Nothing Special   »   [go: up one dir, main page]

WO2019071351A1 - Nouveaux dérivés de 4-aryloxyquinazoline fluorés utilisés en tant qu'inhibiteurs d'egfr utiles pour le traitement de cancers - Google Patents

Nouveaux dérivés de 4-aryloxyquinazoline fluorés utilisés en tant qu'inhibiteurs d'egfr utiles pour le traitement de cancers Download PDF

Info

Publication number
WO2019071351A1
WO2019071351A1 PCT/CA2018/051282 CA2018051282W WO2019071351A1 WO 2019071351 A1 WO2019071351 A1 WO 2019071351A1 CA 2018051282 W CA2018051282 W CA 2018051282W WO 2019071351 A1 WO2019071351 A1 WO 2019071351A1
Authority
WO
WIPO (PCT)
Prior art keywords
compounds
compound
application
egfr
disorder
Prior art date
Application number
PCT/CA2018/051282
Other languages
English (en)
Inventor
Abdelmalik Slassi
Peter Dove
David Alexander ROSA
Original Assignee
Trillium Therapeutics Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Trillium Therapeutics Inc. filed Critical Trillium Therapeutics Inc.
Publication of WO2019071351A1 publication Critical patent/WO2019071351A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/88Oxygen atoms

Definitions

  • the present application relates to novel fluorinated derivatives, to processes for their preparation, to compositions comprising them, and to their use in therapy. More particularly, it relates to compounds useful in the treatment of diseases, disorders or conditions mediated by epidermal growth factor receptor. Such compounds and salts thereof may be useful in the treatment or prevention of a number of different cancers.
  • the application also relates to pharmaceutical compositions comprising said compounds and salts thereof, especially useful polymorphic forms of these compounds and salts, intermediates useful in the manufacture of said compounds and to methods of treatment of diseases mediated by various different forms of EGFR using said compounds and salts thereof.
  • Epidermal Growth Factor Receptor is a transmembrane protein tyrosine kinase of the ErbB receptor family.
  • a growth factor ligand such as epidermal growth factor (EGF)
  • the receptor can homo- dimerise with another EGFR molecule or hetero-dimerise with another family member such as ErbB2 (HER2), ErbB3 (HER3), or ErbB4 (HER4).
  • HER2 ErbB2
  • HER3 ErbB3
  • HER4 ErbB4
  • ErbB family signalling promotes proliferation, invasion, metastasis, angiogenesis, and tumour cell survival and has been described in many human cancers, including those of the lung, head, neck and breast.
  • the ErbB family therefore represents a rational target for anticancer drug development and a number of agents targeting EGFR or ErbB2 are now clinically available, including gefitinib (IRESSATM), erlotinib (TARCEVATM) and lapatinib (TYKERBTM, TYVERBTM).
  • IRESSATM gefitinib
  • TARCEVATM erlotinib
  • TYKERBTM lapatinib
  • Detailed reviews of ErbB receptor signalling and its involvement in tumourigenesis are provided in New England Journal of Medicine (2008) Vol. 358, 1 160-74 and Biochemical and Biophysical Research Communications (2004) Vol. 319, l-ll.
  • This mutation is not believed to hinder the binding of gefitinib or erlotinib to EGFR sterically, it merely alters the affinity to ATP to levels comparable to WT EGFR.
  • agents which inhibit EGFR harbouring the gatekeeper mutation may be especially useful in the treatment of cancer.
  • compounds that exhibit favourable potency against WT EGFR versus activating mutant forms of EGFR for example the L858R EGFR mutant, or the del E746-A750 mutant or the Exon19 deletion EGFR mutant
  • resistant mutant forms of EGFR for example T790M EGFR mutant
  • the human ether-a-go-go related gene (hERG) channels encode the pore- forming subunit of the inward rectifying voltage gated potassium channel expressed in the heart (kr) as well as in brain regions and endocrine cells.
  • This subunit is involved in cardiac action potential repolarization, and reduced function of hERG lengthens ventricular action potentials amd prolongs the QT interval in an electrocardiogram. Blockade of this target increases the risk for potentially fatal ventricular arrhythmia therefore it is desirable for compounds that are to act as drugs to have minimal potency at the hERG channel [Physiological Reviews 2012 92(3) 1393 ⁇
  • compounds that can effectively penetrate the blood brain barrier Such compounds may be expected to be more suitable as therapeutic agents, particularly for the treatment of cancers of the brain.
  • GBM Glioblastoma multiforme
  • PI3K phosphoinositide-3'-kinase
  • Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) tumor suppressor is frequently phosphorylated at a conserved tyrosine residue, Y240, in GBM clinical samples.
  • Phosphorylation of Y240 is associated with shortened overall survival and resistance to EGFR inhibitor therapy in GBM patients and plays an active role in mediating resistance to EGFR inhibition in vitro.
  • Y240 phosphorylation can be mediated by both fibroblast growth factor receptors and SRC family kinases (SFKs) but does not affect the ability of PTEN to antagonize PI3K signaling.
  • a novel class of fluorinated derivatives of Formula I has been prepared and found to be useful in the treatment of cancers and other EGFR related disorders. Surprisingly, it was observed that replacement of a nitrogen atom in a previously known class of non-covalent EGFR inhibitors with an oxygen atom retained biochemical activity and showed reduced hERG channel potency.
  • the compound(s) of the application also exhibit favourable toxicity profiles (for example a decreased hERG blocking liability) in comparison with other known EGFR / EGFR-mutant inhibitors. Therefore, in some embodiments, the compounds of the application are especially useful in the treatment of disease states in which EGFR and/or activating mutations of EGFR and/or resistance mutations of EGFR are implicated, for example in the treatment of cancer.
  • the present application includes a compound of Formula I or a pharmaceutically acceptable salt and/or solvate thereof:
  • R 1 is selected from unsubstituted or substituted phenyl and unsubstituted or substituted naphthyl, wherein the substituents for R 1 are selected from one or more of CI, Br, F, Ci- 4 alkyl, C2-4alkenyl, C2-4alkynyl, Ci- 4 fluoroalkyl, C2- 4 fluoroalkenyl and C2-4fluoroalkynyl;
  • R 2 is selected from (CH 2 )nOR 4 and C(0)X(CH 2 )nOR 4 ;
  • R 3 is selected from Ci- 4 alkyl, Ci- 4 fluoroalkyl and (CH2)mOR 5 ;
  • R 4 is selected from CHF2 and CF3;
  • R 5 is selected from CHF2, CF3 and CH3;
  • X is selected from NH, NCH3 and O;
  • n is selected from 1 , 2, 3 and 4;
  • n is selected from 1 , 2, 3, and 4.
  • the present application also includes a composition comprising one or more compounds of the application and a carrier.
  • the composition is a pharmaceutical composition comprising one or more compounds of the application and a pharmaceutically acceptable carrier.
  • the compounds of the application have been shown to be capable of inhibiting EGFR protein function. Therefore the compounds of the application are useful for treating diseases, disorders or conditions treatable by inhibition of EGFR. Accordingly, the present application also includes a method of treating a disease, disorder or condition treatable by inhibition of EGFR, comprising administering a therapeutically effective amount of one or more compounds of the application to a subject in need thereof.
  • the compounds of the application are used as medicaments. Accordingly, the application also includes a compound of the application for use as a medicament.
  • the present application also includes a use of one or more compounds of the application for treatment of a disease, disorder or condition treatable by inhibition of EGFR as well as a use of one or more compounds of the application for the preparation of a medicament for treatment of a disease, disorder or condition treatable by inhibition of EGFR.
  • the application further includes one or more compounds of the application for use in treating a disease, disorder or condition treatable by inhibition of EGFR.
  • the disease, disorder or condition treatable by inhibition of EGFR is a neoplastic disorder.
  • the treatment is in an amount effective to ameliorate at least one symptom of the neoplastic disorder, for example, reduced cell proliferation or reduced tumor mass in a subject in need of such treatment.
  • the disease, disorder or condition is cancer.
  • the disease, disorder or condition is a disease, disorder or condition associated with an uncontrolled and/or abnormal cellular activity affected directly or indirectly by EGFR.
  • the uncontrolled and/or abnormal cellular activity that is affected directly or indirectly by EGFR is proliferative activity in a cell.
  • the application also includes a method of inhibiting proliferative activity in a cell, comprising administering an effective amount of one or more compounds of the application to the cell.
  • the disease, disorder or condition treatable by inhibition of EGFR is cancer and the one or more compounds of the application are administered in combination with one or more additional cancer treatments.
  • the additional cancer treatment is selected from radiotherapy, chemotherapy, targeted therapies such as antibody therapies and small molecule therapies such as tyrosine-kinase inhibitors, immunotherapy, hormonal therapy and anti-angiogenic therapies.
  • the application additionally provides a process for the preparation of compounds of the applicaition. General and specific processes are discussed in more detail below and set forth in the Examples.
  • composition of the application refers to a composition, such as a pharmaceutical composition, comprising one or more compounds of Formula I , or pharmaceutically acceptable salts, solvates, prodrugs and/or radiolabeled versions thereof .
  • the second component as used herein is chemically different from the other components or first component.
  • a “third” component is different from the other, first, and second components, and further enumerated or “additional” components are similarly different.
  • the second component as used herein is chemically different from the other components or first component.
  • a “third” component is different from the other, first, and second components, and further enumerated or “additional” components are similarly different.
  • suitable means that the selection of the particular compound or conditions would depend on the specific synthetic manipulation to be performed, the identity of the molecule(s) to be transformed and/or the specific use for the compound, but the selection would be well within the skill of a person trained in the art.
  • the compounds described herein may have at least one asymmetric center. Where compounds possess more than one asymmetric center, they may exist as diastereomers. It is to be understood that all such isomers and mixtures thereof in any proportion are encompassed within the scope of the present application. It is to be further understood that while the stereochemistry of the compounds may be as shown in any given compound listed herein, such compounds may also contain certain amounts (for example, less than 20%, suitably less than 10%, more suitably less than 5%) of compounds of the present application having alternate stereochemistry. It is intended that any optical isomers, as separated, pure or partially purified optical isomers or racemic mixtures thereof are included within the scope of the present application.
  • the compounds of the present application may also exist in different tautomeric forms and it is intended that any tautomeric forms which the compounds form, as well as mixtures thereof, are included within the scope of the present application.
  • the compounds of the present application may further exist in varying polymorphic forms and it is contemplated that any polymorphs, or mixtures thereof, which form are included within the scope of the present application.
  • alkyl as used herein, whether it is used alone or as part of another group, means straight or branched chain, saturated alkyl groups. The number of carbon atoms that are possible in the referenced alkyl group are indicated by the prefix "Cni-n2".
  • Ci-6alkyl means an alkyl group having 1 , 2, 3, 4, 5 or 6 carbon atoms.
  • alkylene whether it is used alone or as apart of another group, means straight or branched chain, saturated alkylene group, that is, a saturated carbon chain that contains substituents on two of its ends.
  • the number of carbon atoms that are possible in the referenced alkylene group are indicated by the prefix "Cn-iV.
  • Ci-6alkylene means an alkylene group having 1 , 2, 3, 4, 5 or 6 carbon atoms.
  • alkenyl as used herein, whether it is used alone or as part of another group, means straight or branched chain, unsaturated alkyl groups containing at least one double bond.
  • the number of carbon atoms that are possible in the referenced alkylene groups are indicated by the prefix "Cn-i- n2".
  • C2-6alkenyl means an alkenyl group having 2, 3, 4, 5 or 6 carbon atoms and at least one double bond.
  • alkynyl as used herein, whether it is used alone or as part of another group, means straight or branched chain unsaturated alkyl groups containing at least one triple bond.
  • the number of carbon atoms that are possible in the referenced alkylyne group are indicated by the prefix "Cn-i- n2".
  • C2-6alkynyl means an alkynyl group having 2, 3, 4, 5 or 6 carbon atoms and at least one triple bond.
  • fluoroalkyl refers to an alkyl group wherein one or more, including all of the hydrogen atoms are replaced by a fluorine atom.
  • substituted refers to a structure, molecule or group in which one or more available hydrogen atoms are replaced with one or more other chemical groups.
  • available refers to atoms that would be known to a person skilled in the art to be capable of replacement by a substituent.
  • protecting group refers to a chemical moiety which protects or masks a reactive portion of a molecule to prevent side reactions in those reactive portions of the molecule, while manipulating or reacting a different portion of the molecule. After the manipulation or reaction is complete, the protecting group is removed under conditions that do not degrade or decompose the remaining portions of the molecule.
  • PG protecting group
  • the selection of a suitable protecting group can be made by a person skilled in the art. Many conventional protecting groups are known in the art, for example as described in "Protective Groups in Organic Chemistry” McOmie, J.F.W. Ed., Plenum Press, 1973, in Greene, T.W.
  • cell refers to a single cell or a plurality of cells and includes a cell either in a cell culture or in a subject.
  • subject includes all members of the animal kingdom including mammals, and suitably refers to humans. Thus the methods of the present application are applicable to both human therapy and veterinary applications.
  • the subject is a mammal. In another embodiment, the subject is human.
  • pharmaceutically acceptable means compatible with the treatment of subjects, for example humans.
  • pharmaceutically acceptable carrier means a nontoxic solvent, dispersant, excipient, adjuvant or other material which is mixed with the active ingredient in order to permit the formation of a pharmaceutical composition, i.e., a dosage form capable of administration to a subject.
  • a pharmaceutical composition i.e., a dosage form capable of administration to a subject.
  • a pharmaceutically acceptable oil typically used for parenteral administration.
  • pharmaceutically acceptable salt means either an acid addition salt or a base addition salt which is suitable for, or compatible with the treatment of subjects.
  • An acid addition salt suitable for, or compatible with, the treatment of subjects is any non-toxic organic or inorganic acid addition salt of any basic compound.
  • Basic compounds that form an acid addition salt include, for example, compounds comprising an amine group.
  • Illustrative inorganic acids which form suitable salts include hydrochloric, hydrobromic, sulfuric, nitric and phosphoric acids, as well as acidic metal salts such as sodium monohydrogen orthophosphate and potassium hydrogen sulfate.
  • Illustrative organic acids which form suitable salts include mono-, di- and tricarboxylic acids.
  • organic acids are, for example, acetic, trifluoroacetic, propionic, glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, hydroxymaleic, benzoic, hydroxybenzoic, phenylacetic, cinnamic, mandelic, salicylic, 2-phenoxybenzoic, p-toluenesulfonic acid and other sulfonic acids such as methanesulfonic acid, ethanesulfonic acid and 2- hydroxyethanesulfonic acid.
  • Either the mono- or di-acid salts can be formed, and such salts can exist in either a hydrated, solvated or substantially anhydrous form.
  • acid addition salts are more soluble in water and various hydrophilic organic solvents, and generally demonstrate higher melting points in comparison to their free base forms.
  • the selection criteria for the appropriate salt will be known to one skilled in the art.
  • Other non-pharmaceutically acceptable salts such as but not limited to oxalates may be used, for example in the isolation of compounds of the application for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt.
  • a base addition salt suitable for, or compatible with, the treatment of subjects is any non-toxic organic or inorganic base addition salt of any acidic compound.
  • Acidic compounds that form a basic addition salt include, for example, compounds comprising a carboxylic acid group.
  • Illustrative inorganic bases which form suitable salts include lithium, sodium, potassium, calcium, magnesium or barium hydroxide as well as ammonia.
  • Illustrative organic bases which form suitable salts include aliphatic, alicyclic or aromatic organic amines such as isopropylamine, methylamine, trimethylamine, picoline, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-dimethylaminoethanol, 2- diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N- ethylpiperidine, polyamine resins, and the like.
  • organic amines such as isopropylamine, methylamine, trimethylamine, picoline, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-dimethylaminoethanol, 2- diethylaminoethanol, di
  • Exemplary organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine.
  • exemplary organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine.
  • the selection of the appropriate salt may be useful so that an ester functionality, if any, elsewhere in a compound is not hydrolyzed.
  • the selection criteria for the appropriate salt will be known to one skilled in the art.
  • solvate means a compound, or a salt or prodrug of a compound, wherein molecules of a suitable solvent are incorporated in the crystal lattice.
  • a suitable solvent is physiologically tolerable at the dosage administered. Examples of suitable solvents are ethanol, water and the like. When water is the solvent, the molecule is referred to as a "hydrate”.
  • solvates of the compounds of the application will vary depending on the compound and the solvate. In general, solvates are formed by dissolving the compound in the appropriate solvent and isolating the solvate by cooling or using an antisolvent. The solvate is typically dried or azeotroped under ambient conditions. The selection of suitable conditions to form a particular solvate can be made by a person skilled in the art.
  • treating means an approach for obtaining beneficial or desired results, including clinical results.
  • beneficial or desired clinical results can include, but are not limited to alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disease, stabilized (i.e. not worsening) state of disease, preventing spread of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, diminishment of the reoccurrence of disease, and remission (whether partial or total), whether detectable or undetectable.
  • Treating and “treatment” can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • Treating” and “treatment” as used herein also include prophylactic treatment.
  • a subject with early cancer can be treated to prevent progression, or alternatively a subject in remission can be treated with a compound or composition described herein to prevent recurrence.
  • Treatment methods comprise administering to a subject a therapeutically effective amount of one or more of the compounds of the application and optionally consist of a single administration, or alternatively comprise a series of administrations.
  • the compounds of the application are administered at least once a week.
  • the compounds are administered to the subject from about one time per two weeks, three weeks or one month.
  • the compounds are administered about one time per week to about once daily.
  • the compounds are administered 2, 3, 4, 5 or 6 times daily.
  • the length of the treatment period depends on a variety of factors, such as the severity of the disease, disorder or condition, the age of the subject, the concentration and/or the activity of the compounds of the application, and/or a combination thereof. It will also be appreciated that the effective dosage of the compound used for the treatment may increase or decrease over the course of a particular treatment regime. Changes in dosage may result and become apparent by standard diagnostic assays known in the art. In some instances, chronic administration may be required. For example, the compounds are administered to the subject in an amount and for duration sufficient to treat the patient.
  • "Palliating" a disease, disorder or condition means that the extent and/or undesirable clinical manifestations of a disease, disorder or condition are lessened and/or time course of the progression is slowed or lengthened, as compared to not treating the disorder.
  • prevention or “prophylaxis”, or synonym thereto, as used herein refers to a reduction in the risk or probability of a patient becoming afflicted with a disease, disorder or condition or manifesting a symptom associated with a disease, disorder or condition.
  • the "disease, disorder or condition” as used herein refers to a disease, disorder or condition treatable by inhibition of EGFR activity and particularly using an EGFR inhibitor, such as a compound of the application herein described.
  • EGFR EGFR activity associated with the disease is blocked or inhibited by one or more of the present compounds.
  • an effective amount means an amount of one or more compounds of the application that is effective, at dosages and for periods of time necessary to achieve the desired result.
  • an effective amount is an amount that, for example, inhibits EGFR activity compared to the inhibition without administration of the one or more compounds.
  • effective amounts vary according to factors such as the disease state, age, sex and/or weight of the subject.
  • the amount of a given compound or compounds that will correspond to an effective amount will vary depending upon factors, such as the given drug(s) or compound(s), the pharmaceutical formulation, the route of administration, the type of condition, disease or disorder, the identity of the subject being treated, and the like, but can nevertheless be routinely determined by one skilled in the art.
  • the effective amount is one that following treatment therewith manifests as an improvement in or reduction of any disease symptom.
  • amounts that are effective can cause a reduction in the number, growth rate, size and/or distribution of tumours.
  • administered means administration of a therapeutically effective amount of one or more compounds or compositions of the application to a cell either in cell culture or in a subject.
  • Neoplasm refers to a mass of tissue resulting from the abnormal growth and/or division of cells in a subject having a neoplastic disorder. Neoplasms can be benign (such as uterine fibroids and melanocytic nevi), potentially malignant (such as carcinoma in situ) or malignant (i.e. cancer).
  • neoplastic disorders include but are not limited to carcinoma, sarcoma, metastatic disorders (e.g., tumors arising from the prostate), hematopoietic neoplastic disorders, (e.g., leukemias, lymphomas, myeloma and other malignant plasma cell disorders), metastatic tumors and other cancers.
  • cancer refers to cellular-proliferative disease states.
  • Compounds of the present application were prepared and were found to inhibit uncontrolled and/or abnormal cellular activities affected directly or indirectly by EGFR protein.
  • compounds of the present application exhibited activity as EGFR inhibitors, and are therefore useful in therapy, for example for the treatment of neoplastic disorders such as cancer.
  • Compounds of the application also showed reduced inhibition of hERG compared to closely related arylaminequinazoline compounds indicating that compounds of the application may show a reduced risk for cardiovascular side effects.
  • the present application includes a compound of Formula I or a pharmaceutically acceptable sal or solvate:
  • R 1 is selected from unsubstituted or substituted phenyl and unsubstituted or substituted naphthyl, wherein the substituents for R 1 are selected from one or more of CI, Br, F, Ci- 4 alkyl, C2-4alkenyl, C2-4alkynyl, Ci- 4 fluoroalkyl, C2- 4 fluoroalkenyl and C2-4fluoroalkynyl;
  • R 2 is selected from (CH 2 )nOR 4 and C(0)X(CH 2 )nOR 4 ;
  • R 3 is selected from Ci- 4 alkyl, Ci- 4 fluoroalkyl and (CH2)mOR 5 ;
  • R 4 is selected from CHF2 and CF3;
  • R 5 is selected from CHF2, CF3 and CH3;
  • X is selected from NH, NCH3 and O;
  • n is selected from 1 , 2, 3 and 4;
  • n is selected from 1 , 2, 3, and 4.
  • R 1 has one to three substituents.
  • R 1 is selected from substituted phenyl, wherein the substituents for R 1 are selected from one to three of CI, Br, F, Ci- 4 alkyl, C2-4alkenyl, C2- 4 alkynyl, Ci- 4 fluoroalkyl, C2-4fluoroalkenyl and C2-4fluoroalkynyl.
  • both of R 2 and R 3 are .
  • R 2 is F and R 3 is CH3.
  • R 4 is CHF2.
  • R 5 is CHF2.
  • X is O.
  • n is selected from 1 , 2 and 3. In some embodiments, n is 2.
  • n is selected from 1 , 2 and 3. In some embodiments, m is 2.
  • the compounds of Formula I are selected from: 6,7-bis[2-(difluoromethoxy)ethoxy]- 4-(3-ethynylphenoxy)quinazoline
  • Compounds of the present application can be prepared by various synthetic processes. The choice of particular structural features and/or substituents may influence the selection of one process over another. The selection of a particular process to prepare a given compound of Formula I is within the purview of the person of skill in the art. Some starting materials for preparing compounds of the present application are available from commercial chemical sources. Other starting materials, for example as described below, are readily prepared from available precursors using straightforward transformations that are well known in the art.
  • the compounds of Formula I are prepared according to the processes illustrated in Schemes l-V. Variables in these schemes are as defined above for Formula I unless otherwise specified.
  • commercially available 6-benzyloxy-4- chloro-7-methoxy-quinazoline (A) is a versatile intermediate that can be used to make certain compounds of Formula I as shown in Scheme I . Therefore, for example, compounds of Formula A are reacted with phenols or naphthols R 1 OH in the presence of a suitable base, such as potassium carbonate, in a suitable solvent, such as as dimethylformamide, to provide compounds of Formula B.
  • a suitable base such as potassium carbonate
  • intermediates of Formula C are used to prepare compounds of Formula I wherein R 2 is C(0)X(CH2)nOR 4 and X is NH or NCH3 by reacting the compounds of Formula C with phosgene in the presence of a suitable amine as shown in Scheme I I.
  • 6-benzyloxy-4- chloro-7-methoxy-quinazoline (D) is a versatile intermediate that is used to make certain compounds of Formula I as shown in Scheme II I. Therefore, for example, compounds of Formula D are reacted with phenols or naphthols R 1 OH in the presence of a suitable base, such as potassium carbonate, in a suitable solvent, such as as dimethylformamide, to provide compounds of Formula E. Compounds of Formula E are then deprotected, using acidic conditions, for example in the presence of TFA, to give compounds of Formula F.
  • a suitable base such as potassium carbonate
  • a suitable solvent such as as dimethylformamide
  • known intermediate J is another versatile internmediate that is used to make certain compounds of Formula I as shown in Scheme IV. Therefore, for example, compounds of Formula J are selectively alkylated at the phenolic position using a suitable base, such as potassium carbonate, in a suitable solvent, such as as dimethylformamide, to give compounds of Formula K which are then chlorinated to give compounds of Formula L. Compounds of Formula L then undergo a displacement reaction with various phenols or naphthols (R 1 -OH) in the presence of a suitable base, such as potassium carbonate, in a suitable solvent, such as as dimethylformamide, giving compounds of Fornula M.
  • a suitable base such as potassium carbonate
  • a suitable solvent such as as dimethylformamide
  • R 2 and R 3 are identical or different
  • the substituents on R 1 may be converted using known reactions into other substituents on R 1 .
  • compounds of Formulat I wherein R 1 is substituted at least with one bromine are cross-coupled to trimethylsilylacetylene using bis(triphenylphosphine)palladium(ll) dichloride to give compounds of Formula O which can be deprotected, for example, using potassium carbonate and methanol to give compounds of Formula I, wherein R 1 is substituted with an acetylene (C2alkynyl).
  • Scheme V [0079] In Schemes l-IV the benzyl group (Ph-Chte) is used as a protecting group.
  • benzyl group Ph-Chte
  • other protecting groups may be used in place of the benzyl group.
  • the other protecting groups are also removable under acidic conditions.
  • a transformation of a group or substituent into another group or substituent by chemical manipulation can be conducted on any intermediate or final product on the synthetic path toward the final product, in which the possible type of transformation is limited only by inherent incompatibility of other functionalities carried by the molecule at that stage to the conditions or reagents employed in the transformation.
  • Such inherent incompatibilities, and ways to circumvent them by carrying out appropriate transformations and synthetic steps in a suitable order will be readily understood to one skilled in the art. Examples of transformations are given herein, and it is to be understood that the described transformations are not limited only to the generic groups or substituents for which the transformations are exemplified.
  • the compounds of the present application are suitably formulated in a conventional manner into compositions using one or more carriers. Accordingly, the present application also includes a composition comprising one or more compounds of the application and a carrier. The compounds of the application are suitably formulated into pharmaceutical compositions for administration to subjects in a biologically compatible form suitable for administration in vivo. Accordingly, the present application further includes a pharmaceutical composition comprising one or more compounds of the application and a pharmaceutically acceptable carrier. In embodiments of the application the pharmaceutical compositions are used in the treatment of nay of the diseases, disorders or conditions described herein.
  • the compounds of the application are administered to a subject in a variety of forms depending on the selected route of administration, as will be understood by those skilled in the art.
  • a compound of the application is administered by oral, inhalation, parenteral, buccal, sublingual, nasal, rectal, vaginal, patch, pump, topical or transdermal administration and the pharmaceutical compositions formulated accordingly.
  • administration is by means of a pump for periodic or continuous delivery.
  • Conventional procedures and ingredients for the selection and preparation of suitable compositions are described, for example, in Remington's Pharmaceutical Sciences (2000 - 20th edition) and in The United States Pharmacopeia: The National Formulary (USP 24 NF19) published in 1999.
  • Parenteral administration includes systemic delivery routes other than the gastrointestinal (Gl) tract, and includes, for example intravenous, intraarterial, intraperitoneal, subcutaneous, intramuscular, transepithelial, nasal, intrapulmonary (for example, by use of an aerosol), intrathecal, rectal and topical (including the use of a patch or other transdermal delivery device) modes of administration.
  • Parenteral administration may be by continuous infusion over a selected period of time.
  • a compound of the application is orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it is enclosed in hard or soft shell gelatin capsules, or it is compressed into tablets, or it is incorporated directly with the food of the diet.
  • the compound is incorporated with excipient and used in the form of ingestible tablets, buccal tablets, troches, capsules, caplets, pellets, granules, lozenges, chewing gum, powders, syrups, elixirs, wafers, aqueous solutions and suspensions, and the like.
  • carriers that are used include lactose, corn starch, sodium citrate and salts of phosphoric acid.
  • Pharmaceutically acceptable excipients include binding agents (e.g., pregelatinized maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g. , lactose, microcrystalline cellulose or calcium phosphate); lubricants (e.g., magnesium stearate, talc or silica); disintegrants (e.g., potato starch or sodium starch glycolate); or wetting agents (e.g., sodium lauryl sulphate).
  • the tablets are coated by methods well known in the art.
  • Oral dosage forms also include modified release, for example immediate release and timed- release, formulations.
  • modified-release formulations include, for example, sustained-release (SR), extended-release (ER, XR, or XL), time- release or timed-release, controlled-release (CR), or continuous-release (CR or Contin), employed, for example, in the form of a coated tablet, an osmotic delivery device, a coated capsule, a microencapsulated microsphere, an agglomerated particle, e.g., as of molecular sieving type particles, or, a fine hollow permeable fiber bundle, or chopped hollow permeable fibers, agglomerated or held in a fibrous packet.
  • SR sustained-release
  • ER extended-release
  • CR controlled-release
  • Contin continuous-release
  • Timed-release compositions are formulated, for example as liposomes or those wherein the active compound is protected with differentially degradable coatings, such as by microencapsulation, multiple coatings, etc.
  • Liposome delivery systems include, for example, small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles.
  • liposomes are formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.
  • useful carriers or diluents include lactose and dried corn starch.
  • liquid preparations for oral administration take the form of, for example, solutions, syrups or suspensions, or they are suitably presented as a dry product for constitution with water or other suitable vehicle before use.
  • aqueous suspensions and/or emulsions are administered orally, the compound of the application is suitably suspended or dissolved in an oily phase that is combined with emulsifying and/or suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents are added.
  • Such liquid preparations for oral administration are prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, methyl cellulose or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters or ethyl alcohol); and preservatives (e.g., methyl or propyl p-hydroxybenzoates or sorbic acid).
  • suspending agents e.g., sorbitol syrup, methyl cellulose or hydrogenated edible fats
  • emulsifying agents e.g., lecithin or acacia
  • non-aqueous vehicles e.g., almond oil, oily esters or ethyl alcohol
  • preservatives e.g., methyl or propyl p-hydroxybenzoates or sorbic acid.
  • Useful diluents include lactose and high mo
  • a compound of the application is administered parenterally.
  • solutions of a compound of the application are prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose.
  • dispersions are prepared in glycerol, liquid polyethylene glycols, DMSO and mixtures thereof with or without alcohol, and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. A person skilled in the art would know how to prepare suitable formulations.
  • sterile solutions of the compounds of the application are usually prepared, and the pH's of the solutions are suitably adjusted and buffered.
  • ointments or droppable liquids are delivered, for example, by ocular delivery systems known to the art such as applicators or eye droppers.
  • ocular delivery systems known to the art such as applicators or eye droppers.
  • such compositions include mucomimetics such as hyaluronic acid, chondroitin sulfate, hydroxypropyl methylcellulose or polyvinyl alcohol, preservatives such as sorbic acid, EDTA or benzyl chromium chloride, and the usual quantities of diluents or carriers.
  • diluents or carriers will be selected to be appropriate to allow the formation of an aerosol.
  • a compound of the application is formulated for parenteral administration by injection, including using conventional catheterization techniques or infusion.
  • Formulations for injection are, for example, presented in unit dosage form, e.g., in ampoules or in multi- dose containers, with an added preservative.
  • the compositions take such forms as sterile suspensions, solutions or emulsions in oily or aqueous vehicles, and contain formulating agents such as suspending, stabilizing and/or dispersing agents. In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists.
  • the compounds of the application are suitably in a sterile powder form for reconstitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • compositions for nasal administration are conveniently formulated as aerosols, drops, gels and powders.
  • the compounds of the application are conveniently delivered in the form of a solution, dry powder formulation or suspension from a pump spray container that is squeezed or pumped by the patient or as an aerosol spray presentation from a pressurized container or a nebulizer.
  • Aerosol formulations typically comprise a solution or fine suspension of the active substance in a physiologically acceptable aqueous or non-aqueous solvent and are usually presented in single or multidose quantities in sterile form in a sealed container, which, for example, take the form of a cartridge or refill for use with an atomising device.
  • the sealed container is a unitary dispensing device such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve which is intended for disposal after use.
  • the dosage form comprises an aerosol dispenser
  • it will contain a propellant which is, for example, a compressed gas such as compressed air or an organic propellant such as fluorochlorohydrocarbon.
  • a propellant include but are not limited to dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, heptafluoroalkanes, carbon dioxide or another suitable gas.
  • the dosage unit is suitably determined by providing a valve to deliver a metered amount.
  • the pressurized container or nebulizer contains a solution or suspension of the active compound.
  • Capsules and cartridges made, for example, from gelatin) for use in an inhaler or insufflator are, for example, formulated containing a powder mix of a compound of the application and a suitable powder base such as lactose or starch.
  • the aerosol dosage forms can also take the form of a pump-atomizer.
  • compositions suitable for buccal or sublingual administration include tablets, lozenges, and pastilles, wherein a compound of the application is formulated with a carrier such as sugar, acacia, tragacanth, or gelatin and glycerine.
  • Compositions for rectal administration are conveniently in the form of suppositories containing a conventional suppository base such as cocoa butter.
  • Suppository forms of the compounds of the application are useful for vaginal, urethral and rectal administrations.
  • Such suppositories will generally be constructed of a mixture of substances that is solid at room temperature but melts at body temperature.
  • the substances commonly used to create such vehicles include but are not limited to theobroma oil (also known as cocoa butter), glycerinated gelatin, other glycerides, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol. See, for example: Remington's Pharmaceutical Sciences, 16th Ed., Mack Publishing, Easton, PA, 1980, pp. 1530-1533 for further discussion of suppository dosage forms.
  • a compound of the application is coupled with soluble polymers as targetable drug carriers.
  • soluble polymers include, for example, polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxy-ethylaspartamide- phenol, or polyethyleneoxide-polylysine substituted with palmitoyl residues.
  • a compound of the application is coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and crosslinked or amphipathic block copolymers of hydrogels.
  • a drug for example, polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and crosslinked or amphipathic block copolymers of hydrogels.
  • a compound of the application including pharmaceutically acceptable salts, solvates and/or prodrugs thereof is suitably used on their own but will generally be administered in the form of a pharmaceutical composition in which the one or more compounds of the application (the active ingredient) is in association with a pharmaceutically acceptable carrier.
  • the pharmaceutical composition will comprise from about 0.05 wt% to about 99 wt% or about 0.10 wt% to about 70 wt%, of the active ingredient, and from about 1 wt% to about 99.95 wt% or about 30 wt% to about 99.90 wt% of a pharmaceutically acceptable carrier, all percentages by weight being based on the total composition.
  • a compound of the application is either used alone or in combination with other known agents useful for treating diseases, disorders or conditions that treatable by inhibition of EGFR, and those that are treatable with an EGFR inhibitor.
  • a compound of the application is administered contemporaneously with those agents.
  • "contemporaneous administration" of two substances to a subject means providing each of the two substances so that they are both active in the individual at the same time.
  • the exact details of the administration will depend on the pharmacokinetics of the two substances in the presence of each other, and can include administering the two substances within a few hours of each other, or even administering one substance within 24 hours of administration of the other, if the pharmacokinetics are suitable. Design of suitable dosing regimens is routine for one skilled in the art.
  • two substances will be administered substantially simultaneously, i.e., within minutes of each other, or in a single composition that contains both substances.
  • a combination of agents is administered to a subject in a non-contemporaneous fashion.
  • a compound of the present application is administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form.
  • the present application provides a single unit dosage form comprising one or more compounds of the application, an additional therapeutic agent, and a pharmaceutically acceptable carrier.
  • the dosage of a compound of the application varies depending on many factors such as the pharmacodynamic properties of the compound, the mode of administration, the age, health and weight of the recipient, the nature and extent of the symptoms, the frequency of the treatment and the type of concurrent treatment, if any, and the clearance rate of the compound in the subject to be treated.
  • a compound of the application is administered initially in a suitable dosage that is adjusted as required, depending on the clinical response. Dosages will generally be selected to maintain a serum level of the compound of the application from about 0.01 ⁇ g/cc to about 1000 g/cc, or about 0.1 ⁇ g/cc to about 100 ⁇ g/cc.
  • oral dosages of one or more compounds of the application will range between about 1 mg per day to about 1000 mg per day for an adult, suitably about 1 mg per day to about 500 mg per day, more suitably about 1 mg per day to about 200 mg per day.
  • a representative amount is from about 0.001 mg/kg to about 10 mg/kg, about 0.01 mg/kg to about 10 mg/kg, about 0.01 mg/kg to about 1 mg/kg or about 0.1 mg/kg to about 1 mg/kg will be administered.
  • a representative amount is from about 0.001 mg/kg to about 10 mg/kg, about 0.1 mg/kg to about 10 mg/kg, about 0.01 mg/kg to about 1 mg/kg or about 0.1 mg/kg to about 1 mg/kg.
  • a representative amount is from about 0.1 mg/kg to about 10 mg/kg or about 0.1 mg/kg to about 1 mg/kg.
  • compositions are formulated for oral administration and the one or more compounds are suitably in the form of tablets containing 0.25, 0.5, 0.75, 1 .0, 5.0, 10.0, 20.0, 25.0, 30.0, 40.0, 50.0, 60.0, 70.0, 75.0, 80.0, 90.0, 100.0, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950 or 1000 mg of active ingredient per tablet.
  • the one or more compounds of the application are administered in a single daily, weekly or monthly dose or the total daily dose is divided into two, three or four daily doses.
  • a compound also includes embodiments wherein one or more compounds are referenced.
  • the present application includes a method for inhibiting EGFR in a cell, either in a biological sample or in a patient, comprising administering an effective amount of one or more compounds of the application to the cell.
  • the application also includes a use of one or more compounds of the application for inhibiting EGFR in a cell as well as a use of one or more compounds of the application for the preparation of a medicament for inhibiting EGFR in a cell.
  • the application further includes one or more compounds of the application for use in inhibiting EGFR in a cell.
  • the compounds of the application have been shown to be capable of inhibiting EGFR protein activity, the compounds of the application are useful for treating diseases, disorders or conditions treatable by the inhibition of EGFR. Therefore the compounds of the present application are useful as medicaments. Accordingly, the present application includes a compound of the application for use as a medicament.
  • the present application also includes a method of treating a disease, disorder or condition treatable by inhibition of EGFR comprising administering a therapeutically effective amount of one or more compounds of the application to a subject in need thereof.
  • the present application also includes a use of one or more compounds of the application for treatment of a disease, disorder or condition treatable by inhibition of EGFR as well as a use of one or more compounds of the application for the preparation of a medicament for treatment of a disease, disorder or condition treatable by inhibition of EGFR.
  • the application further includes one or more compounds of the application for use in treating a disease, disorder or condition treatable by inhibition of EGFR.
  • the disease, disorder or condition is a neoplastic disorder.
  • the present application also includes a method of treating a neoplastic disorder comprising administering a therapeutically effective amount of one or more compounds of the application to a subject in need thereof.
  • the present application also includes a use of one or more compounds of the application for treatment of a neoplastic disorder as well as a use of one or more compounds of the application for the preparation of a medicament for treatment of a neoplastic disorder.
  • the application further includes one or more compounds of the application for use in treating a neoplastic disorder.
  • the treatment is in an amount effective to ameliorate at least one symptom of the neoplastic disorder, for example, reduced cell proliferation or reduced tumor mass, among others, in a subject in need of such treatment.
  • the disease, disorder or condition treatable by inhibition of EGFR is cancer.
  • the present application also includes a method of treating cancer comprising administering a therapeutically effective amount of one or more compounds of the application to a subject in need thereof.
  • the present application also includes a use of one or more compounds of the application for treatment of cancer as well as a use of one or more compounds of the application for the preparation of a medicament for treatment of cancer.
  • the application further includes one or more compounds of the application for use in treating cancer.
  • the compound is administered for the prevention of cancer in a subject such as a mammal having a predisposition for cancer.
  • the cancer is a solid cancer or a so-called liquid cancer, and can be selected from a cancer of the skin, blood, prostate, colorectum, pancreas, kidney, ovary, breast, for example mammary, liver, tongue and lung.
  • the cancer is selected from leukaemia, lymphoma, non-Hodgkin's lymphoma and multiple myeloma.
  • the cancer target includes particularly those for which regulatory approval has already been granted for other EGFR inhibitors. These cancers include colorectal cancer, head and neck cancer, pancreatic cancer, non-small cell lung cancer, and glioma.
  • the disease, disorder or condition is a disease, disorder or condition associated with an uncontrolled and/or abnormal cellular activity affected directly or indirectly by alteration of EGFR protein activity.
  • the uncontrolled and/or abnormal cellular activity that is affected directly or indirectly by altered EGFR activity is proliferative activity in a cell.
  • the application also includes a method of inhibiting proliferative activity in a cell, comprising administering an effective amount of one or more compounds of the application to the cell.
  • the present application also includes a use of one or more compounds of the application for inhibition of proliferative activity in a cell as well as a use of one or more compounds of the application for the preparation of a medicament for inhibition of proliferative activity in a cell.
  • the application further includes one or more compounds of the application for use in inhibiting proliferative activity in a cell.
  • the present application also includes a method of inhibiting uncontrolled and/or abnormal cellular activities affected directly or indirectly by EGFR protein in a cell, either in a biological sample or in a subject, comprising administering an effective amount of one or more compounds of the application to the cell.
  • the application also includes a use of one or more compounds of the application for inhibition of uncontrolled and/or abnormal cellular activities affected directly or indirectly by EGFR protein in a cell as well as a use of one or more compounds of the application for the preparation of a medicament for inhibition of uncontrolled and/or abnormal cellular activities affected directly or indirectly by EGFR protein inhibition in a cell.
  • the application further includes one or more compounds of the application for use in inhibiting uncontrolled and/or abnormal cellular activities affected directly or indirectly by EGFR.
  • the present application also includes a method of treating a disease, disorder or condition that is treatable by inhibition of EGFR comprising administering a therapeutically effective amount of one or more compounds of the application in combination with another known agent useful for such treatment.
  • the present application also includes a use of one or more compounds of the application in combination with another known agent useful for treatment of a disease, disorder or condition treatable by inhibition of EGFR for treatment of a disease, disorder or condition treatable by inhibition of EGFR as well as a use of one or more compounds of the application in combination with another known agent useful for treatment of a disease, disorder or condition treatable by inhibition of EGFR, for the preparation of a medicament for treatment of a disease, disorder or condition treatable by inhibition of EGFR.
  • the application further includes one or more compounds of the application in combination with another known agent useful for treatment of a disease, disorder or condition treatable by inhibition of EGFR for use in treating a disease, disorder or condition treatable by inhibition of EGFR.
  • the disease, disorder or condition treatable by inhibition of EGFR is a cancer such as multiple myeloma, lymphoma, leukemia, ovarian cancer, brain cancer, lung cancer, and pancreatic cancer.
  • Treatable EGFR-mediated cancers thus include benign or malignant tumors (e.g., renal, liver, kidney, bladder, breast, gastric, ovarian, colorectal, prostate, pancreatic, lung, vulva, and thyroid); hepatic carcinomas; sarcomas; glioblastomas; and various head and neck tumors including particularly head and neck cancers and especially squamous cell carcinoma of the head and neck, colorectal cancers, gastrointestinal cancers, brain tumours including glioblastomas, and tumours of the lung including non-small-cell lung carcinoma, and of the breast, pancreas, esophagus, kidney, ovary, cervix and prostate.
  • benign or malignant tumors e.g., renal, liver, kidney, bladder, breast, gastric, ovarian, colorectal, prostate, pancreatic, lung, vulva, and thyroid
  • hepatic carcinomas e.g., renal, liver, kidney, bladder, breast, gastric
  • the disease, disorder or condition treatable by inhibition of EGFR is cancer and the one or more compounds of the application are administered in combination with one or more additional cancer treatments.
  • the additional cancer treatment is selected from radiotherapy, chemotherapy, targeted therapies such as antibody therapies and small molecule therapies such as tyrosine-kinase inhibitors, immunotherapy, hormonal therapy and anti-angiogenic therapies.
  • Example 17 Determination of hERG activity by Manual Patch-Clamp
  • HEK 293 cell line stably expressing hERG channel (Cat. K1236) was purchased from Invitrogen. The cells were cultured in medium containing of 85% DMEM, 10% dialyzed FBS, 0.1 mM NEAA, 25mM HEPES, 100U/ml_ Penicillin-Streptomycin and 5 ⁇ g/mL Blasticidin and 400 ⁇ g/mL Geneticin. Cells were split using TrypLETM Express about three times a week, and maintained between -40% to -80% confluence. Before the assay, the cells were seeded onto the coverslips at 5 ⁇ 105cells /per 6 cm cell culture dish and induced with doxycycline at 1 ⁇ g/ml for 48 hours.
  • Test compounds were initially prepared in DMSO with final concentration of 30 mM as stock solution according to SOP-ADMET-MAN-007.
  • Normal test pulse current waveform e.g., hERG peak tail
  • current amplitude greater than prepulse current amplitude and the peak tail amplitude >250 pA
  • ICso values for representative compounds of Formula I are presented in table 1 .
  • Representative compounds of the application have IC50 values that are approximately 1 order of magnitude greater than their corresponding nitrogen counterparts (disclosed in WO2016/123706 A1 ) and therefor these compounds have a significantly lower risk for cardiovascular side effects.
  • Table 1
  • Streptavidin-coated magnetic beads were treated with biotinylated small molecule ligands for 30 minutes at room temperature to generate affinity resins for kinase assays.
  • the liganded beads were blocked with excess biotin and washed with blocking buffer (SeaBlock (Pierce), 1 % BSA, 0.05 % Tween 20, 1 mM DTT) to remove unbound ligand and to reduce non-specific phage binding.
  • Binding reactions were assembled by combining kinases, liganded affinity beads, and test compounds in 1x binding buffer (20 % SeaBlock, 0.17x PBS, 0.05 % Tween 20, 6 mM DTT).
  • Test compounds were prepared as 40x stocks in 100% DMSO and directly diluted into the assay. All reactions were performed in polypropylene 384-well plates in a final volume of 0.04 ml. The assay plates were incubated at room temperature with shaking for 1 hour and the affinity beads were washed with wash buffer (1x PBS, 0.05 % Tween 20). The beads were then re- suspended in elution buffer (1x PBS, 0.05 % Tween 20, 0.5 ⁇ non-biotinylated affinity ligand) and incubated at room temperature with shaking for 30 minutes. The kinase concentration in the eluates was measured by qPCR.
  • the hydrochloride salt of compound 1-1 (1-1 -HCI) was assessed against a panel of 486 kinases, including WT EGFR, EGFR mutant and ephrin receptor tyrosine kinases.
  • Ultrasensitive quantitative PCR qPCR was used to measure levels of immobilized kinases after treatment with 1-1 -HCI @ 300 nM.
  • 1-1 -HCI exhibited inhibitory activity against WT and certain mutant EGFR kinases containing activating mutations, such as the E746-A750del, L747- E749del/A750P and L747-S752del/P753S mutants (see Table 2).
  • the compound proved ineffective against the T790M mutant as well as related kinases ERBB2 and ERBB3.
  • P-glycoprotein is a member of the ABC-transporter family that transports substances across cellular membranes acting as an energy- dependent efflux pump extruding drugs out of the cells. Increased expression of Pgp in cancer cells is one of the major mechanisms of cancer resitances and chemotherapy and thus Pgp plays a key role on the pharmacokinetics of drug absorption and distribution.
  • Caco-2 cells were diluted to 6.86x10 5 cells/mL with culture medium and 50 ⁇ _ of cell suspension were dispensed into the filter well of the 96-well HTS Transwell plate. Cells were cultivated for 14-18 days in a cell culture incubator at 37 °C, 5% C02, 95% relative humidity. Cell culture medium was replaced every 2 days, beginning no later than 24 hours after initial plating.
  • TEER of each well was determined.
  • the TEER value of each well should be greater than 230 ohms- cm 2 .
  • TEER value (ohm- cm 2 ) TEER measurement (ohms) x Area of membrane (cm 2 )
  • Stock solutions of 1-1 and control compounds were prepared in DMSO at 10 mM and then diluted to 1 ⁇ with HBSS (10 mM HEPES, pH 7.4,) to get compound working solution.
  • HBSS mM HEPES, pH 7.4
  • 75 ⁇ _ of compound working solution was added to the filter well (apical compartment) and 235 ⁇ _ of HBSS (10 mM HEPES, pH 7.4) to receiver plate (basolateral compartment).
  • Plates were incubated for 2 hours at 37 °C with shaking at 150 rpm on a rotary shaker. At the end of the transport period, aliquots of 50 ⁇ _ were removed directly from the apical and basolateral wells and transferred to wells of new plates. Four volumes of cold methanol containing internal standards (IS, 100 nM Alprazolam, 200 nM Labetalol and 200 nM Diclofenac) was added into each well. Samples were centrifuged at 3,220 g for 30 minutes. Aliquots of 200 ⁇ _ supernatant were used for LC-MS/MS analysis.
  • LC/MS analysis was performed for all samples from this study using a Shimadzu liquid chromatograph separation system equipped with degasser DGU-20A3, solvent delivery unit LC-20AD, system controller CBM- 20A, column oven CTO-10ASVP and CTC Analytics HTC PAL System. Mass spectrometric analysis was performed using an API 5500 instrument from AB Inc (Canada) with an ESI interface. The data acquisition and control system were created using Analyst 1 .5 software from ABI Inc. Results
  • Activity as a Pgp substrate is an undesirable trait for an anticancer compound.
  • Compounds that are substrates for Pgp will be more readily transported out of a cancer cell and therefore show reduced activity.
  • Results for representative compounds of the application are shown in Table 3. As can be seen, 1-1 exhibits a significantly low efflux ratio. In fact the efflux ratio for 1-1 is well below the desirable range for anticancer drugs.
  • Base Reaction buffer 20 mM Hepes (pH 7.5), 10 mM MgC , 1 mM EGTA, 0.02% Brij35, 0.02 mg/ml BSA, 0.1 mM Na 3 V0 4 , 2 mM DTT, 1 % DMSO
  • Example 21 Plasma, Brain and Lung exposure ofl-1-HCI in Male SD Rats
  • the objective of this study was to determine the plasma pharmacokinetic profile and the brain penetration, lung penetration of 1-1 -HCI in fed male SD Rats after single intravenous (IV) and oral (PO) administrations.
  • Study Design Total 36 male SD Rats were assigned to 3 groups as shown in the table below. 1-1 -HCI was administered once intravenously (2 mg/kg, free form) at 10 mL/kg and orally by gavage (25 mg/kg, free form) at a dose volume of 10 mL/kg, respectively. Plasma samples were collected at each time point after IV and PO administration, lung and brain samples were just collected at each time point after PO administration at 25 mg/kg (free form).
  • DOSE 2 Dissolved 133.96 mg of 1-1 -HCI in 0.250 mL of DMSO with vortexing 1 minute, sonification 2 minutes, and added 0.499 mL of Tween-20 with vortexing 1 minute, sonification 2 minutes, then added 49.157 mL of 0.5% ⁇ - ⁇ -CD in water (w/v) with vortexing 5 minutes to obtain a suspension with concentration at 2.5 mg/mL of 1-1 -HCI.
  • Blood Samples For IV (2 mg/kg) administration, blood samples were collected from each animal at 0.083, 0.25, 0.5, 1 .5, 3, 6, 9, 12, 24 and 48 hour post-dose.
  • For PO (25 mg/kg) administration blood samples were collected from each animal at 0.25, 0.5, 1 .5, 3, 6, 9, 12, 24 and 48 hour post- dose.
  • For PO (25 mg/kg) administration blood, lung and brain samples were collected from each animal at 0.5, 1.5, 3, 6, 9, 12 and 24 hour post-dose.
  • Blood samples were collected from each animal via jugular vein (PO 25 mg/kg: heart puncture). These blood samples were placed into the tubes containing K2EDTA. The whole blood tubes were inverted several times and then centrifuged at 2000 g for 5 minutes at 4°C to obtain plasma. The plasma samples were stored frozen at -75 ⁇ 15°C until analysis.
  • Brain and lung samples were collected after animals being fully exsanguinated. Procedure: opened chest cavity, cut ventricle and performed a gentle IV saline flush (saline flush volume ⁇ 20 mL) with the animal head placed down at a 45 degree angle to facilitate blood removal. Brain and lung samples were collected at adopted time points, quick frozen in ice box. The brain and lung samples were stored frozen at -75 ⁇ 15 °C until analysis.
  • Sample treatment All of the brain and lung samples were weighed and homogenized with water by brain weight and lung weight (g) to water volume (mL) using a ratio of 1 :3 before analysis. The actual concentration was the detected value multiplied by the dilution factor. 5 ⁇ of each calibration standard working solution (5, 10, 20, 50, 100, 500, 1000, 5000 and 10000 ng/mL) was added to 50 ⁇ _ of the blank SD rat plasma (or blank SD rat brain homogenate and lung homogenate) to achieve calibration standards of 0.5- 1000 ng/mL (0.5, 1 , 2, 5, 10, 50, 100, 500, 1000 ng/mL) in a total volume of 55 ⁇ .
  • Quality Control (QC) samples at 1 ng/mL (low), 2 ng/mL (low), 50 ng/mL (mid), 800 ng/mL (high) were prepared from the QC working solutions in the same way as calibration standards.
  • 55 ⁇ of standards, 55 ⁇ of QC samples and 55 ⁇ of unknown samples 50 ⁇ of plasma or brain homogenate, lung homogenate with 5 ⁇ 50% acetonitrile
  • the samples were vortexed for 30 sec. After centrifugation at 4°C, 4000 rpm for 15 min, 15 ⁇ of the supernatant was injected into the LC-MS/MS system for quantitative analysis. All samples were processed on ice.
  • LC-MS/MS conditions The LC-MS/MS system consisted of two Shimadzu LC-30AD pumps, a DGU-20A5R degasser, a Rack changer II and an AB API4000LC-MS/MS mass spectrometer. Chromatographic separation was performed on a Phenomenex Synergi 4 ⁇ Polar-RP (50 ⁇ 2.00 mm) 80A column at room temperature. The mobile phase was composed of A: 5% acetonitrile (0.1 % formic acid); B: 95% acetonitrile (0.1 % formic acid). The flow rate was 0.7 mL/min. The injection volume was 15 ⁇ .
  • ESI Positive mode electrospray ionization
  • ISO Dexamethasone
  • MRM multiple reaction monitoring
  • the optimized transitions were 495.99 ⁇ 334.00 and 393.40 ⁇ 373.30 for 1-1 -HCI and Dexamethasone, respectively.
  • the instrument parameters were set as follows: ion spray voltage: 5500 V; curtain gas: 40 psi; nebulizer gas: 50 psi; turbo gas: 50 psi; collision gas: 10 psi; temperature: 450 °C.
  • the ultimate test of a lead drug candidate is efficacy in an in vivo model.
  • various ADME parameters are tested in isolation of one another to ascertain whether a drug can be viable in an in vivo setting.
  • all ADME parameters are working in unison to elucidate a proper pharmacokinetic profile for the drug.
  • 1-1 -HCI brain, lung and plasma distribution was evaluated after administration to male SD Rats.
  • the Cmax ratio and AUCIast ratio of plasma: brain were 0.552 and 0.512.
  • the Cmax ratio and AUCIast ratio of plasma: lung were 0.412 and 0.341 .
  • distribution to brain and lung were more than plasma exposure.
  • Approximately a 195 % and 293% increase in 1-1 -HCI exposure was observed in brain and lung, respectively, compared to plasma.
  • Table 9 presents a comparison of the brain penetrations of 1-1 -HCI and Erlotinib.
  • 1-1 -HCI was dissolved in 0.5% CMC/ 1 % Tween-20/ 0.5%HP- ⁇ - CD with vortexing for 5 minutes, followed by sonication for 20 minutes to prepare a uniform suspension with concentrations of 5, 10, and 20 mg/kg.
  • 1-1 - HCI was administered to male SD rats at the aforementioned concentrations orally at a dosing frequency of 1 dose/day for 7 days. All study animals were monitored for behavior, such as mobility, food and water consumption, body weight (BW), eye/hair matting and other signs of physical decline. Animals that were observed to be in a continuing deteriorating condition were euthanized prior to death, or before reaching a comatose state. Animals exhibiting obvious signs of severe distress and/or pain were humanely sacrificed by carbon dioxide followed by cervical dislocation to ensure death.
  • the organs (adrenals, heart, liver, spleen, lung, kidneys, brain (1 ⁇ 2 snap frozen and 1 ⁇ 2 fix), Gl (stomach, small and large intestine), pancreas, testes, thymus) were collected and organs weight were recorded followed by fixation of all collected tissue samples in 10% formalin. Bone marrow was collected and prepared immediately into bone marrow smear slides for later examination.
  • Serum samples were used for blood chemistry analysis of the following parameters: TBA, AST, ALT, ALP, ALB, TP, TBIL, DBIL, GGT, UA, UREA, CRE, TG, TCHO, HDL-C, LDL-C, FFA and CK.
  • Blood smears were prepared by placing a drop of blood approximately 4 mm in diameter on a glass slide and speading the droplet with the edge of a separate glass slide. Smears were air-dried, and then dipped into 100% methanol for 5 minutes to complete the fixing process.
  • Bone marrow smears were extracted from the femur at the level of the proximal diaphysis. Marrow was removed using curved forceps and was added to a drop of bovine serum albumin on a glass slide. The marrow was gently mixed with the albumin and spread using the edge of a separate glass slide. The smear was air-dried and fixed in 100% methanol for 5 minutes.
  • 1-1 -HCI exhibited no signs of significant body weight loss or any gross clinical abnormalities at the doses administered (5, 10, and 20 mg/kg, p.o).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne une nouvelle classe de dérivés fluorés de formule I qui ont été préparés et se sont avérés utiles dans le traitement de cancers et d'autres troubles liés au récepteur du facteur de croissance épidermique (EGFR). Les composés de formule I présentent une excellente pénétration du cerveau et un profil d'effet secondaire amélioré par rapport aux composés précédemment connus (par exemple) l'erlotinib.
PCT/CA2018/051282 2017-10-12 2018-10-11 Nouveaux dérivés de 4-aryloxyquinazoline fluorés utilisés en tant qu'inhibiteurs d'egfr utiles pour le traitement de cancers WO2019071351A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762571441P 2017-10-12 2017-10-12
US62/571,441 2017-10-12

Publications (1)

Publication Number Publication Date
WO2019071351A1 true WO2019071351A1 (fr) 2019-04-18

Family

ID=66100207

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2018/051282 WO2019071351A1 (fr) 2017-10-12 2018-10-11 Nouveaux dérivés de 4-aryloxyquinazoline fluorés utilisés en tant qu'inhibiteurs d'egfr utiles pour le traitement de cancers

Country Status (1)

Country Link
WO (1) WO2019071351A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020261156A1 (fr) 2019-06-28 2020-12-30 Array Biopharma Inc. Dérivés de quinazoline-4-one utiles pour le traitement de maladies et de troubles associés à braf
WO2021250521A1 (fr) 2020-06-09 2021-12-16 Array Biopharma Inc. Composés de 4-oxo-3,4-dihydroquinazolinone pour le traitement de maladies et d'affections associées à braf
WO2022208391A1 (fr) 2021-03-31 2022-10-06 Pfizer Inc, 3,4-dihydro-2,7-naphtyridine-1,6 (2h,7h)-diones utilisés en tant qu'inhibiteurs de mek
WO2024218632A1 (fr) 2023-04-17 2024-10-24 Array Biopharma Inc. Inhibiteurs de la protéine kinase erk

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6251912B1 (en) * 1997-08-01 2001-06-26 American Cyanamid Company Substituted quinazoline derivatives
WO2009030224A2 (fr) * 2007-09-07 2009-03-12 Schebo Biotech Ag Nouveaux produits pharmaceutiques, procédés pour les produire et leur utilisation en thérapie médicale
WO2016123706A1 (fr) * 2015-02-03 2016-08-11 Trillium Therapeutics Inc. Nouveaux dérivés fluorés utilisés en tant qu'inhibiteurs d'egfr utiles pour le traitement de cancers

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6251912B1 (en) * 1997-08-01 2001-06-26 American Cyanamid Company Substituted quinazoline derivatives
WO2009030224A2 (fr) * 2007-09-07 2009-03-12 Schebo Biotech Ag Nouveaux produits pharmaceutiques, procédés pour les produire et leur utilisation en thérapie médicale
WO2016123706A1 (fr) * 2015-02-03 2016-08-11 Trillium Therapeutics Inc. Nouveaux dérivés fluorés utilisés en tant qu'inhibiteurs d'egfr utiles pour le traitement de cancers

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
NEIL VASEDEV ET AL.: "Metabolic stability of 6,7-dialkoxy-4(2-,3- and 4-[18F]fluoroanilino)quinazolines, potential EGFR imaging probes", BIOORGANIC &MEDICINAL CHEMISTRY, vol. 19, 2011, pages 2959 - 2965, XP055592865 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020261156A1 (fr) 2019-06-28 2020-12-30 Array Biopharma Inc. Dérivés de quinazoline-4-one utiles pour le traitement de maladies et de troubles associés à braf
WO2021250521A1 (fr) 2020-06-09 2021-12-16 Array Biopharma Inc. Composés de 4-oxo-3,4-dihydroquinazolinone pour le traitement de maladies et d'affections associées à braf
WO2022208391A1 (fr) 2021-03-31 2022-10-06 Pfizer Inc, 3,4-dihydro-2,7-naphtyridine-1,6 (2h,7h)-diones utilisés en tant qu'inhibiteurs de mek
WO2024218632A1 (fr) 2023-04-17 2024-10-24 Array Biopharma Inc. Inhibiteurs de la protéine kinase erk

Similar Documents

Publication Publication Date Title
EP3037424B1 (fr) Nouveau composé substitué par une quinoline
AU2016214923A1 (en) Novel fluorinated derivatives as EGFR inhibitors useful for treating cancers
KR102603153B1 (ko) C5-아닐리노퀴나졸린 화합물 및 암의 치료에서의 이의 용도
JP6235078B2 (ja) 置換ピリダジンカルボキサミド化合物
US20190016689A1 (en) Novel fluorinated quinazoline derivatives as egfr inhibitors
WO2019071351A1 (fr) Nouveaux dérivés de 4-aryloxyquinazoline fluorés utilisés en tant qu'inhibiteurs d'egfr utiles pour le traitement de cancers
Liu et al. Design, synthesis and structure-activity relationships of novel 4-phenoxyquinoline derivatives containing 1, 2, 4-triazolone moiety as c-Met kinase inhibitors
CN109311852B (zh) 作为egfr抑制剂的苯胺嘧啶化合物的结晶
TW201638092A (zh) 嘧啶並吡咯類化合物、其製備方法、藥用組合物及其應用
WO2014040549A1 (fr) Composé cyclique hétéroaromatique alcynylique et son application
US20190100509A1 (en) Salt of pyridinyl amino pyrimidine derivative, preparation method therefor, and application thereof
EP3897848B1 (fr) Traitements du cancer
US9624218B2 (en) Pyrido[2,3-d]pyrimidin-4-one compounds as tankyrase inhibitors
WO2017023899A1 (fr) Inhibiteurs de la tyrosine kinase ack1/tnk2
CN105130907A (zh) 嘧啶类化合物及其用途
KR100714370B1 (ko) 항종양 활성을 갖는〔1,2,4〕―트리아졸로〔4,3―c〕퀴나졸린 유도체

Legal Events

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

Ref document number: 18866706

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18866706

Country of ref document: EP

Kind code of ref document: A1