-4.22 (m, 2H), 5.69-5.73 (m, 1H), 6.20-6.22 (m, 1H), 6.42-6.48 (m, 1H), 6.90-7.11 (m, 2H), 7.15-7.40 (m, 2H), 8.10-8.59 (overlapping m, 4H), 8.72-8.96 (m, 2H), 10.13 (s, 1H) ppm.
Example 10 iV-(2-(2-(DimethylamÎno)ethoxy)-4-methoxy-5-((4-(l-methyI-l//-indazol-3-yl)pyriniidin-2yl)amino)phenyl)acrylamide (10) jV-(4-Fluoro-2-methoxy-5-nitrophenyl)-4-(l-methyl-lH-indazol-3-yl)pyrimidin“2-amine (Scheme 8, Intermediate R). Into a 1000-mL 3-necked round-bottom flask purged and maintained with an inert atmosphère of nitrogen, was placed a solution of IH-indazole (10 g, 84.65 mmol, 1.00 equiv) in A^V-dimethylformamide (500 mL), I2 (21.5 g, 84.65 mmol, 1.00 equiv), This was followed by the addition of KOH (19 g, 338.62 mmol, 4.00 equiv) in several batches at 0°C. The resulting solution was stirred ovemight at room température. The reaction was then quenched by the addition of 200 mL of aqueous Na2S2O3. The resulting solution was extracted with 3x500 mL of ethyl acetate and the organic layers combined. The resulting mixture was washed with 3x500 mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The resulting mixture was washed with 1x100 mL of hexane. This resulted in 14 g (68%) of 3-iodo-lH-indazole as a white solid.
Scheme 8
KOH, lz,DMF
RT,overnigh1
68%
I
CHjl.NaH (65%)
THF
RT, 1 h
-SnSn- (HsCfeSn
---------------- N
Pd(PPh3)4,1,4-dioxane
100 °C, 6 h 1
54%
68%
Into a 500-mL 3-necked round-bottom flask purged and maintained with an inert atmosphère of nitrogen, was placed a solution of 3-iodo-lH-indazole (14 g, 57.37 mmol, 1.00 equiv) in tetrahydrofuran (200 mL). This was followed by the addition of NaH (65%) (2.5 g, 1.20 equiv) in several batches at 0°C. The mixture was stirred for 30 min at 0°C. To this was added iodomethane (9.7 g, 68.34 mmol, 1.20 equiv) dropwise with stirring at 0°C. The resulting solution was stirred for 1 h at room température. The reaction was then quenched by the addition of 300 mL of water/ice. The resulting solution was extracted with 2x300 mL of ethyl acetate and the organic layers combîned. The resulting mixture was washed with 1x300 mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:5). This resulted in 8 g (54%) of 3iodo-l-methyl-lH-indazole as a yellow solid
Into a 500-mL 3-necked round-bottom flask purged and maintained with an inert atmosphère of nitrogen, was placed a solution of 3-iodo-l-methyl-lH-indazole (5 g, 19.38 mmol, 1.00 equiv) in 1,4-dioxane (200 mL), hexamethyldistannane (12 g, 36.63 mmol, 2.00 equiv), tetrakis(triphenylphosphane) palladium (2.2 g, 1.90 mmol, 0.10 equiv). The resulting solution was stirred for 6 h at 100°C. The reaction mixture was cooled to room température with a water/ice bath. The reaction was then quenched by the addition of 30 mL of aqueous KF (1 N) dropwise with stirring. The resulting solution was stirred for 0.5 h at room température. The resulting solution was diluted with 200 mL of H2O. The resulting solution was extracted with 2x200 mL of ethyl acetate and the organic layers combined. The resulting mixture was washed with 3x200 mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:5). This resulted in 3.9 g (68%) of l-methyl-3-(trimethylstannyl)-lH-indazole as yellow liquid.
Into a 250-mL 3-necked round-bottom flask purged and maintained with an inert atmosphère of nitrogen, was placed l-methyl-3-(trimethylstannyl)-lH-indazole (3.9 g, 13.22 mmol, 1.00 equiv), 1,4-dioxane (100 mL), 2,4-dichloropyrimidine (2.0 g, 13.42 mmol, 1.00 equiv), tetrakis(triphenylphosphane) palladium (1.5 g, 1.30 mmol, 0.10 equiv). The resulting solution was stirred ovemight at 105°C. The reaction mixture was cooled to room température with a water/ice bath. The reaction was then quenched by the addition of 200 mL of water/ice. The solids were collected by filtration. The fil ter cake was washed with 1x100 mL of Eî2O. This resulted in 2.1 g (65%) of 3-(2-chloropyrimidin-4-yl)-l-methyl-lH-indazole as a light yellow solid.
Into a 250-mL 3-necked round-bottom flask, was placed 3-(2-chloropyrimidin-4-yl)-lmethyl-lH-indazole (2,9 g, 11.85 mmol, 1.00 equiv), 4-fluoro-2-methoxy-5-nitroaniline (2.2 g, 11.82 mmol, 1.00 equiv), 2-propanol (80 rnL), TsOH (2.4 g, 13.94 mmol, 1.20 equiv). The resulting solution was stirred ovemight at 80°C. The reaction mixture was cooled to room température with a water/ice bath. The solids were collected by filtration. The fîlter cake was washed with 100 mL of CHjCN. The solid was dried in an oven. This resulted in 1.06 g (23%) of A-(4-fluoro-2-methoxy-5-nitrophenyl)-4-(l-methyl-lH-indazol-3-yl)pyrimidin-2-amine (Intermediate R) as a yellow solid. (ES, îm/z): [M+H]+=395; ’H-NMR (300MHz, DMSO-c4,) δ 8.96 (br, 1H), 8.87-8.85 (d, J=8.4Hz, 2H), 8.56-8.54 (d, J=5.4Hz, 1H), 8.49-8.46 (d, J=8.1Hz,
1H), 7.77-.775 (d, J=8.4Hz, 1H),7.58-7.57 (d, J=5.1Hz, 1H), 7.52-7.47 (t, J=7.2Hz, 1H), 7.447.40 (d, >13.5Hz, 1H), 7.26-7.21 (t, >7.5Hz, 1H), .4.19 (s, 1H), 4.01 (s, 1H) ppm.
A-(4-(2-(Dimethylamino)ethoxy)-2-methoxy-5-nitrophenyl)-4-(l-methyl-Lff-indazol-3yl)pyrimidine-2-amine (Scheme 8, Intermediate S). To a suspension of NaH (31 mg, 1.3 mmol) in 10 mL of 1,4-dioxane was added 2-dimethylaminoethanol (0.16 mL, 1.3 mmol) dropwise with stirring under N2. After stirring for 1.5 h, Intermediate R (0.2 g, 0.51 mmol) was added portionwise. After 0.5 h, the reaction mixture was quenched with water and extracted with CH2CI2. The organic phase was dried over Na2SO4, filtered and concentrated to yield 0.23 g of A-(4-(2-(dimethylamino)ethoxy) -2-methoxy-5 -nitrophenyl)-4-( 1 -methyl- 177-indazol-3 20 yl)pyrimidine-2-amine (Intermediate S): m/z MH+ = 464.
4-(2-(Dimethylamino)ethoxy)-6-methoxy-Arl-(4-(l-methyI-lH-indazol-3yl)pyrimidm2-yI)bcnzcnc-l,3-diaminc (Scheme 8, Intermediate T). A suspension of 7V-(4-(2(dimethylamino)ethoxy)-2-methoxy-5-nitrophenyl)-4-(l-methyl-l//-indazol-3-yl)pyrimidine-225 amine (0.23 g), 0.28 g of Fe°, 10 mL of 70% ethanol/H2O, and 0.5 mL of acetic acid was heated at reflux with stirring for 2 h. The mixture was cooled to room température, then filtered. The filtrate was adjusted to pH 10, then extracted with CH2C12. The organic phases were combined, dried over Na2SO4, filtered and concentrated. The crude product was purified by chromatography (silica gel, CH2C12-1% NH4OH/MeOH gradient) to afford 4-(2-(dimethylamino)ethoxy)-630 methoxy-vV1 -(4-(1 -methyl-lH-îndazol-3yl)pyrimidin-2-yI)benzene-l,3-diamine (Intermediate T) as an off-white solid: m/z MH+ 434.
Ar-(2-(2-(Dimethylamino)ethoxy)-4-methoxy-5-((4-( 1 -methyl- lH-indazol-3- ;
i yl)pyrimidin-2-yl)amino)phenyl)acrylamide (Example 10). To a solution of 4-(233
I !
I !
’ i (dimethylamino)ethoxy)-6-methoxy-M-(4-(l-methyl-lH-indazol-3yl)pyrimidin-2-yl)benzene1,3-diamine (60 mg, 0.14 mmol) dissolved in 10 mL of 4:1 THF:H2O was added 3chloropropionyl chloride (17 mg, 0.14 mmol). After 4h, NaOH (1.4 mmol, 56 mg) was added and the mixture was heated at reflux for 5 h. THF was removed under reduced pressure, and the aqueous phase was extracted with ethyl acetate. The organic phases were combined, washed with H2O, dried (Νη24) and concentrated. The crude product was purified by chromatography (silica gel, CH2Cl2-MeOH gradient) to afford Example 10 as a solid: C^H^NyOj; m/z MH+ 48S; Ή NMR (300 MHz, DMSO) δ 2.28 (s, 6H), 2.51-2.63 (m, 2H), 3.80 (s, 3H), 4.14-4.44 (overlappingm, 5H), 5.68-5.76 (m 1H), 6.11-6.19 (m, 1H), 6.43-6.48 (m, 1H), 6.95 (s, 1H), 7.117.17 (m, 1H), 7.37-7.45 (overlapping m, 2H), 7.68-7.07 (d, 1H, J = 8.4 Hz ), 8.39-8.43 (overlappingm, 4H), 9.75 (s, 1H) ppm.
Example 11 ;V-(2-(2-(Dimethy]amino)ethoxy)-5-((4-(l-(2-iluorocthyl)-lH-indolvl-3-yl)pyrimidin-2yl)amino)-4-methoxyphenyl)acrylamide (11) l-(2-FluoroethyI)-lH-indole (Scheme 9). Sodium hydride, 60 wt.% in oil (2.3g, 57.5 mmol) was added to stirred, 0°C, clear, colorless solution of indole (10.1g, 86.2 mmol) in anhydrous tetrahydrofuran at as rapid a rate consistent with maintaining control of the concomitant hydrogen évolution. Solution was stirred at 0°C under N2 blanket until gas évolution ceased, and reaction had become a fine white suspension. A solution of l-fluoro-2-iodoethane (5 g, 29 mmol) in anhydrous tetrahydrofuran (6mL) was then slowly added via syringe, the ice bath was removed and the pot heated to reflux ovemight. The reaction mixture was cooled, diluted with a solution of ammonium chloride (4.6g, 86 mmol) in DI water (300mL), transferred to a separatory funnel, and extracted with ethyl acetate. The extract was dried (CaSCL) and evaporated to provide a yellow oil, which was flash chromatographed (silica gel, 100% hexanes) to provide 4.2g of yellow oil, characterized by LC-MS as a 60/40 mixture of indole to desired product. This impure product was treated with benzene sulfonyl chloride to modify the elution characteristics of the mixture to allow for isolation of the desired product as follows: To a 0aC solution of the above isolated 60/40 mixture of indole to desired product and tetrabutyl ammonium bisulfate 1.2 g, 3.4 mmol) in anhydrous toluene (lOOmL) was added a solution of sodium hydroxide (24.7g, 617.5 mmol) in DI water (25 mL). To the rapidly stirred, 0°C, mixture was then added benzene sulfonyl chloride (5.5 mL, 43.1 mmol) and the reaction allowed to stir and warm to ambient température under N2 blanket ovemight.
Scheme 9
1. NaH, 0°C, 0.5h 2.1-fluoro-2-iodoethane
FeCI3; DME 60°C, 17h
Cl
THF
The reaction mixture was then partitioned between ethyl acetate and DI water, the organic phase dried (CaSO4) and flash chromatographed (silica gel, 10% acetone / hexanes) to cleanly 5 résolve the 1-phenylsulfonyl indole from the desired product affording 1.3g of 1-(2-fluoroeth.yl)IH-indole as a clear, colorless liquid. *H NMR (300 MHz, DMSO) δ 4.45 (t, 1H, J=4.9Hz), 4.54 (t, 1H, J=4.9Hz), 4.64 (t, 1H, J=4.6Hz), 4.80 (t, 1H, J = 4.4Hz), 6.46 (dd, 1H, J = 3.1, 0.8Hz), 7.03 (m, 1H), 7.13 (m, 1H), 7.37 (d, 1H, J=3.2Hz), 7.49 (d, 1H, J=8.3Hz), 7.55 (m, 1H) ppm. 13C NMR (75 MHz, DMSO) δ 46.4 (d, Jcl^19.5IIz), 83.3 (d, JCF =166.5Hz), 101.4, 110.3, 119.6, 10 120.9, 121.6, 128.6, 129.3, 136.4 ppm. C10Hi0NF m/z MH+ 164.
3-(2-Chloropyrimidin-4-yl)-l-(2-fluoroethyl)-lH-indole (Scheme 9, Intermediate U). Ferrie chloride (1.3 g, 7.9 mmol) was rapidly added to a stirring, ambient température, degassed, clear, colorless solution of l-(2-fluoroethyl)-lH-indole and 2,4-dichloropyrimidine (1.2 g, 8.3 mmol) dissolved in anhydrous 1,2-dimethoxyethane (80mL). The résultant black, opaque, solution was stirred at 60°C for 17 h under dry nitrogen atmosphère, cooled, and partitioned between ethyl acetate and saturated aqueous sodium chloride. The organic phase was dried (CaSO4) and evaporated to provide 2.3g of purple oïl which was purified by flash ebromatography (silica gel, 0 to 90% ethyl acetate in hexanes) to yield 557.5 mg of 3-(2chloropyrimidin-4-yl)-l-(2-fluoroethyl)-lH-indole (U) as a light yellow powder. ]H NMR (300 MHz, DMSO) δ 4.60 (t, 1H, J=4.7Hz), 4.69 (t, 1H, J=4.8Hz), 4.75 (t, 1H, J=4.4Hz), 4.90 (t, 1H, J = 4.4 Hz), 7.31 (m, 2H), 7.67 (m, 1H), 7.88 (d, 1H, J=5.5Hz), 8.44 (m, 1H), 8.57 (m, 2H) ppm. 13CNMR(75 MHz, DMSO) δ 47.2 (d, JCF=19.8Hz), 82.8 (d, JCF =167.7Hz), 111.6, 111.9, 115.0, 122.1, 122.3, 123.4, 125.8, 134.6, 137.8,159.4, 160.8,164.9 ppm. C^nClFNj m/zMH+276.
N-(4-Fhioro-2-methoxy-5-nitrophenyI)-4-(l-(2-fhioroethyl)-lH-mdol-3-yl)pyrimidin-2amine (Scheme 9, Intermediate V). p-Toluene sulfonic acid monohydrate (442.8 mg, 2.3 mmol) was added to a stirred suspension of 3-(2-chloropyrimidin-4-yl)-l-(2-fluoroethyl)-lH-indole (U) (535.3 mg, 1.9 mmol) and 4-fluoro-2-methoxy-5-nitroaniline (361.4 mg, 1.9 mmol) in 1,4dioxane (20 mL) and heated to reflux under nitrogen blanket. While approaching reflux température the suspended solid dissolved. Reflux was continued ovemight, then the reaction was cooled and poured into a rapidly stirred 5% (w/v) solution sodium hydrogen carbonate in DI water (200 mL) to precipitate product. Product was isolated by filtration, washed with water and allowed to dry to yield 921.4 mg of N-(4-fluoro-2-methoxy-5-nitrophenyl)-4-(l-(2-fluoroethyl)lH-indol-3-yl)pyrimidin-2-amine (V) as a fine yellow powder. CjiHpF^Oj m/z MH+=426.
N-(4-(2-(Dimethylamino)ethoxy)-2-methoxy-5-nitrophenyl)-4-(l-(2-fluoroethyl)-lHindol-3-yl)pyrimidin-2-amine (Scheme 9, Intermediate W). 2-(Dimethylamino)ethanol (0.8 ml., 7.7 mmol) was slowly added to a stirred, N2 blanketed, ambient température, suspension of sodium hydride, 60 wt.% in oil (306.4 mg, 7.7 mmol) in anhydrous 1,4-dioxane (24 mL). Anion formation was allowed to proceed for 0.5 h, then N-(4-fluoro-2-methoxy-5-nitrophenyl)-4-(l-(2fluoroethyl)-lH-indol-3-yl)pyrimidin-2-amine (intermediate V) (652.0 mg, 1.53 mmol) was added ail at once. The reaction immediately tumed to a red color, and was allowed to stir. After 10 min., LC-MS reported the reaction to be complété. DI water (5 mL) was added to quench, then the mixture was partitioned between ethyl acetate and saturated aqueous sodium chloride. The organic extract was dried (CaSO4) and evaporated to afford a yellow solid. This solid was recrystallized from boiling ethyl acetate / heptane, which upon cooling, precipitated a bright yellow crystalline powder. Thé powder was isolated by filtration, washed with heptane, and allowed to dry providing 572.0 mg of N-(4-(2-(dimethylamino)ethoxy)-2-methoxy-5nitrophenyl)-4-(l-(2-fluoroethyl)-lH-indol-3-yl)pyrimidin-2-amine (W). [H NMR (300 MHz, DMSO) δ 2.27 (s, 6H), 2.71 (t, 2H, J = 5.7Hz), 4.01 (s, 3H), 4.33 (t, 2H, J-5.6Hz ), 4.56 (t, 1H, J=4.6Hz), 4.65 (t, 1H, J=4.6Hz), 4.73 (t, 1H, J=4.2Hz), 4.89 (t, 1H, J=4.6Hz), 7.01 (s, 1H), 7.10 (m, 1H), 7.25 (m, 2H), 7.61 (d, 1H, J=8.4Hz), 8.22 (s, 1H), 8.36 (m, 3H), 8.76 (s, 1H) ppm. [3C NMR (75 MHz, DMSO) δ 46.2, 47.0 (d, JCF=19.5Hz), 57.3, 58.0, 69.0, 82.8 (d, JCF =166.6Hz),
99.2, 108.2, 111.1, 113.4, 119.2, 121.4, 122.4, 122.6, 122.8, 126.0, 131.3, 132.8, 137.6, 150.6,
156.2, 157.7, 160.5,162.5 ppm. C25H27 F N6O4m/z MH+= 495.
4-(2-(Dimethylamino)ethoxy)-Nl-(4-(l-(2-fluoroethyl)-lH-indol-3-yl)pyrimidin-2yl)-6-methoxybenzene-l,3-diamine (Scheme 9, Intermediate X). Stannous chloride dihydrate (708.3 mg, 3.1 mmol) was added to a stirred, ambient température yellow suspension of N-(4-(2(dimethylamino)ethoxy)-2-methoxy-5-nitrophenyl)-4-(l-(2-fluoroethyl)-lH-indol-3- .
yl)pyrimidin-2-amine (W) (303.8 mg, 0.6 mmol) in ethyl acetate (30 mL) and heated at reflux under nitrogen blanket for 4h. The reaction was allowed to cool, then poured into a 5%(w/v) solution of sodium hydrogen carbonate in DI water (200 mL) and stirred for 0.5 h. The multiphase mixture was then filtered through tightly packed celite, with ethyl acetate rinsing of the filter cake. The filtrate was transferred to a separatory funnel and the liquid phases separated. The retained ethyl acetate solution of product was washed with saturated aqueous sodium chloride, dried (CaSO4), and evaporated to provide a red oil which was purifîed by flash chromatography (silica gel, 2% NH4OH(aq.) in methanol / ethyl acetate; 0 to 10%,) to isolate X as 165.4 mg of red oil. C25H29FN6O2 m/z MH+ = 465.
N-(2-(2-(DimethyIamino)ethoxy)-5-((4-(l-(2-fluoroethyl)-lH-indol-3-yl)pyrimidin-2yl)amiiio)-4-methoxyphenyl)acrylamide (11, Scheme 9). 3-Chloropropanoyl chloride (38 mL, 0.4 mmol) was slowly added, by syringe, to a rapidly stirred, 0°C, nitrogen blanketed solution of 4-(2-(dimethylamino)ethoxy)-N 1-(4-(1 -(2-fluoroethyl)-1 H-indol-3 -yl)pyrimidin-2-yl) -6methoxybenzene-l,3-diamine (Intermediate X) in anhydrous tetrahydrofuran (20 mL). Upon this addition, precipitate immediately formed. The suspension was stirred at 0 °C for an additional 5 min. then the ice bath was removed. Upon confirmation of complété conversion to the 3chloropropanamide intermediate, a solution of sodium hydroxîde (726.0 mg, 18.2 mmol) in DI water (5.0 mL) was added to the réaction suspension which was heated to reflux for Ih then cooled and partitioned with brine and additional tetrahydrofuran. The organic extract was dried (CaSO4) and evaporated to yield 445.Img of solid orange foam which was purifïed by gradient flash chromatography (silica gel, 2% NH4OH(aq.) in methanol / ethyl acetate; 0 to 10%), and 5 crystalized from ethyl acetate / heptane to isolate 130 mg of Example 11 as a fine light yellow powder. Ή NMR (300 MHz, DMSO) Ô 2.28 (s, 6H), 2.58 (t, 2H, J=5.3Hz), 3.86 (s, 3H), 4.19 (t, 2H, J=5.3Hz), 4.58 (t, 1H, J=4.6Hz), 4.67 (t, 1H, J=4.5Hz), 4.72 (t, 1H, J=4.6Hz), 4.88 (t, 1H, J=4.6Hz), 5.75 (dd, 1H, J=10.4, 1.7Hz), 6.22 (dd, 1H, 1=17.0, 1.9Hz), 6.48 (m, 1H), 6.95 (s, 1H), 7.14 (t, 1H, J=7.4Hz), 7.22 (m, 2H), 7.60 (d, 1H, J=8.2Hz), 7.94 (s, 1H), 8.30 (m, 2H), 8.56 (s, 10 1H), 8.80 (s, 1H), 9.83 (s, 1H) ppm. 13C NMR (75 MHz, DMSO) δ 45.6, 46.9 (d, JCF =19.9Hz),
56.6, 57.9, 60.2, 69.4, 82.9 (d, JCF =168.2Hz), 101.6, 107.5, 111.1, 113.6, 116.9, 121.4, 122.3,
122.6, 123.2, 126.0, 126.6, 132.6, 133.2, 137.6, 145.3, 147.8, 158.0, 160.7, 162.1, 163.2 ppm. C28H31FN6O3 m/z MH+ = 519.
The following non-limiting Examples further illustrate certain aspects of the présent 15 invention, which are prepared according to the general Synthetic Schemes 1 to 9 above:
'1
Biological Assays
Compounds of the formula I as novel EGFR tyrosine kinase inhibitors were evaluated for their activity against EGFR according to the procedures described below.
Cell Culture. A431 (passage 3) and NCI-H1975 (passage 5) cells (ATCC) were started from frozen stocks and cultured in RPMI 1640 medium supplemented with 10% heat-inactivated fêtai bovine sérum, IX penicillin/streptomycin/glutamine, 1 mM sodium pyruvate, 10 mM 4-(2hydroxyethyl)-l-piperazineethanesulfonic acid (HEPES), and 0.25% D-glucose (growth medium) in T175 flasks in a humidified 30°C, 5% CO2 incubator. The cell monolayer was dispersed by 5 minute exposure to 0.25% Trypsin/EDTA solution (Life Technologies) and the solution was neutralized with a fresh growth medium. Pooled cells were pelleted by centrifugation (200xg, 8 min.), resuspended in the growth medium, and an aliquot was removed for cell counting using an automated cell counter (Logos Biosystems). The cells maintained normal moiphology and growth characteristics during the period of the study.
Cell Prolifération Assay. Dispersed cells were pooled by centrifugation (200xg, 8 min.) and resuspended in a fresh medium to a concentration of 1.00F.+04 cells/ml. 200 pL of the cell suspension was added to each well (2,000 cells/well) of a black-walled 96 well plate and the cells were allowed to attach overnight under normal culture conditions. After ovemight culturing, 1 pL of a test compound (n = 3 per concentration) was added per well to achieve final concentrations of 10, 3.33, 1.11, 0.370, 0.124, 0.0412, 0.0137, 0.0046, and 0.0015 pM. The final DMSO concentration in the well was 0.5% v/v. Vehicle, non-treated, and cell-free wells were also included in the assay. The cells were cultured under normal conditions for 72 hours with daily visual inspection,
Cell prolifération was measured using the dye Alamar Blue (resazurin). Resazurin is reduced by cellular enzymes to resorufin, which is fluorescent (544 nM excitation, 612 nm émission). Fluorescence intensity was proportional to cell number. A resazurin stock solution was prepared in a phosphate-buffered saline (PB S) to a stock concentration of 440 μΜ. The resazurin stock solution (40 pL each) was added to each well at hour 67 of the 72 hour incubation period. The plate was retumed to normal culture conditions and fluorescence measurements were collected using a Cytation 3 multimode plate reader (Biotek) at 72 hours.
Data Analysis. Fluorescence measurements were normalized against cell-free (background) readings and the total growth over 72 hour time period was determined versus the average of the vehicle control wells. Average and standard déviation values were determined for each condition (n=3).
Table 1 contains illustrative data from study of représentative compounds of the présent invention, which demonstrate their excellent selectivity for inhibition of the growth of H1975 (double mutant) cells over A431 (wild type) cells.
Table 1. Biological activity of selected compounds in the A431 (wild type) and H1975 (double mutant) cell prolifération assays.
Example A431IC (pM)' H1975IC50 (pM)a
1 + +++
2 + -H-+
3 + ++
4 + -H-+
5 ++ ++
8 -H- +++
9 + -H-+
10 ++
11 -H- | | |
“An IC50 value greater than 1.0 μΜ is represented by an IC50 in the range of 0.1 -1.0 μΜ is represented by and an IC50 value below 0.1 μΜ is represented by “+++”.
The in vivo anticancer activity of Examples 1 and 2 is also illustrated in FIGs. 1-4.
Antitumor Activity of Example 1 in the H1975 Mouse Xenograft Model. The in vivo anticancer activity of Example 1 against tumors with the L858R/T79OM double mutation is illustrated in FIG. 1. Example 1 was evaluated in subcutaneously-implanted H1975 human nonsmall cell lung carcinoma xenographs in female nude mice at 6.25, 12.5 and 25 mg/kg. Example 1 was dosed orally once a day for 14 days (days 6-19). At ail doses, Example 1 was well tolerated, resulting in no treatment-related mortality. Treatment with 1 at 6.25, 12.5 and 25 mg/kg produced a médian time to évaluation size of 28.9, 31.6 and 34,3 days, respectively, resulting in a statistically significant (P<0.05) tumor growth delay of 14, 16.7 and 19.3 days, respectively. At 25 mg/kg, treatment produced a 100% incidence of complété régressions and 10% of the mice were tumor free survivors,
Antitumor Activity of Example 2 in the H1975 Mouse Xenograft Model. The in vivo anticancer activity of Example 2 against tumors with the L858R/T790M double mutation is illustrated in FIG. 2. Example 2 was evaluated in subcutaneously-implanted H1975 human nonsmall cell lung carcinoma xenographs in female nude mice at 50 and 100 mg/kg. Example 2 was dosed orally once a day for 14 days (days 7-20). At 100 mg/kg oral dosing, Example 2 was well tolerated and produced significant (P<0.05) anticancer activity based upon the % tumor growth inhibition values (%TGI) of 110.5%, 116.6% and 116.6%, which were calculated from the médian tumor burdens on days 10, 14 and 17, respectively. Time to évaluation size (750 mm3) was 39.6 days, resulting in a tumor growth delay (T-C) of 22.2 days, which is also statistically significant. Treatment produced a 100% incidence of complété tumor régression and 12.5% of the mice remained tumor free (TF S) at the completion of the study.
Antitumor Activity of Example 1 in the HCC827 Mouse Xenograft Model. The in vivo anticancer activity of Example 1 against tumors with the deIE746-A750 activating mutation is illustrated in FIG. 3. Example 1 was evaluated in a subcutaneously-implanted HCC827 human non-small cell lung carcinoma xenographs in female nude mice at 6.25 mg/kg. Example 1 was dosed orally once a day for 14 days (days 13-26). At 6.25 mg/kg oral dosing, 1 was well tolerated, resulting in no treatment-related mortality. Treatment with 1 produced a médian time to évaluation size of 61.5 days, resulting in a statistically significant (P<0.05) tumor growth delay of 33.2 days. Treatment produced a 100% incidence of complété tumor régression at the completion of dosing. FIG. 4 shows the average concentration of Example 1 in plasma, brain and tumor tissues following a 25 mg/kg oral dose in this model.
The foregoing examples and description of the preferred embodiments should be taken as illustrating, rather than as limiting the présent invention as defined by the daims. As will be readily appreciated, numerous variations and combinations of the features set forth above 5 can be utilized without departing from the présent invention as set forth in the daims.

Claims (5)

CLAIMS (Amended) What is claimed is:
1. A compound of formula I :
R3
or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein:
G is selected from substituted or unsubstituted 17T-indol-3-yl, substituted or unsubstituted 777-indazol-3-yl, substituted or unsubstituted 277-indazol-3-yl, and substituted or unsubstituted pyrazolo[l,5-a]-pyridin-3-yl, and substituted or unsubstituted 4,5,6,7-tetrahydropyrazolo[l,5a]pyridin-3-yl;
X is selected from oxygen, sulfur, and methylene;
R1 is selected from hydrogen, halogen, methyl, trifluoromethyl, andcyano;
R2, R3, and R4 are the same or different and are independently selected from hydrogen, halogen, and trifluoromethyl;
R5 is selected from lower alkyl, optionally substituted 3- to 6-membered heterocyclyl, R7R8N-(lower alkyl), and R7RsN-(cycloalkylaIkyl), wherein R7 and R8 are the same or different and are independently selected from hydrogen and lower alkyl; and
R6 is selected from lower alkoxy and lower alkyl.
2. The compound of claim 1, or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein:
G is selected from the group consisting of 7/f-indol-3-yl, 1 -methyl-7/7-indol-3-yl, l-(2fluoroethyl)-777-indol-3-yl, 1,2-dimethyl-7/7-indol-3-yl, pyrazolo[l,5-a]-pyridin-3-yl, 4,5,6,7tetrahydropyrazoIo[l,5-a]pyridin-3-yl, l-mcthyl-777-indazol-3-yl, and 2-methyl-277-indazol-3-yl.
3. The compound of claim 1 or 2, or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein:
R5 is selected from Cj-Ce alkyl, substituted or unsubstituted azetidinyl, substituted or unsubstituted pyrrolidinyl, substituted or unsubstituted piperidinyl, R7R8N-(CH2)n- (η = 1 to 5), R7R8N-(C3-Ce cycloalkyl)-(CH2)m- (m = 1 to 3), wherein R7 and R8 are the same or different and are independently selected from hydrogen and lower alkyl.
4. The compound of claim 3, or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein:
R5 is selected from methyl, l-(dimethylamino)-cyclopropylmethyl, 3(dimethylamino)cyclobutyl, l-methylazetidin-3-yl, (7?)-l-methylpynOlidin-3-yl, (5)-1methylpyrrolidin-3-yl, and 1-methylpiperidin-4-yl, and 2-dimethylamino-ethyl.
5. The compound of any one of daims 1 to 4, or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein R1 is hydrogen, halogen, or methyl.
6. The compound of any one of daims 1 to 5, or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein R2 is hydrogen or halogen.
Ί. The compound of any one of daims 1 to 6, or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein R4 is hydrogen.
. 8. The compound of any one of daims 1 to 5, or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein:
R2 is hydrogen, F, or Cl;
R3 is hydrogen, F, Cl, or -CF3; and
R4 is hydrogen.
9. The compound of daim 1, characterized by a structure of formula II:
R9
Πγ- f '\JAQ A„ γ hn 0
o. NNy H I OCH3 i
II or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, wherein:
X is O, S, or CH2;
5 Q is C-R10 or N
R9 is CH3 or CH2CH2F; and
R10 is H or CH3.
10. The compound of claim 1, selected from the group consisting of:
or a pharmaceutically acceptable sait, solvaté, or prodrug thereof,
11. The compound of claim 1, 9, or 10, or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, selected from the group consisting of:
12. A composition comprising a compound according to any one of claîms 1 to 11, or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, and a pharmaceutically
10 acceptable carrier, adjuvant, diluent, or vehicle.
13. Use of a compound according to any one of daims 1 to 11, or a pharmaceutically acceptable sait, solvaté, or prodrug thereof, or a composition of claim 12, in the manufacture of a médicament for treatment of a disease or disorder associated with an EGFR activity.
14. The Use of claim 13, wherein said disease or disorder is a cancer selected from 15 the group consisting of brain cancer, lung cancer, kidney cancer, bone cancer, liver cancer, bladder cancer, head and neck cancer, esophageal cancer, stomach cancer, colon cancer, rectum cancer, breast cancer, ovarian cancer, melanoma, skin cancer, adrenal cancer, cervical cancer, lymphoma, and thyroid tumors and their complications,
15. The use of claim 13 or 14, wherein said disease or disordèr is brain cancer or
5 lung cancer.
OA1201700215 2014-12-11 2015-12-11 Substituted 2-anilinopyrimidine derivatives as EGFR modulators. OA18657A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US62/090869 2014-12-11
US62/166883 2015-05-27

Publications (1)

Publication Number Publication Date
OA18657A true OA18657A (en) 2019-04-08

Family

ID=

Similar Documents

Publication Publication Date Title
US11414401B2 (en) Substituted 2-anilinopyrimidine derivatives as EGFR modulators
AU2015292425B2 (en) 2-H-indazole derivatives as cyclin-dependent kinase (CDK) inhibitors and therapeutic uses thereof
EP3746072B1 (en) 2h-indazole derivatives as cdk4 and cdk6 inhibitors and therapeutic uses thereof
AU2014289762B2 (en) Protein tyrosine kinase modulators and methods of use
US10851092B2 (en) Pyridine compound
JP6101341B2 (en) Substituted pyridopyrimidine compounds and their use as FLT3 inhibitors
JP2007534624A (en) Biaryl piperazinyl-pyridine analogues
WO2021231400A1 (en) Bis-aryl ethers containing n-acyl azetidine as egfr/her2 inhibitors
EP3471730B1 (en) Pharmaceutical salts of n-(2-(2-(dimethylamino)ethoxy)-4-methoxy-5-((4-(1-methyl-1h-indol-3-yl)pyrimidin-2-yl)amino)phenyl)acrylamide and crystalline forms thereof
OA18657A (en) Substituted 2-anilinopyrimidine derivatives as EGFR modulators.
CN115803325A (en) EGFR inhibitor and preparation method and application thereof
OA19873A (en) 2H-indazole derivatives as CDK4 and CDK6 inhibitors and therapeutic uses thereof.
KR20160030296A (en) Protein tyrosine kinase modulators and methods of use