EP3325447A1 - Substituted quinazoline compounds and their use as inhibitors of g12c mutant kras, hras and/or nras proteins - Google Patents

Substituted quinazoline compounds and their use as inhibitors of g12c mutant kras, hras and/or nras proteins

Info

Publication number
EP3325447A1
EP3325447A1 EP16747678.7A EP16747678A EP3325447A1 EP 3325447 A1 EP3325447 A1 EP 3325447A1 EP 16747678 A EP16747678 A EP 16747678A EP 3325447 A1 EP3325447 A1 EP 3325447A1
Authority
EP
European Patent Office
Prior art keywords
compound
alkyl
cancer
halo
cyano
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
EP16747678.7A
Other languages
German (de)
French (fr)
Inventor
Liansheng Li
Jun Feng
Yun Oliver Long
Yuan Liu
Tao Wu
Pingda Ren
Yi Liu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Araxes Pharma LLC
Original Assignee
Araxes Pharma LLC
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
Priority claimed from JOP/2016/0150A external-priority patent/JOP20160150B1/en
Application filed by Araxes Pharma LLC filed Critical Araxes Pharma LLC
Publication of EP3325447A1 publication Critical patent/EP3325447A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/38Nitrogen atoms
    • C07D215/42Nitrogen atoms attached in position 4
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings

Definitions

  • the present invention is generally directed to novel compounds and methods for their preparation and use as therapeutic or prophylactic agents, for example for treatment of cancer.
  • RAS represents a group of closely related monomeric globular proteins of 189 amino acids (21 kDa molecular mass) which are associated with the plasma membrane and which bind either GDP or GTP.
  • RAS acts as a molecular switch. When RAS contains bound GDP, it is in the resting or off position and is "inactive". In response to exposure of the cell to certain growth promoting stimuli, RAS is induced to exchange its bound GDP for a GTP. With GTP bound, RAS is "switched on” and is able to interact with and activate other proteins (its "downstream targets”).
  • the RAS protein itself has a very low intrinsic ability to hydrolyze GTP back to GDP, thus turning itself into the off state.
  • GTPase-activating proteins GTPase-activating proteins
  • Any mutation in RAS which affects its ability to interact with GAP or to convert GTP back to GDP will result in a prolonged activation of the protein and consequently a prolonged signal to the cell telling it to continue to grow and divide. Because these signals result in cell growth and division, overactive RAS signaling may ultimately lead to cancer.
  • RAS proteins contain a G domain which is responsible for the enzymatic activity of RAS - the guanine nucleotide binding and the hydrolysis (GTPase reaction). It also contains a C-terminal extension, known as the CAAX box, which may be post-translationally modified and is responsible for targeting the protein to the membrane.
  • the G domain is approximately 21-25 kDa in size and it contains a phosphate binding loop (P-loop).
  • the P-loop represents the pocket where the nucleotides are bound in the protein, and this is the rigid part of the domain with conserved amino acid residues which are essential for nucleotide binding and hydrolysis (Glycine 12, Threonine 26 and Lysine 16).
  • the G domain also contains the so called Switch I (residues 30-40) and Switch II (residues 60-76) regions, both of which are the dynamic parts of the protein which are often represented as the "spring- loaded” mechanism because of their ability to switch between the resting and loaded state.
  • the key interaction is the hydrogen bonds formed by Threonine-35 and glycine- 60 with the ⁇ -phosphate of GTP which maintain Switch 1 and Switch 2 regions respectively in their active conformation. After hydrolysis of GTP and release of phosphate, these two relax into the inactive GDP conformation.
  • KRAS mutations are detected in 25-30% of tumors.
  • the rates of oncogenic mutation occurring in the NRAS and HRAS family members are much lower (8% and 3% respectively).
  • the most common KRAS mutations are found at residue G12 and G13 in the P-loop and at residue Q61.
  • G12C is a frequent mutation of KRAS gene (glycine-12 to cysteine). This mutation had been found in about 13% of cancer occurrences, about 43% of lung cancer occurrences, and in almost 100% of MYH-associates polyposis (familial colon cancer syndrome). However targeting this gene with small molecules is a challenge. Accordingly, while progress has been made in this field, there remains a need in the art for improved compounds and methods for treatment of cancer, for example by inhibition of KRAS, HRAS or NRAS. The present invention fulfills this need and provides further related advantages. BRIEF SUMMARY
  • the present invention provides compounds, including stereoisomers, pharmaceutically acceptable salts, tautomers and prodrugs thereof, which are capable of modulating G12C mutant KRAS, HRAS and/or NRAS proteins.
  • the compounds act as electrophiles which are capable of forming a covalent bond with the cysteine residue at position 12 of a KRAS, HRAS or NRAS G12C mutant protein. Methods for use of such compounds for treatment of various diseases or conditions, such as cancer, are also provided.
  • compositions comprising one or more compounds of structure (I) and a pharmaceutically acceptable carrier are also provided in various other embodiments.
  • the present invention provides a method for treatment of cancer, the method comprising administering an effective amount of a pharmaceutical composition comprising any one or more of the compounds of structure (I) to a subject in need thereof.
  • a method for inhibiting proliferation of a cell population comprising contacting the cell population with any one of the compounds of structure (I) is also provided.
  • the invention is directed to a method for treating a disorder mediated by a KRAS, HRAS or NRAS G12C mutation in a subject in need thereof, the method comprising:
  • a pharmaceutical composition comprising any one or more compounds of structure (I).
  • the invention is directed to a method for preparing a labeled KRAS, HRAS or NRAS G12C mutant protein, the method comprising reacting the KRAS, HRAS or NRAS G12C mutant with a compound of structure (I), to result in the labeled KRAS, HRAS or NRAS G12C protein.
  • Fig. 1 illustrates the enzymatic activity of RAS.
  • Fig. 2 depicts a signal transduction pathway for RAS.
  • Fig. 3 shows some common oncogenes, their respective tumor type and cumulative mutation frequencies (all tumors).
  • Amino refers to the -NH 2 radical.
  • Carboxy or “carboxyl” refers to the -C0 2 H radical.
  • Niro refers to the -N0 2 radical.
  • Alkyl refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds), having from one to twelve carbon atoms (C 1 -C 12 alkyl), preferably one to eight carbon atoms (Ci-C 8 alkyl) or one to six carbon atoms (Ci-C 6 alkyl), and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, ⁇ -propyl, 1-methylethyl (z ' so-propyl), «-butyl, «-pentyl, 1, 1-dimethylethyl (t-butyl), 3-methylhexyl, 2-methylhexyl, ethenyl, prop-l-enyl, but-l-enyl, pent-l-enyl, penta-l,4-dienyl
  • alkyl is unsaturated and thus includes alkenyls (one or more carbon- carbon double bonds) and/or alkynyls (one or more carbon-carbon triple bonds such as ethynyl and the like). Unless stated otherwise specifically in the specification, an alkyl group is optionally substituted.
  • Alkylene or “alkylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds), and having from one to twelve carbon atoms, e.g., methylene, ethylene, propylene, «-butylene, ethenylene, propenylene, «-butenylene, propynylene, «-butynylene, and the like.
  • the alkylene chain is attached to the rest of the molecule through a single or double bond and to the radical group through a single or double bond.
  • the points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, an alkylene chain is optionally substituted.
  • Alkoxy refers to a radical of the formula -OR a where R a is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkoxy group is optionally substituted.
  • Alkoxyalkyl refers to a radical of the formula -Rt,OR a where R a is an alkyl radical as defined above containing one to twelve carbon atoms and Rb is an alkylene radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkoxyalkyl group is optionally substituted.
  • Alkylaminyl refers to a radical of the formula - HR a or - R a R a where each R a is, independently, an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkylaminyl group is optionally substituted.
  • Aminylalkyl refers to an alkyl group comprising at least one aminyl substituent (- R a R b wherein R a and R are each independently H or Ci-C 6 alkyl).
  • the aminyl substituent can be on a tertiary, secondary or primary carbon. Unless stated otherwise specifically in the specification, an aminylalkyl group is optionally substituted.
  • Aminylalkylaminyl refers to a radical of the formula - R a R b wherein R a is H or Ci-C 6 alkyl and R b is aminylalkyl. Unless stated otherwise specifically in the specification, an aminylalkylaminyl group is optionally substituted.
  • Aminylalkoxy refers to a radical of the formula -OR a H 2 wherein R a is alkylene. Unless stated otherwise specifically in the specification, an aminylalkoxy group is optionally substituted.
  • Alkylaminylalkoxy refers to a radical of the formula -OR a R b R c wherein R a is alkylene and R b and R c are each independently H or Ci-C 6 alkyl, provided one of R b or R c is Ci-C 6 alkyl. Unless stated otherwise specifically in the specification, an alkylaminylalkoxy group is optionally substituted.
  • R a is an alkyl radical as defined above containing one to twelve carbon atoms and R is alkylene. Unless stated otherwise specifically in the specification, an alkylcarbonylaminylalkoxy group is optionally substituted.
  • Aryl refers to a carbocyclic ring system radical comprising hydrogen, 6 to 18 carbon atoms and at least one aromatic ring.
  • the aryl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems.
  • Aryl radicals include, but are not limited to, aryl radicals derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, as-indacene, s-indacene, indane, indene, naphthalene, phenalene, phenanthrene, pleiadene, pyrene, and triphenylene.
  • Aralkyl refers to a radical of the formula -R b -R c where R b is an alkylene chain as defined above and R c is one or more aryl radicals as defined above, for example, benzyl, diphenylmethyl and the like. Unless stated otherwise specifically in the specification, an aralkyl group is optionally substituted.
  • Carboxyalkyl refers to a radical of the formula -R b -R c where R b is an alkylene chain as defined above and R c is a carboxyl group as defined above. Unless stated otherwise specifically in the specification, carboxyalkyl group is optionally substituted.
  • Cyanoalkyl refers to a radical of the formula -R b -R c where R b is an alkylene chain as defined above and R c is a cyano group as defined above. Unless stated otherwise specifically in the specification, a cyanoalkyl group is optionally substituted.
  • Carbocyclic or “carbocycle” refers to a ring system, wherein each of the ring atoms are carbon.
  • Cycloalkyl refers to a stable non-aromatic monocyclic or polycyclic carbocyclic radical consisting solely of carbon and hydrogen atoms, which may include fused or bridged ring systems, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond.
  • Monocyclic radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • Polycyclic radicals include, for example, adamantyl, norbornyl, decalinyl,
  • cycloalkylene is a divalent cycloalkyl, which typically connects one portion a molecule to another portion. Unless otherwise stated specifically in the specification, a cycloalkyl (or cycloalkylene) group is optionally substituted.
  • Cycloalkylalkyl refers to a radical of the formula -3 ⁇ 43 ⁇ 4 where 3 ⁇ 4 is an alkylene chain as defined above and R d is a cycloalkyl radical as defined above. Unless stated otherwise specifically in the specification, a cycloalkylalkyl group is optionally substituted.
  • Cycloalkylaminyl refers to a radical of the formula - R a R b wherein R a is H or alkyl and R is cycloalkyl. Unless stated otherwise specifically in the specification, an heterocyclylalkoxy group is optionally substituted.
  • fused refers to any ring structure described herein which is fused to an existing ring structure in the compounds of the invention.
  • the fused ring is a heterocyclyl ring or a heteroaryl ring, any carbon atom on the existing ring structure which becomes part of the fused heterocyclyl ring or the fused heteroaryl ring is replaced with a nitrogen atom.
  • Halo or halogen refers to bromo, chloro, fluoro or iodo.
  • Haloalkyl refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difluorom ethyl, trichlorom ethyl, 2,2,2-trifluoroethyl, 1,2-difluoroethyl, 3-bromo-2-fluoropropyl, 1,2-dibromoethyl, and the like. Unless stated otherwise specifically in the specification, a haloalkyl group is optionally substituted.
  • Heterocyclyl or “heterocyclic ring” refers to a stable 3- to 18-membered non-aromatic ring radical having one to twelve ring carbon atoms (e.g., two to twelve) and from one to six ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur.
  • the heterocyclyl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused, spirocyclic ("spiro-heterocyclyl”) and/or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl radical is optionally oxidized; the nitrogen atom is optionally quaternized; and the heterocyclyl radical is partially or fully saturated.
  • heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl[l,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl,
  • Heterocyclylene refers to a bivalent heterocyclyl group, which typically connects one portion of a molecule to another. Unless stated otherwise specifically in the specification, a heterocyclyl and/or heterocyclylene group is optionally substituted.
  • N-heterocyclyl refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a nitrogen atom in the heterocyclyl radical. Unless stated otherwise specifically in the specification, a N-heterocyclyl group is optionally substituted.
  • Heterocyclylalkyl refers to a radical of the formula -RbRe where 3 ⁇ 4, is an alkylene chain as defined above and Re is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl is optionally attached to the alkyl radical at the nitrogen atom. Unless stated otherwise specifically in the specification, a heterocyclylalkyl group is optionally substituted.
  • Heterocyclylalkoxy refers to a radical of the formula -OR a R b wherein R a is alkylene and R is heterocyclyl. Unless stated otherwise specifically in the specification, an heterocyclylalkoxy group is optionally substituted.
  • Heterocyclylaminyl refers to a radical of the formula - R a R b wherein R a is H or alkyl and R b is heterocyclyl. Unless stated otherwise specifically in the specification, an heterocyclylalkoxy group is optionally substituted.
  • Heteroaryl refers to a 5- to 14-membered ring system radical comprising hydrogen atoms, one to thirteen ring carbon atoms, one to six ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and at least one aromatic ring.
  • the heteroaryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized.
  • Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzothiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl,
  • benzothiazolyl benzothiadiazolyl, benzo[£][l,4]dioxepinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[l,2-a]pyridinyl, carbazolyl, cinnolinyl,
  • a heteroaryl is optionally substituted.
  • N-heteroaryl refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical. Unless stated otherwise specifically in the specification, an N-heteroaryl group is optionally substituted.
  • Heteroarylalkyl refers to a radical of the formula -3 ⁇ 4R f where R is an alkylene chain as defined above and R f is a heteroaryl radical as defined above. Unless stated otherwise specifically in the specification, a heteroarylalkyl group is optionally substituted.
  • Hydroxylalkyl refers to an alkyl group comprising at least one hydroxyl substituent.
  • the -OH substituent may be on a primary, secondary or tertiary carbon. Unless stated otherwise specifically in the specification, a hydroxylalkyl group is optionally substituted. Unless stated otherwise specifically in the specification, a hydroxylalkyl group is optionally substituted.
  • Phosphoalkoxy refers to an alkoxy group, as defined herein, which is substituted with at least one phosphate group, as defined herein. Unless stated otherwise specifically in the specification, an phosphoalkoxy group is optionally substituted.
  • Thioalkyl refers to a radical of the formula -SR a where R a is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, a thioalkyl group may be optionally substituted.
  • substituted used herein means any of the above groups (e.g., alkyl, alkylene, alkoxy, alkoxyalkyl, alkoxycarbonyl, aminylalkyl, aminylalkylaminyl, aminylalkoxy, alkylaminylalkoxy, alkylaminyl, alkylcarbonylaminyl, alkylcarbonylaminylalkoxy, aminylcarbonylalkyl, thioalkyl, aryl, aralkyl, carboxyalkyl, cyanoalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclyloxy, heterocyclylaminyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N- heteroaryl, heteroarylalkyl, phosphoalkoxy, and/or hydroxy
  • “Substituted” also means any of the above groups in which one or more hydrogen atoms are replaced by a higher-order bond (e.g., a double- or triple-bond) to a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and ester groups; and nitrogen in groups such as imines, oximes, hydrazones, and nitriles.
  • substituted includes any of the above groups in which one or more hydrogen atoms are replaced
  • R g and R h are the same or different and independently hydrogen, alkyl, alkoxy, alkylaminyl, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl.
  • Substituted further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to an aminyl, cyano, hydroxyl, imino, nitro, oxo, thioxo, halo, alkyl, alkoxy, alkylaminyl, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl group.
  • each of the foregoing substituents may also be optionally substituted with one or more of the above substituents.
  • Electrophile or “electrophilic moiety” is any moiety capable of reacting with a nucleophile (e.g., a moiety having a lone pair of electrons, a negative charge, a partial negative charge and/or an excess of electrons, for example a -SH group).
  • Electrophiles typically are electron poor or comprise atoms which are electron poor.
  • an electrophile contains a positive charge or partial positive charge, has a resonance structure which contains a positive charge or partial positive charge or is a moiety in which delocalization or polarization of electrons results in one or more atom which contains a positive charge or partial positive charge.
  • the electrophiles comprise conjugated double bonds, for example an ⁇ , ⁇ -unsaturated carbonyl or ⁇ , ⁇ -unsaturated thiocarbonyl compound.
  • an effective amount refers to that amount of a compound described herein that is sufficient to effect the intended application including but not limited to disease treatment, as defined below.
  • the therapeutically effective amount may vary depending upon the intended treatment application (in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of
  • the term also applies to a dose that will induce a particular response in target cells, e.g. reduction of platelet adhesion and/or cell migration.
  • the specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of
  • treatment refers to an approach for obtaining beneficial or desired results with respect to a disease, disorder or medical condition including but not limited to a therapeutic benefit and/or a prophylactic benefit.
  • therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated.
  • a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the subject,
  • compositions are administered to a subject at risk of developing a particular disease, or to a subject reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
  • a prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
  • co-administration encompass administration of two or more agents to an animal, including humans, so that both agents and/or their metabolites are present in the subject at the same time.
  • Co-administration includes simultaneous administration in separate compositions, administration at different times in separate compositions, or administration in a composition in which both agents are present.
  • “Pharmaceutically acceptable salt” includes both acid and base addition salts.
  • “Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, camphoric acid, camphor- 10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane- 1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic
  • “Pharmaceutically acceptable base addition salt” refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol,
  • 2-diethylaminoethanol dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like.
  • Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine,
  • antagonists are used interchangeably, and they refer to a compound having the ability to inhibit a biological function of a target protein, whether by inhibiting the activity or expression of the protein, such as KRAS, HRAS or RAS G12C. Accordingly, the terms “antagonist” and “inhibitors” are defined in the context of the biological role of the target protein. While preferred antagonists herein specifically interact with (e.g. bind to) the target, compounds that inhibit a biological activity of the target protein by interacting with other members of the signal transduction pathway of which the target protein is a member are also specifically included within this definition. A preferred biological activity inhibited by an antagonist is associated with the development, growth, or spread of a tumor.
  • agonist refers to a compound having the ability to initiate or enhance a biological function of a target protein, whether by inhibiting the activity or expression of the target protein. Accordingly, the term
  • agonist is defined in the context of the biological role of the target polypeptide.
  • While preferred agonists herein specifically interact with (e.g. bind to) the target, compounds that initiate or enhance a biological activity of the target polypeptide by interacting with other members of the signal transduction pathway of which the target polypeptide is a member are also specifically included within this definition.
  • agent refers to a biological, pharmaceutical, or chemical compound or other moiety.
  • Non-limiting examples include a simple or complex organic or inorganic molecule, a peptide, a protein, an oligonucleotide, an antibody, an antibody derivative, antibody fragment, a vitamin derivative, a carbohydrate, a toxin, or a chemotherapeutic compound.
  • Various compounds can be synthesized, for example, small molecules and oligomers (e.g., oligopeptides and oligonucleotides), and synthetic organic compounds based on various core structures.
  • various natural sources can provide compounds for screening, such as plant or animal extracts, and the like.
  • Signal transduction is a process during which stimulatory or inhibitory signals are transmitted into and within a cell to elicit an intracellular response.
  • a modulator of a signal transduction pathway refers to a compound which modulates the activity of one or more cellular proteins mapped to the same specific signal transduction pathway.
  • a modulator may augment (agonist) or suppress (antagonist) the activity of a signaling molecule.
  • an “anti-cancer agent”, “anti-tumor agent” or “chemotherapeutic agent” refers to any agent useful in the treatment of a neoplastic condition.
  • One class of anti- cancer agents comprises chemotherapeutic agents.
  • “Chemotherapy” means the administration of one or more chemotherapeutic drugs and/or other agents to a cancer patient by various methods, including intravenous, oral, intramuscular, intraperitoneal, intravesical, subcutaneous, transdermal, buccal, or inhalation or in the form of a suppository.
  • cell proliferation refers to a phenomenon by which the cell number has changed as a result of division. This term also encompasses cell growth by which the cell morphology has changed (e.g., increased in size) consistent with a proliferative signal.
  • selective inhibition refers to a biologically active agent refers to the agent's ability to preferentially reduce the target signaling activity as compared to off-target signaling activity, via direct or indirect interaction with the target.
  • Subject refers to an animal, such as a mammal, for example a human.
  • the methods described herein can be useful in both human therapeutics and veterinary applications.
  • the subject is a mammal, and in some
  • the subject is human.
  • “Mammal” includes humans and both domestic animals such as laboratory animals and household pets (e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like.
  • Radionuclides e.g., actinium and thorium radionuclides
  • LET low linear energy transfer
  • beta emitters conversion electron emitters
  • high-energy radiation including without limitation x-rays, gamma rays, and neutrons.
  • An "anti-cancer agent”, “anti-tumor agent” or “chemotherapeutic agent” refers to any agent useful in the treatment of a neoplastic condition.
  • chemotherapeutic agents comprises chemotherapeutic agents.
  • “Chemotherapy” means the administration of one or more chemotherapeutic drugs and/or other agents to a cancer patient by various methods, including intravenous, oral, intramuscular, intraperitoneal, intravesical, subcutaneous, transdermal, buccal, or inhalation or in the form of a suppository.
  • Prodrug is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound described herein (e.g., compound of structure (I)).
  • prodrug refers to a precursor of a biologically active compound that is pharmaceutically acceptable.
  • a prodrug is inactive when administered to a subject, but is converted in vivo to an active compound, for example, by hydrolysis.
  • the prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam).
  • prodrugs are also meant to include any covalently bonded carriers, which release the active compound in vivo when such prodrug is administered to a
  • Prodrugs of an active compound are typically prepared by modifying functional groups present in the active compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent active compound.
  • Prodrugs include compounds wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the active compound is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively.
  • Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of a hydroxy functional group, or acetamide, formamide and benzamide derivatives of an amine functional group in the active compound and the like.
  • prodrugs include compounds of structure (I) having a phosphate, phosphoalkoxy, ester or boronic ester substituent. Without being bound by theory, it is believed that such substituents are converted to a hydroxyl group under physiological conditions. Accordingly, embodiments include any of the compounds disclosed herein, wherein a hydroxyl group has been replaced with a phosphate, phosphoalkoxy, ester or boronic ester group, for example a phosphate or phosphoalkoxy group. For example, in some embodiments a hydroxyl group on the R 1 moiety is replaced with a phosphate, phosphoalkoxy, ester or boronic ester group, for example a phosphate or alkoxy phosphate group. Exemplary prodrugs of certain embodiments thus include com ounds having one of the following R 1 moieties:
  • each R' is independently H or an optional substituent, and n is 1, 2, 3 or 4.
  • in vivo refers to an event that takes place in a subject's body.
  • Embodiments of the invention disclosed herein are also meant to encompass all pharmaceutically acceptable compounds of structure (I) being
  • isotopically-labelled by having one or more atoms replaced by an atom having a different atomic mass or mass number.
  • isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2 H, 3 H, U C, 13 C, 14 C, 13 N, 15 N, 15 0, 17 0, 18 0, 31 P, 32 P, 35 S, 18 F, 36 C1, 123 I, and 125 I, respectively.
  • radiolabeled compounds could be useful to help determine or measure the effectiveness of the compounds, by characterizing, for example, the site or mode of action, or binding affinity to pharmacologically important site of action.
  • isotopically-labeled compounds of structure (I) for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies.
  • substitution with heavier isotopes such as deuterium, i.e. 2 H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence are preferred in some circumstances.
  • Isotopically-labeled compounds of structure (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Preparations and Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
  • Certain embodiments are also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result from, for example, the oxidation, reduction, hydrolysis, amidation, esterification, and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the embodiments include compounds produced by a process comprising administering a compound of this invention to a mammal for a period of time sufficient to yield a metabolic product thereof. Such products are typically identified by administering a radiolabeled compound of the invention in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to human, allowing sufficient time for metabolism to occur, and isolating its conversion products from the urine, blood or other biological samples.
  • an animal such as rat, mouse, guinea pig, monkey, or to human
  • “Stable compound” and “stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent. Often crystallizations produce a solvate of the compound of the invention.
  • the term “solvate” refers to an aggregate that comprises one or more molecules of a compound of the invention with one or more molecules of solvent.
  • the solvent is water, in which case the solvate is a hydrate.
  • the solvent is an organic solvent.
  • the compounds of the present invention may exist as a hydrate, including a
  • the compound of the invention is a true solvate, while in other cases, the compound of the invention merely retains adventitious water or is a mixture of water plus some adventitious solvent.
  • Optional or “optionally” means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not.
  • optionally substituted aryl means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
  • a “pharmaceutical composition” refers to a formulation of a compound of the invention and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g. , humans.
  • a medium includes all pharmaceutically acceptable carriers, diluents or excipients therefor.
  • “Pharmaceutically acceptable carrier, diluent or excipient” includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
  • the compounds of the invention may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that are defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids.
  • Embodiments thus include all such possible isomers, as well as their racemic and optically pure forms.
  • Optically active (+) and (-), (R)- and (5)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization.
  • Embodiments of the present invention include all manner of rotamers and conformationally restricted states of a compound of the invention.
  • Atropisomers which are stereoisomers arising because of hindered rotation about a single bond, where energy differences due to steric strain or other contributors create a barrier to rotation that is high enough to allow for isolation of individual conformers, are also included.
  • certain compounds of the invention may exist as mixtures of atropisomers or purified or enriched for the presence of one atropisomer.
  • Non-limiting examples of compounds which exist as atropisomers include the following compounds:
  • the compound of structure (I) is a mixture of atropisomers. In other embodiments, the compound of structure (I) is a substantially purified atropisomer. In some embodiments, the compound of structure (I) is a substantially purified R-atropisomer. In some other embodiments, the compound of structure (I) is a substantially purified R-atropisomer.
  • stereoisomer refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable.
  • the present invention contemplates various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another.
  • a “tautomer” refers to a proton shift from one atom of a molecule to another atom of the same molecule. Embodiments thus include tautomers of the disclosed compounds.
  • the chemical naming protocol and structure diagrams used herein are a modified form of the I.U.P.A.C. nomenclature system, using the ACD/Name Version 9.07 software program and/or ChemDraw Ultra Version 11.0.1 software naming program (CambridgeSoft).
  • a substituent group is typically named before the group to which it attaches.
  • cyclopropylethyl comprises an ethyl backbone with a cyclopropyl substituent.
  • all bonds are identified in the chemical structure diagrams herein, except for all bonds on some carbon atoms, which are assumed to be bonded to sufficient hydrogen atoms to complete the valency.
  • the invention provides compounds which are capable of selectively binding to and/or modulating a G12C mutant KRAS, HRAS or RAS protein.
  • the compounds may modulate the G12C mutant KRAS, HRAS or NRAS protein by reaction with an amino acid. While not wishing to be bound by theory, the present applicants believe that, in some embodiments, the compounds of the invention selectively react with the G12C mutant KRAS, HRAS or NRAS proteins by forming a covalent bond with the cysteine at the 12 position of a G12C mutant KRAS, HRAS or NRAS protein. By binding to the Cysteine 12, the compounds of the invention may lock the switch II of the G12C mutant KRAS, HRAS or NRAS into an inactive stage.
  • This inactive stage may be distinct from those observed for GTP and GDP bound KRAS, HRAS or NRAS.
  • Some compounds of the invention may also be able to perturb the switch I conformation. Some compounds of the invention may favor the binding of the bound KRAS, HRAS or NRAS to GDP rather than GTP and therefore sequester the KRAS, HRAS or NRAS into an inactive KRAS, HRAS or NRAS GDP state. Because effector binding to KRAS, HRAS or NRAS is highly sensitive to the conformation of switch I and II, the irreversible binding of these compounds may disrupt KRAS, HRAS or NRAS downstream signaling.
  • compounds having activity as modulators of a G12C mutant KRAS, HRAS or NRAS protein are provided, the compounds have the followin structure (I):
  • A is N or C
  • B is oxo, cyano, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, heteroaryl, cycloalkylalkyl, heterocycloalkyl, heteroarylalkyl, amino, alkylamino, arylamino, -C0 2 H, -CONH 2 , aminylcarbonyl, aminylcarbonylalkyl, heteroarylamino, halo, haloalkyl, alkoxy, haloalkoxy, aryl or -X-L 2 -R a ;
  • X is -NR b - or -0-;
  • L 1 is alkylene, cycloalkylene, heterocyclylene or absent;
  • L 2 is alkylene or absent
  • R b is, at each occurrence, independently H or Ci-C 6 alkyl
  • R 1 is aryl or heteroaryl
  • alkylaminylalkyl cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aminylcarbonyl, heteroaryl or aryl;
  • is a single or double bond such that all valences are satisfied
  • E is an electrophilic moiety capable of forming a covalent bond with the cysteine residue at position 12 of a KRAS, HRAS or NRAS G12C mutant protein.
  • A is N or C
  • B is oxo, alkyl, cycloalkyl, heterocyclyl, heteroaryl, cycloalkylalkyl, heterocycloalkyl, heteroarylalkyl, amino, alkylamino, arylamino, -C0 2 H, -CONH 2 , aminylcarbonyl, aminylcarbonylalkyl, heteroarylamino, halo, haloalkyl, alkoxy, haloalkoxy, aryl or -X-L 2 -R a ;
  • X is -NR b - or -0-;
  • L 1 is alkylene, cycloalkylene, heterocyclylene or absent;
  • L 2 is alkylene or absent
  • R is H, cyano, amino, halo, haloalkyl, hydroxyl, cycloalkyl, heterocyclyl, heterocycloalkyl, aryl, heteroaryl, -C0 2 H, -CONH 2, aminylcarbonyl, Ci- C 6 alkyl, Ci-C 6 alkylaminyl or Ci-C 6 alkoxy;
  • R b is, at each occurrence, independently H or Ci-C 6 alkyl
  • R 1 is aryl or heteroaryl
  • alkylaminylalkyl cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aminylcarbonyl, heteroaryl or aryl;
  • is a single or double bond such that all valences are satisfied; and E is an electrophilic moiety capable of forming a covalent bond with the cysteine residue at position 12 of a KRAS, HRAS or NRAS G12C mutant protein.
  • A is N or C
  • B is oxo, alkyl, cycloalkyl, heterocyclyl, heteroaryl, cycloalkylalkyl, heterocycloalkyl, heteroarylalkyl or -X-L 2 -R a ;
  • X is -NR b - or -0-;
  • L 1 is alkylene, cycloalkylene, heterocyclylene or absent;
  • L 2 is alkylene or absent
  • R is H, cyano, amino, Ci-C 6 alkyl, Ci-C 6 alkylaminyl or Ci-C 6 alkoxy;
  • R b is H or Ci-C 6 alkyl
  • R 1 is aryl or heteroaryl
  • is a single or double bond such that all valences are satisfied
  • E is an electrophilic moiety capable of forming a covalent bond with the cysteine residue at position 12 of a KRAS, HRAS or NRAS G12C mutant protein.
  • B is cycloalkyl, heterocyclyl or heteroaryl.
  • L 1 is alkylene or absent.
  • B is cycloalkyl or heterocyclyl, for example in some embodiments the compound has the following structure (IA):
  • G 1 is N or CH
  • G 2 is R C or CHR C ;
  • R c is H, alkyl, alkylcarbonyl, aminocarbonyl, alkylcarbonylaminyl, aminocarbonylaminyl or heteroarylcarbonyl;
  • R a and R are, at each occurrence, independently
  • R 3a and R 3b join to form oxo a carbocyclic or heterocyclic ring; or R 3a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, Ci-C 6 haloalkyl, Ci-C 6 haloalkoxy, C 2 -C 6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R 3a and R 3b join to form oxo a carbocyclic or heterocyclic ring; or R 3a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, Ci-C 6 haloalkyl, Ci-C 6 haloalkoxy
  • R a and R are, at each occurrence, independently
  • R 4a and R 4b join to form oxo, a carbocyclic or heterocyclic ring; or R 4a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, Ci-C 6 haloalkyl, Ci-C 6 haloalkoxy, C 2 - C 6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R 4a and R 4b join to form oxo, a carbocyclic or heterocyclic ring; or R 4a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, Ci-C 6 haloalkyl, Ci-C 6 haloal
  • n 1 and m 2 are each independently 1, 2 or 3;
  • n is an integer from 0 to 5.
  • R a and R are, at each occurrence, independently
  • R 3a and R 3b join to form a carbocyclic or heterocyclic ring; or R 3a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, C 2 -C 6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R 3b joins with R 4b to form a carbocyclic or heterocyclic ring;
  • R a and R are, at each occurrence, independently
  • R 4a and R 4b join to form a carbocyclic or heterocyclic ring; or R 4a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, C 2 -C 6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R 4b joins with R 3b to form carbocyclic or heterocyclic ring;
  • the compound has the following structure (IAa) or (IAb):
  • p is an integer from 0 to 3.
  • the compound has one of the following structures (I Ac), (IAd) or (IAe):
  • R c is alkylcarbonyl, aminocarbonyl, alkylcarbonylaminyl, aminocarbonylaminyl or heteroarylcarbonyl, for example, in some embodiments the alkylcarbonyl is substituted with aminocarbonyl, hydroxylaminocarbonyl, hydroxyl or amino.
  • R c has one of the following structures:
  • p 2 is an integer from 1 to 3.
  • R c has one of the following structures:
  • B is alkyl, cycloalkylalkyl, heterocycloalkyl, heteroarylalkyl or -X-L 2 -R a .
  • B is -X-L 2 -R a .
  • R a is heterocyclyl or heteroaryl.
  • L 1 is alkylene or absent. In some embodiments, L 1 is alkylene. In other embodiments, L 1 is absent.
  • B is -X-L 2 -R a and L 1 is alkylene or absent, for example in some embodiments the compound has one of the following structures (IB) or (IC):
  • H represents a 5 or 6-membered heteroaryl ring optionally substituted with one or more of R 3a , R 3b , R 4a and R 4b ;
  • G 1 is N or CH
  • G 2 is R C or CHR C ;
  • R c is H, alkyl, alkylcarbonyl, aminocarbonyl, alkylcarbonylaminyl, aminocarbonylaminyl or heteroaryl carbonyl;
  • R 3a and R 3b are, at each occurrence, independently
  • R 3a and R 3b join to form oxo a carbocyclic or heterocyclic ring; or R 3a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, Ci-C 6 haloalkyl, Ci-C 6 haloalkoxy, C 2 -C 6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R 3a and R 3b join to form oxo a carbocyclic or heterocyclic ring; or R 3a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, Ci-C 6 haloalkyl, Ci-C 6 haloalkoxy
  • R 4a and R 4b are, at each occurrence, independently
  • R 4a and R 4b join to form oxo a carbocyclic or heterocyclic ring; or R 4a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, Ci-C 6 haloalkyl, Ci-C 6 haloalkoxy, C 2 -C 6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R 4a and R 4b join to form oxo a carbocyclic or heterocyclic ring; or R 4a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, Ci-C 6 haloalkyl, Ci-C 6 haloalkoxy
  • n 1 and m 2 are each independently 1, 2 or 3; and n is an integer from 0 to 5.
  • R 3a and R 3b are, at each occurrence, independently
  • R 3a and R 3b join to form a carbocyclic or heterocyclic ring; or R 3a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, C 2 -C 6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R 3b joins with R 4b to form a carbocyclic or heterocyclic ring;
  • R a and R are, at each occurrence, independently
  • R 4a and R 4b join to form a carbocyclic or heterocyclic ring; or R 4a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, C 2 -C 6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R 4b joins with R 3b to form a carbocyclic or heterocyclic ring;
  • H is pyrrolidinyl or pyridinyl.
  • the compound has one of the following structures
  • R d is, at each occurrence, independently H, halo or hydroxyl
  • p 3 is an integer from 0 to 3.
  • the compound has one of the following structures (IBc
  • X is - H-. In some of other the foregoing embodiments, X is -0-.
  • B is -X-L 2 -R a , and the compound has the following structure (ID):
  • the compound has the following structure
  • X is - H-. In some other embodiments of compounds (ID) or (IDa) X is -0-.
  • B is heteroarylalkyl.
  • the heteroarylalkyl is pyrrolidinylalkyl or pyridinylalkyl.
  • B is oxo.
  • L 1 is heterocyclylene.
  • B is oxo, and L 1 is heterocyclylene.
  • the compound has the following structure (IE):
  • G 1 is CH
  • G 2 is N or CH
  • R 3a and R 3b are, at each occurrence, independently
  • R 3a and R 3b join to form oxo a carbocyclic or heterocyclic ring; or R 3a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, Ci-C 6 haloalkyl, Ci-C 6 haloalkoxy, C 2 -C 6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R 3a and R 3b join to form oxo a carbocyclic or heterocyclic ring; or R 3a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, Ci-C 6 haloalkyl, Ci-C 6 haloalkoxy
  • R 4a and R 4b are, at each occurrence, independently
  • R 4a and R 4b join to form oxo a carbocyclic or heterocyclic ring; or R 4a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, Ci-C 6 haloalkyl, Ci-C 6 haloalkoxy, C 2 -C 6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R 4a and R 4b join to form oxo a carbocyclic or heterocyclic ring; or R 4a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, Ci-C 6 haloalkyl, Ci-C 6 haloalkoxy
  • R 3a and R 3b are, at each occurrence, independently
  • R 3a and R 3b join to form a carbocyclic or heterocyclic ring; or R 3a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, C 2 -C 6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R 3b joins with R 4b to form carbocyclic or heterocyclic ring; R 4a and R 4b are, at each occurrence, independently
  • R 4a and R 4b join to form a carbocyclic or heterocyclic ring; or R 4a is H, -OH, - H 2 , -C0 2 H, halo, cyano, Ci-C 6 alkyl, C 2 -C 6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R 4b joins with R 3b to form a carbocyclic or heterocyclic ring;
  • the compound has one of the following structures (IEa), (IEb), (IEc) or (IEd) :
  • the compound has one of the following structures IEe), (IEf), (IEg), (IEh (IEi) of (IEj):
  • the compound has the following structure (IEI):
  • R 1 is aryl or heterocyclyl (e.g., heteroaryl or aliphatic heterocyclyl), each of which is optionally substituted with one or more substituents.
  • R 1 is aryl or heteroaryl.
  • R 1 is capable of reversible interaction with KRAS, HRAS or NRAS G12C mutant protein.
  • R 1 has high affinity towards KRAS, HRAS or NRAS and is highly specific towards G12C KRAS, HRAS or NRAS.
  • R 1 is capable of hydrophobic interaction with KRAS, HRAS or NRAS G12C.
  • R 1 is able to form hydrogen bonds with various residues of G12C KRAS, HRAS or NRAS protein.
  • R 1 is aryl.
  • R 1 is phenyl, and in other embodiments R 1 is naphthyl.
  • R 1 is substituted or unsubstituted.
  • R 1 is substituted with one or more substituents.
  • R 1 is substituted with halo, amino, hydroxyl, Ci-C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, cyano, Ci-C 6 haloalkyl, Ci-C 6 alkoxy, alkylaminyl, cycloalkyl, heterocyclylalkyl, heterocyclylalkoxy, heterocyclylaminyl,
  • R 1 is substituted with halo, hydroxyl, Ci-C 6 alkyl, Ci-C 6 haloalkyl, Ci-C 6 alkoxy or Ci-C 6 alkylcarbonyloxy, or combinations thereof.
  • R 1 is substituted with fluoro, chloro, cyclopropyl, cyclobutyl, hydroxyl, amino, methyl, ethyl, isopropyl, trifluoromethyl or methoxy, or combinations thereof.
  • R 1 is substituted with fluoro, hydroxyl, methyl, isopropyl, trifluoromethyl or methoxy, or combinations thereof.
  • R 1 has one of the following structures:
  • R 1 is heteroaryl, for example a heteroaryl comprising nitrogen.
  • R 1 is indazolyl or quinolinyl.
  • R 1 is heteroaryl which is substituted with one or more substituents.
  • R 1 is substituted with hydroxyl or Ci-C 6 alkyl, or both.
  • R 1 has one of the following structures: , for example:
  • R c is H.
  • R 2a and R 2b are each halo.
  • R 2a is fiuoro, and in other embodiments, R 2b is chloro.
  • R 2a and R 2b are each independently halo, haloalkyl, alkyl, amino, hydroxyl or alkoxy. In other embodiments, R 2a and R 2b are each independently halo, haloalkyl, alkyl or alkoxy. In some embodiments, R 2a is fluoro, chloro, CF 3 or methoxy. In some embodiments, R 2a is fluoro, chloro or methoxy. In different embodiments, R 2b is chloro, fluoro, amino, hydroxyl or CF 3 . In still other different embodiments, R 2b is chloro, fluoro or CF 3 .
  • the compounds have the following structure (IF):
  • R is H.
  • is a double bond such that all valences are satisfied.
  • A is N. In other embodiments, A is C.
  • nucleophile such as the cysteine residue at position 12 of a KRAS, HRAS or RAS
  • E moieties which are capable of reaction with (e.g., by covalent bond formation) a nucleophile are preferred.
  • E is capable of reacting in a conjugate addition manner (e.g., 1,4-conjugate addition) with an appropriately reactive nucleophile.
  • E comprises conjugated pi bonds such that derealization of electrons results in at least one atom (e.g., a carbon atom) having a positive charge, partial positive charge or a polarized bond.
  • E comprises one or more bonds wherein the electronegativity of the two atoms forming the bonds is sufficiently different such that a partial positive charge (e.g., by polarization of the bond) resides on one of the atoms, for example on a carbon atom.
  • E moieties comprising carbon-halogen bonds, carbon-oxygen bonds or carbon bonds to various leaving groups known in the art are examples of such E moieties.
  • E has the following structure:
  • R 8 is H, -OH, -CN or Ci-C 6 alkyl
  • R 9 and R 10 are each independently H, halo, cyano, carboxyl, Ci-C 6 alkyl, alkoxycarbonyl, aminylalkyl, alkylaminylalkyl, aryl, heterocyclyl, heterocyclylalkyl, heteroaryl or hydroxylalkyl, or R 9 and R 10 join to form a carbocyclic, heterocyclic or heteroaryl ring.
  • E has the following structure:
  • R 8 is H, Ci-C 6 alkyl or hydroxylalkyl
  • R 10 is H, Ci-C 6 alkyl, aminylalkyl, alkylaminylalkyl or hydroxylalkyl.
  • the Q moiety is typically selected to optimize the reactivity (i.e., electrophilicity) of E.
  • R 8 is H.
  • R 8 is -CN.
  • R 8 is -OH.
  • R 8 is H. In other of these embodiments, R 8 is hydroxylalkyl, for example in some embodiments the hydroxylalkyl is 2-hydroxylalkyl.
  • At least one of R 9 or R 10 is H.
  • each of R 9 and R 10 are H.
  • R 10 is alkylaminylalkyl. In some of these embodiments, R 10 has the following structure:
  • R 10 is hydroxylalkyl, such as 2-hydroxylalkyl.
  • R 9 and R 10 join to form a carbocyclic ring.
  • the carbocyclic ring is a cyclopentene, cyclohexene or phenyl ring.
  • the carbocyclic ring is a cyclopentene or cyclohexene ring.
  • the carbocyclic ring is a phenyl ring, for example a phenyl ring having the following structure:
  • E is an electrophile capable of bonding with a KRAS, HRAS or NRAS protein comprising G12C mutation.
  • the electrophile E is capable of forming an irreversible covalent bond with a G12C mutant KRAS, HRAS or NRAS protein. In some cases, the electrophile E may bind with the cysteine residue at the position 12 of a G12C mutant KRAS, HRAS or NRAS protein. In various embodiments of any of the foregoing, E has one of the followin structures:
  • E has one of the following structures:
  • E has one of the following structures:
  • E has one of the following structures:
  • R is H or Ci-Cealkyl
  • R 9 is H, cyano or Ci-C 6 alkyl, or R 9 joins with R 10 to form a carbocycle;
  • R 10 is H or Ci-C 6 alkyl or R 10 joins with R 9 to form a carbocycle and
  • R 10a is H or Ci-C 6 alkyl.
  • E is
  • R a and R are, at each occurrence, independently H, -OH, - H 2 , -C0 2 H, halo, cyano, hydroxylalkly, aminylalkyl, cyanoalkyl, carboxyalkyl or aminylcarbonyl
  • R 4a and R 4b are, at each occurrence, independently H, -OH, - H 2 , -C0 2 H, halo, cyano, hydroxylalkly, aminylalkyl, cyanoalkyl, carboxyalkyl or aminylcarbonyl.
  • R 3a and R 4a are, at each occurrence, independently H, -OH, hydroxylalkly, cyano, or aminylcarbonyl and R 3b and R 4b are H.
  • R a and R a are H and R and R are, at each occurrence, independently H, -OH, - H 2 , -C0 2 H, halo, cyano, hydroxylalkly, aminylalkyl, cyanoalkyl, carboxyalkyl or aminylcarbonyl.
  • At least one of R 3a , R 3b , R 4a or R 4b is H. In some embodiments, each of R 3a , R 3b , R 4a and R 4b are H.
  • R 3a and R 4a are, at each occurrence, independently H or Ci-C 6 alkyl, provided at least one of R 3a or R 4a is Ci-C 6 alkyl.
  • at least one of R 3a , R 4a , R 3b and R 4b is independently Ci- C 6 alkyl, such as methyl.
  • one occurrence of R 3a is Ci-C 6 alkyl, such as methyl, and the remaining R 3a and each R 4a is H.
  • two occurrences of R 3a are Ci-C 6 alkyl, such as methyl, and the remaining R 3a and each R 4a is H.
  • one occurrence of R 3a and one occurrence of R 4a is independently Ci-C 6 alkyl, such as methyl, and the remaining R 3a and R 4a are each H.
  • R 3a is -OH, - H 2 , -C0 2 H, halo, cyano, hydroxylalkly, aminylalkyl, cyanoalkyl, carboxyalkyl or aminylcarbonyl, and R 3b , R 4a and R 4b are H.
  • R 4a is -OH, - H 2 , -C0 2 H, halo, cyano, hydroxylalkly, aminylalkyl, cyanoalkyl, carboxyalkyl or aminylcarbonyl, and R 3a , R 3b and R 4b are H.
  • R 3a is H, -OH, - H 2 , -C0 2 H, halo, cyano, hydroxylalkly, aminylalkyl, cyanoalkyl, carboxyalkyl or aminylcarbonyl, and R 3b joins with R 4b to form a carbocyclic or heterocyclic ring;
  • R 4a is H, -OH, - H 2 , -C0 2 H, halo, cyano, hydroxylalkly, aminylalkyl, cyanoalkyl, carboxyalkyl or aminylcarbonyl, and R 4b joins with R 3b to form a carbocyclic or heterocyclic ring.
  • R 3a and R 3b join to form a carbocyclic or heterocyclic ring.
  • R 4a and R 4b join to form a carbocyclic or heterocyclic ring.
  • R a or R a is aminylcarbonyl.
  • the aminylcarbonyl is .
  • R 4a is cyano. In other embodiments, R 3a or R 4a is -OH. In other embodiments, R 3a or R 4a is hydroxylalkyl, for example hydroxylmethyl.
  • R 1 is aryl or heteroaryl and R 2a , R 2b and R 2c are independently selected from H and halo, for example in some further embodiments R 1 is aryl or heteroaryl and R 2a and R 2b are independently selected from halo, such as chloro and fluoro, and R 2c is H.
  • R 1 is aryl or heteroaryl
  • R 2a is chloro
  • R 2b is fluoro
  • R 2c is H.
  • R 1 is aryl or heteroaryl
  • one of R 2a or R 2b is halo, such as chloro or fluoro, and the other one of R 2a or R 2b is H.
  • 1, and m 2 is 1. In other embodiments, m 1 is 1 and, m 2 is 2. In still other embodiments and m 2 is 2. In more embodiments, 1, and m 2 is 3.
  • stereoisomer Other embodiments are directed to a single stereoisomer.
  • the compounds are racemic (e.g., mixture of atropisomers), while in other embodiments the compounds are substantially a single isomer, for example a substantially purified atropisomer.
  • the compound has one of the structures set forth in Table 1 below.
  • Table 1 The compounds in Table 1 were each prepared and analyzed by mass spectrometry and/or 1 H MR. Experimental mass spectrometry data is included in Table 1. Exemplary synthetic procedures are described in more detail below and in the Examples. General methods by which the compounds may be prepared are provided below and indicated in Table 1. Table 1
  • starting components may be obtained from sources such as Sigma Aldrich, Lancaster Synthesis, Inc., Maybridge, Matrix Scientific, TCI, and Fluorochem USA, etc. or synthesized according to sources known to those skilled in the art (see, for example, Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5th edition (Wiley, December 2000)) or prepared as described in this invention.
  • Embodiments of the compound of structure (I) can be prepared according to General Reaction Scheme 1 ("Method A"), wherein R 1 , R 2a , R 2b , R 2c , R 3a , R 3b , R 4a , R 4b , R 9 , R 10 , Q, m 1 and m 2 are as defined herein.
  • Method A General Reaction Scheme 1
  • compounds of structure A-1 are purchased from commercial sources or prepared according to techniques known in the art.
  • the desired R 1 substituent added to A-1 by way of Suzuki coupling to yield A-2.
  • Cyclization of A- 2 with a reagent such as formamidine acetate provides quinazolinone A-3.
  • Treatment of A-3 with an appropriately substituted cyclic group and an activating agent, such as HATU provides A-4 which can then be deprotected by treatment with acid.
  • the "E” moiety is then installed under conditions known in the art to yield A-6.
  • Embodiments of the compound of structure (I) can be prepared according to General Reaction Scheme 2 ("Method B"), wherein R 1 , R 2a , R 2b , R 2c , R 3a , R 3b , R 4a , R 4b , R 9 , R 10 , m 1 and m 2 are as defined herein.
  • Method B General Reaction Scheme 2
  • compounds of structure A-3 are prepared as described in General Scheme 1.
  • Treatment of A-3 with an appropriately substituted cyclic group and an activating agent, such as HATU, provides B-1.
  • B-1 is deprotected and the "E" moiety installed to yield B-3 in a manner analogous to that described above with regard to General Reaction Scheme 1.
  • Embodiments of the compound of structure (I) can be prepared according to General Reaction Scheme 3 ("Method C"), wherein R 1 , R 2a , R 2b , R 2c , R 3a , R 3b , R 4a , R 4b , R 9 , R 10 , m 1 and m 2 are as defined herein.
  • Method C General Reaction Scheme 3
  • compounds of structure C-1 are purchased from commercial sources or prepared according to methods known in the art.
  • C-1 is reacted with diethyl 2-(ethoxymethylene)malonate to yield C-2.
  • C-2 can then be cyclized by heating in an appropriate high-boiling solvent (e.g., Ph 2 0) to yield quinolone C-3. Chlorination of C-3 yields C-4 which is reacted with an appropriate heterocyclic moiety to yield C-5.
  • the desired R 1 moiety is installed using Suzuki chemistry as described above.
  • the protecting group of C-6 is removed, and the free amine is optionally functionalized to yield C-7. Saponification of C-7 followed by decarboxylation and amination yields C-8.
  • C-8 is then treated in a manner analogous to that described for Method A to yield C-9.
  • Suitable protecting groups include, but are not limited to, hydroxy, amino, mercapto and carboxylic acid.
  • Suitable protecting groups for hydroxy include trialkylsilyl or diarylalkylsilyl (for example, t-butyldimethylsilyl, t-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like.
  • Suitable protecting groups for amino, amidino and guanidino include t-butoxycarbonyl, benzyloxycarbonyl, and the like.
  • Suitable protecting groups for mercapto include -C(0)-R" (where R" is alkyl, aryl or arylalkyl), /?-methoxybenzyl, trityl and the like.
  • Suitable protecting groups for carboxylic acid include alkyl, aryl or arylalkyl esters.
  • Protecting groups are optionally added or removed in accordance with standard techniques, which are known to one skilled in the art and as described herein. The use of protecting groups is described in detail in Green, T.W. and P.G.M. Wutz, Protective Groups in Organic Synthesis (1999), 3rd Ed., Wiley.
  • the protecting group may also be a polymer resin such as a Wang resin, Rink resin or a 2-chlorotrityl-chloride resin.
  • compositions comprising any one (or more) of the foregoing compounds and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition is formulated for oral administration.
  • the pharmaceutical composition is formulated for injection.
  • the pharmaceutical compositions comprise a compound as disclosed herein and an additional therapeutic agent (e.g., anticancer agent).
  • additional therapeutic agent e.g., anticancer agent
  • Suitable routes of administration include, but are not limited to, oral, intravenous, rectal, aerosol, parenteral, ophthalmic, pulmonary, transmucosal, transdermal, vaginal, otic, nasal, and topical administration.
  • parenteral delivery includes intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intralymphatic, and intranasal injections.
  • a compound as described herein is administered in a local rather than systemic manner, for example, via injection of the compound directly into an organ, often in a depot preparation or sustained release formulation.
  • long acting formulations are administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the drug is delivered in a targeted drug delivery system, for example, in a liposome coated with organ-specific antibody.
  • the liposomes are targeted to and taken up selectively by the organ.
  • the compound as described herein is provided in the form of a rapid release formulation, in the form of an extended release formulation, or in the form of an intermediate release formulation.
  • the compound described herein is administered topically.
  • the compounds according to the invention are effective over a wide dosage range.
  • dosages from 0.01 to 1000 mg, from 0.5 to 100 mg, from 1 to 50 mg per day, and from 5 to 40 mg per day are examples of dosages that are used in some embodiments.
  • An exemplary dosage is 10 to 30 mg per day. The exact dosage will depend upon the route of administration, the form in which the compound is administered, the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician.
  • a compound of the invention is administered in a single dose.
  • administration will be by injection, e.g., intravenous injection, in order to introduce the agent quickly.
  • injection e.g., intravenous injection
  • other routes are used as appropriate.
  • a single dose of a compound of the invention may also be used for treatment of an acute condition.
  • a compound of the invention is administered in multiple doses. In some embodiments, dosing is about once, twice, three times, four times, five times, six times, or more than six times per day. In other embodiments, dosing is about once a month, once every two weeks, once a week, or once every other day. In another embodiment a compound of the invention and another agent are administered together about once per day to about 6 times per day. In another embodiment the administration of a compound of the invention and an agent continues for less than about 7 days. In yet another embodiment the administration continues for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, continuous dosing is achieved and maintained as long as necessary.
  • a compound of the invention is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, or 28 days. In some embodiments, a compound of the invention is administered for less than 28, 14, 7, 6, 5, 4, 3, 2, or 1 day. In some embodiments, a compound of the invention is administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
  • the compounds of the invention are administered in dosages. It is known in the art that due to intersubject variability in compound pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy. Dosing for a compound of the invention may be found by routine experimentation in light of the instant disclosure.
  • the compounds described herein are formulated into pharmaceutical compositions.
  • pharmaceutical compositions are formulated in a conventional manner using one or more
  • physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any pharmaceutically acceptable techniques, carriers, and excipients are used as suitable to formulate the pharmaceutical compositions described herein: Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa. : Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975; Liberman, H.A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and
  • compositions comprising a compound of structure (I) and a pharmaceutically acceptable diluent(s), excipient(s), or carrier(s).
  • the compounds described are administered as pharmaceutical compositions in which compounds of structure (I) are mixed with other active ingredients, as in combination therapy.
  • the pharmaceutical compositions include one or more compounds of structure (I).
  • a pharmaceutical composition refers to a mixture of a compound of structure (I) with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients.
  • the pharmaceutical composition facilitates administration of the compound to an organism.
  • therapeutically effective amounts of compounds of structure (I) provided herein are administered in a pharmaceutical composition to a mammal having a disease, disorder or medical condition to be treated.
  • the mammal is a human.
  • therapeutically effective amounts vary depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors.
  • the compounds described herein are used singly or in combination with one or more therapeutic agents as components of mixtures.
  • one or more compounds of structure (I) is formulated in an aqueous solutions.
  • the aqueous solution is selected from, by way of example only, a physiologically compatible buffer, such as Hank's solution, Ringer's solution, or physiological saline buffer.
  • a physiologically compatible buffer such as Hank's solution, Ringer's solution, or physiological saline buffer.
  • one or more compound of structure (I) is/are formulated for transmucosal administration.
  • transmucosal formulations include penetrants that are appropriate to the barrier to be permeated.
  • appropriate formulations include aqueous or nonaqueous solutions.
  • such solutions include physiologically compatible buffers and/or excipients.
  • compounds described herein are formulated for oral administration. Compounds described herein are formulated by combining the active compounds with, e.g., pharmaceutically acceptable carriers or excipients.
  • the compounds described herein are formulated in oral dosage forms that include, by way of example only, tablets, powders, pills, dragees, capsules, liquids, gels, syrups, elixirs, slurries, suspensions and the like.
  • pharmaceutical preparations for oral use are obtained by mixing one or more solid excipient with one or more of the compounds described herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as: for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methylcellulose,
  • microcrystalline cellulose hydroxypropylmethylcellulose, sodium
  • disintegrating agents are optionally added.
  • Disintegrating agents include, by way of example only, cross-linked
  • croscarmellose sodium polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • dosage forms such as dragee cores and tablets, are provided with one or more suitable coating.
  • suitable coating In specific embodiments, concentrated sugar solutions are used for coating the dosage form.
  • the sugar solutions optionally contain additional components, such as by way of example only, gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs and/or pigments are also optionally added to the coatings for identification purposes.
  • dyestuffs and/or pigments are optionally utilized to characterize different combinations of active compound doses.
  • Oral dosage forms include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • push-fit capsules contain the active ingredients in admixture with one or more filler.
  • Fillers include, by way of example only, lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • soft capsules contain one or more active compound that is dissolved or suspended in a suitable liquid. Suitable liquids include, by way of example only, one or more fatty oil, liquid paraffin, or liquid polyethylene glycol.
  • stabilizers are optionally added.
  • therapeutically effective amounts of at least one of the compounds described herein are formulated for buccal or sublingual
  • Formulations suitable for buccal or sublingual administration include, by way of example only, tablets, lozenges, or gels.
  • the compounds described herein are formulated for parental injection, including
  • formulations for injection are presented in unit dosage form (e.g., in ampoules) or in multi-dose containers. Preservatives are, optionally, added to the injection formulations.
  • the pharmaceutical compositions are formulated in a form suitable for parenteral injection as sterile suspensions, solutions or emulsions in oily or aqueous vehicles.
  • Parenteral injection formulations optionally contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form.
  • suspensions of the active compounds e.g., compounds of structure (I) are prepared as appropriate oily injection suspensions.
  • Suitable lipophilic solvents or vehicles for use in the pharmaceutical compositions described herein include, by way of example only, fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • aqueous injection suspensions contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension contains suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the active ingredient is in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • the compounds of structure (I) are administered topically.
  • the compounds described herein are formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, pastes, medicated sticks, balms, creams or ointments.
  • Such pharmaceutical such as solutions, suspensions, lotions, gels, pastes, medicated sticks, balms, creams or ointments.
  • compositions optionally contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.
  • the compounds of structure (I) are formulated for transdermal administration.
  • transdermal formulations employ transdermal delivery devices and transdermal delivery patches and can be lipophilic emulsions or buffered, aqueous solutions, dissolved and/or dispersed in a polymer or an adhesive.
  • patches are constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • the transdermal delivery of the compounds of structure (I) is
  • transdermal patches provide controlled delivery of the compounds of structure (I).
  • the rate of absorption is slowed by using rate-controlling membranes or by trapping the compound within a polymer matrix or gel.
  • absorption enhancers are used to increase absorption.
  • Absorption enhancers or carriers include absorbable pharmaceutically acceptable solvents that assist passage through the skin.
  • transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
  • the compounds of structure (I) are formulated for administration by inhalation.
  • Various forms suitable for administration by inhalation include, but are not limited to, aerosols, mists or powders.
  • Pharmaceutical compositions of any of compound of structure (I) are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant (e.g., dichlorodifluoromethane, trichlorofluoromethane,
  • the dosage unit of a pressurized aerosol is determined by providing a valve to deliver a metered amount.
  • capsules and cartridges of, such as, by way of example only, gelatin for use in an inhaler or insufflator are formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the compounds of structure (I) are formulated in rectal compositions such as enemas, rectal gels, rectal foams, rectal aerosols, suppositories, jelly suppositories, or retention enemas, containing conventional suppository bases such as cocoa butter or other glycerides, as well as synthetic polymers such as polyvinylpyrrolidone, PEG, and the like.
  • a low-melting wax such as, but not limited to, a mixture of fatty acid glycerides, optionally in combination with cocoa butter is first melted.
  • compositions are formulated in any conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any pharmaceutically acceptable techniques, carriers, and excipients are optionally used as suitable. Pharmaceutical compositions comprising a compound of structure (I) are manufactured in a
  • compositions include at least one pharmaceutically acceptable carrier, diluent or excipient and at least one compound of structure (I), described herein as an active ingredient.
  • the active ingredient is in free-acid or free- base form, or in a pharmaceutically acceptable salt form.
  • the methods and pharmaceutical compositions described herein include the use of N-oxides, crystalline forms (also known as polymorphs), as well as active metabolites of these compounds having the same type of activity. All tautomers of the compounds described herein are included within the scope of the compounds presented herein.
  • the compounds described herein encompass unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. The solvated forms of the compounds presented herein are also considered to be disclosed herein.
  • compositions optionally include other medicinal or pharmaceutical agents, carriers, adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure, buffers, and/or other therapeutically valuable substances.
  • adjuvants such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure, buffers, and/or other therapeutically valuable substances.
  • compositions comprising the compounds described herein include formulating the compounds with one or more inert, pharmaceutically acceptable excipients or carriers to form a solid, semi-solid or liquid.
  • Solid compositions include, but are not limited to, powders, tablets, dispersible granules, capsules, cachets, and suppositories.
  • Liquid compositions include solutions in which a compound is dissolved, emulsions comprising a compound, or a solution containing liposomes, micelles, or nanoparticles comprising a compound as disclosed herein.
  • Semi-solid compositions include, but are not limited to, gels, suspensions and creams.
  • compositions described herein include liquid solutions or suspensions, solid forms suitable for solution or suspension in a liquid prior to use, or as emulsions. These compositions also optionally contain minor amounts of nontoxic, auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, and so forth.
  • composition comprising at least one compound of structure (I) illustratively takes the form of a liquid where the agents are present in solution, in suspension or both. Typically when the composition is administered as a solution or suspension a first portion of the agent is present in solution and a second portion of the agent is present in particulate form, in suspension in a liquid matrix.
  • a liquid composition includes a gel formulation. In other embodiments, the liquid composition is aqueous.
  • useful aqueous suspensions contain one or more polymers as suspending agents.
  • Useful polymers include water-soluble polymers such as cellulosic polymers, e.g., hydroxypropyl methylcellulose, and water-insoluble polymers such as cross-linked carboxyl-containing polymers.
  • Certain pharmaceutical compositions described herein comprise a mucoadhesive polymer, selected for example from carboxymethylcellulose, carbomer (acrylic acid polymer),
  • poly(methylmethacrylate), polyacrylamide, polycarbophil, acrylic acid/butyl acrylate copolymer, sodium alginate and dextran are examples of poly(methylmethacrylate), polyacrylamide, polycarbophil, acrylic acid/butyl acrylate copolymer, sodium alginate and dextran.
  • Useful pharmaceutical compositions also, optionally, include solubilizing agents to aid in the solubility of a compound of structure (I).
  • solubilizing agent generally includes agents that result in formation of a micellar solution or a true solution of the agent.
  • Certain acceptable nonionic surfactants for example polysorbate 80, are useful as solubilizing agents, as can ophthalmically acceptable glycols, polyglycols, e.g., polyethylene glycol 400, and glycol ethers.
  • useful pharmaceutical compositions optionally include one or more pH adjusting agents or buffering agents, including acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane; and buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride.
  • acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids
  • bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane
  • buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride.
  • acids, bases and buffers are included in an amount required to maintain pH of the composition in an acceptable range.
  • useful compositions also, optionally, include one or more salts in an amount required to bring osmolality of the composition into an acceptable range.
  • salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions; suitable salts include sodium chloride, potassium chloride, sodium thiosulfate, sodium bisulfite and ammonium sulfate.
  • Other useful pharmaceutical compositions optionally include one or more preservatives to inhibit microbial activity.
  • Suitable preservatives include mercury-containing substances such as merfen and thiomersal; stabilized chlorine dioxide; and quaternary ammonium compounds such as benzalkonium chloride, cetyltrimethylammonium bromide and cetylpyridinium chloride.
  • compositions include one or more surfactants to enhance physical stability or for other purposes.
  • Suitable nonionic surfactants include polyoxyethylene fatty acid glycerides and vegetable oils, e.g., polyoxyethylene (60) hydrogenated castor oil; and polyoxyethylene alkylethers and alkylphenyl ethers, e.g., octoxynol 10, octoxynol 40.
  • compositions include one or more antioxidants to enhance chemical stability where required.
  • Suitable antioxidants include, by way of example only, ascorbic acid and sodium metabisulfite.
  • aqueous suspension compositions are packaged in single-dose non-reclosable containers.
  • multiple-dose reclosable containers are used, in which case it is typical to include a preservative in the composition.
  • hydrophobic pharmaceutical compounds are employed. Liposomes and emulsions are examples of delivery vehicles or carriers useful herein. In certain embodiments, organic solvents such as N-methylpyrrolidone are also employed. In additional embodiments, the compounds described herein are delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials are useful herein. In some embodiments, sustained-release capsules release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization are employed.
  • the formulations described herein comprise one or more antioxidants, metal chelating agents, thiol containing compounds and/or other general stabilizing agents.
  • stabilizing agents include, but are not limited to: (a) about 0.5% to about 2% w/v glycerol, (b) about 0.1% to about 1% w/v methionine, (c) about 0.1% to about 2% w/v monothioglycerol, (d) about 1 mM to about 10 mM EDTA, (e) about 0.01% to about 2% w/v ascorbic acid, (f) 0.003% to about 0.02% w/v polysorbate 80, (g) 0.001% to about 0.05% w/v.
  • polysorbate 20 (h) arginine, (i) heparin, (j) dextran sulfate, (k) cyclodextrins, (1) pentosan poly sulfate and other heparinoids, (m) divalent cations such as magnesium and zinc; or (n)
  • the concentration of one or more compounds provided in the pharmaceutical compositions of the present invention is less than 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 1 1%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002%, or 0.0001% w/w, w/v or v/v.
  • the concentration of one or more compounds of the invention is greater than 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19.75%, 19.50%, 19.25% 19%, 18.75%, 18.50%, 18.25% 18%, 17.75%, 17.50%, 17.25% 17%, 16.75%, 16.50%, 16.25% 16%, 15.75%, 15.50%, 15.25% 15%, 14.75%, 14.50%, 14.25% 14%, 13.75%, 13.50%, 13.25% 13%, 12.75%, 12.50%, 12.25% 12%, 1 1.75%, 1 1.50%, 1 1.25% 1 1%, 10.75%, 10.50%, 10.25% 10%, 9.75%, 9.50%, 9.25% 9%, 8.75%, 8.50%, 8.25% 8%, 7.75%, 7.50%, 7.25% 7%, 6.75%, 6.50%, 6.25% 6%, 5.75%, 5.50%, 5.25% 5%, 4.75%, 4.50%,
  • the concentration of one or more compounds of the invention is in the range from approximately 0.0001%) to approximately 50%, approximately 0.001 ) to approximately 40 %, approximately 0.01% to approximately 30%, approximately 0.02% to approximately 29%, approximately 0.03%> to
  • the concentration of one or more compounds of the invention is in the range from approximately 0.001%) to approximately 10%, approximately 0.01% to approximately 5%, approximately 0.02% to approximately 4.5%o, approximately 0.03% to approximately 4%, approximately 0.04% to
  • the amount of one or more compounds of the invention is equal to or less than 10 g, 9.5 g, 9.0 g, 8.5 g, 8.0 g, 7.5 g, 7.0 g, 6.5 g, 6.0 g, 5.5 g, 5.0 g, 4.5 g, 4.0 g, 3.5 g, 3.0 g, 2.5 g, 2.0 g, 1.5 g, 1.0 g, 0.95 g, 0.9 g, 0.85 g, 0.8 g, 0.75 g, 0.7 g, 0.65 g, 0.6 g, 0.55 g, 0.5 g, 0.45 g, 0.4 g, 0.35 g, 0.3 g, 0.25 g, 0.2 g, 0.15 g, 0.1 g, 0.09 g, 0.08 g, 0.07 g, 0.06 g, 0.05 g, 0.04 g, 0.03 g, 0.02 g, 0.01 g, 0.009 g, 0.00
  • the amount of one or more compounds of the invention is more than 0.0001 g, 0.0002 g, 0.0003 g, 0.0004 g, 0.0005 g, 0.0006 g, 0.0007 g, 0.0008 g, 0.0009 g, 0.001 g, 0.0015 g, 0.002 g, 0.0025 g, 0.003 g, 0.0035 g, 0.004 g, 0.0045 g, 0.005 g, 0.0055 g, 0.006 g, 0.0065 g, 0.007 g, 0.0075 g, 0.008 g, 0.0085 g, 0.009 g, 0.0095 g, 0.01 g, 0.015 g, 0.02 g, 0.025 g, 0.03 g, 0.035 g, 0.04 g, 0.045 g, 0.05 g, 0.055 g, 0.06 g, 0.065 g, 0.07 g, 0.075
  • the amount of one or more compounds of the invention is in the range of 0.0001-10 g, 0.0005-9 g, 0.001-8 g, 0.005-7 g, 0.01-6 g, 0.05-5 g, 0.1-4 g, 0.5-4 g, or 1-3 g.
  • kits and articles of manufacture are also provided.
  • such kits comprise a carrier, package, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in a method described herein.
  • Suitable containers include, for example, bottles, vials, syringes, and test tubes.
  • the containers are formed from a variety of materials such as glass or plastic.
  • Packaging materials for use in packaging pharmaceutical products include those found in, e.g., U.S. Pat. Nos. 5,323,907, 5,052,558 and 5,033,252. Examples of
  • pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of
  • the container(s) includes one or more compounds described herein, optionally in a composition or in combination with another agent as disclosed herein.
  • the container(s) optionally have a sterile access port (for example the container is an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • kits optionally comprising a compound with an identifying description or label or instructions relating to its use in the methods described herein.
  • a kit typically includes one or more additional containers, each with one or more of various materials (such as reagents, optionally in concentrated form, and/or devices) desirable from a commercial and user standpoint for use of a compound described herein.
  • Non-limiting examples of such materials include, but not limited to, buffers, diluents, filters, needles, syringes; carrier, package, container, vial and/or tube labels listing contents and/or instructions for use, and package inserts with instructions for use.
  • a set of instructions will also typically be included.
  • a label is optionally on or associated with the container. For example, a label is on a container when letters, numbers or other characters forming the label are attached, molded or etched into the container itself, a label is associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert. In addition, a label is used to indicate that the contents are to be used for a specific therapeutic application.
  • the label indicates directions for use of the contents, such as in the methods described herein.
  • the pharmaceutical compositions is presented in a pack or dispenser device which contains one or more unit dosage forms containing a compound provided herein.
  • the pack for example contains metal or plastic foil, such as a blister pack.
  • the pack or dispenser device is accompanied by instructions for administration.
  • the pack or dispenser is
  • compositions containing a compound provided herein formulated in a compatible pharmaceutical carrier are prepared, placed in an
  • Embodiments of the present invention provide a method of inhibiting RAS-mediated cell signaling comprising contacting a cell with an effective amount of one or more compounds disclosed herein. Inhibition of RAS-mediated signal transduction can be assessed and demonstrated by a wide variety of ways known in the art.
  • Non-limiting examples include a showing of (a) a decrease in GTPase activity of RAS; (b) a decrease in GTP binding affinity or an increase in GDP binding affinity; (c) an increase in K off of GTP or a decrease in K off of GDP; (d) a decrease in the levels of signaling transduction molecules downstream in the RAS pathway, such as a decrease in pMEK level; and/or (e) a decrease in binding of RAS complex to downstream signaling molecules including but not limited to Raf. Kits and
  • Embodiments also provide methods of using the compounds or pharmaceutical compositions of the present invention to treat disease conditions, including but not limited to conditions implicated by G12C KRAS, HRAS or NRAS mutation, G12C HRAS mutation and/or G12C NRAS mutation (e.g., cancer).
  • disease conditions including but not limited to conditions implicated by G12C KRAS, HRAS or NRAS mutation, G12C HRAS mutation and/or G12C NRAS mutation (e.g., cancer).
  • a method for treatment of cancer comprising administering an effective amount of any of the foregoing pharmaceutical compositions comprising a compound of structure (I) to a subject in need thereof.
  • the cancer is mediated by a KRAS, HRAS or NRAS G12C mutation.
  • the cancer is pancreatic cancer, colon cancer, MYH associated polyposis, colorectal cancer or lung cancer.
  • the invention provides method of treating a disorder in a subject in need thereof, wherein the said method comprises determining if the subject has a KRAS, HRAS or NRAS G12C mutation and if the subject is determined to have the KRAS, HRAS or NRAS G12C mutation, then administering to the subject a therapeutically effective dose of at least one compound of structure (I) or a pharmaceutically acceptable salt, ester, prodrug, tautomer, solvate, hydrate or derivative thereof.
  • the disclosed compounds strongly inhibit anchorage-independent cell growth and therefore have the potential to inhibit tumor metastasis. Accordingly, in another embodiment the disclosure provides a method for inhibiting tumor metastasis, the method comprising administering an effective amount a pharmaceutical composition of comprising any of the compounds disclosed herein and a
  • KRAS, HRAS or RAS G12C mutations have also been identified in hematological malignancies (e.g., cancers that affect blood, bone marrow and/or lymph nodes). Accordingly, certain embodiments are directed to administration of a disclosed compounds (e.g., in the form of a pharmaceutical composition) to a patient in need of treatment of a hematological malignancy.
  • hematological malignancies e.g., cancers that affect blood, bone marrow and/or lymph nodes.
  • Such malignancies include, but are not limited to leukemias and lymphomas.
  • the presently disclosed compounds can be used for treatment of diseases such as Acute lymphoblastic leukemia (ALL), Acute myelogenous leukemia (AML), Chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), Chronic myelogenous leukemia (CML), Acute monocytic leukemia (AMoL) and/ or other leukemias.
  • ALL Acute lymphoblastic leukemia
  • AML Acute myelogenous leukemia
  • CLL Chronic lymphocytic leukemia
  • SLL small lymphocytic lymphoma
  • CML Chronic myelogenous leukemia
  • Acute monocytic leukemia Acute monocytic leukemia
  • the compounds are useful for treatment of lymphomas such as all subtypes of Hodgkin's lymphoma or non-Hodgkin's lymphoma.
  • a tumor or cancer comprises a G12C KRAS
  • HRAS or NRAS mutation can be undertaken by assessing the nucleotide sequence encoding the KRAS, HRAS or NRAS protein, by assessing the amino acid sequence of the KRAS, HRAS or NRAS protein, or by assessing the characteristics of a putative KRAS, HRAS or NRAS mutant protein.
  • the sequence of wild-type human KRAS, HRAS or NRAS is known in the art, (e.g. Accession No. NP203524).
  • Methods for detecting a mutation in a KRAS, HRAS or NRAS nucleotide sequence are known by those of skill in the art. These methods include, but are not limited to, polymeRASe chain reaction-restriction fragment length
  • PCR-RFLP polymeRASe chain reaction-single strand conformation polymorphism
  • PCR-SSCP polymeRASe chain reaction-single strand conformation polymorphism
  • MASA mutant allele-specific PCR amplification
  • direct sequencing primer extension reactions
  • electrophoresis oligonucleotide ligation assays
  • hybridization assays TaqMan assays
  • SNP genotyping assays high resolution melting assays and microarray analyses.
  • samples are evaluated for G12C KRAS, HRAS or NRAS mutations by real-time PCR.
  • fluorescent probes specific for the KRAS, HRAS or RAS G12C mutation are used. When a mutation is present, the probe binds and fluorescence is detected.
  • the KRAS, HRAS or NRAS G12C mutation is identified using a direct sequencing method of specific regions (e.g., exon 2 and/or exon 3) in the KRAS, HRAS or NRAS gene. This technique will identify all possible mutations in the region sequenced.
  • Methods for detecting a mutation in a KRAS, HRAS or NRAS protein are known by those of skill in the art. These methods include, but are not limited to, detection of a KRAS, HRAS or NRAS mutant using a binding agent (e.g., an antibody) specific for the mutant protein, protein electrophoresis and Western blotting, and direct peptide sequencing.
  • a binding agent e.g., an antibody
  • Methods for determining whether a tumor or cancer comprises a G12C KRAS, HRAS or NRAS mutation can use a variety of samples.
  • the sample is taken from a subject having a tumor or cancer.
  • the sample is taken from a subject having a cancer or tumor.
  • the sample is a fresh tumor/cancer sample.
  • the sample is a frozen tumor/cancer sample.
  • the sample is a formalin-fixed paraffin- embedded sample.
  • the sample is processed to a cell lysate.
  • the sample is processed to DNA or RNA.
  • Embodiments of the invention also relate to a method of treating a hyperproliferative disorder in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
  • said method relates to the treatment of cancer such as acute myeloid leukemia, cancer in adolescents, adrenocortical carcinoma childhood, AIDS- related cancers (e.g.
  • Lymphoma and Kaposi's Sarcoma anal cancer, appendix cancer, astrocytomas, atypical teratoid, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumors, burkitt lymphoma, carcinoid tumor, atypical teratoid, embryonal tumors, germ cell tumor, primary lymphoma, cervical cancer, childhood cancers, chordoma, cardiac tumors, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myleoproliferative disorders, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, extrahepatic ductal carcinoma in situ (DCIS), embryonal tumors, CNS cancer, endometrial cancer, ependymoma, esophageal cancer, esthesioneuroblastoma,
  • myelodysplastic syndromes my elodysplastic/my el oproliferative neoplasms, multiple myeloma, merkel cell carcinoma, malignant mesothelioma, malignant fibrous histiocytoma of bone and osteosarcoma, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-Hodgkin's lymphoma, non-small cell lung cancer (NSCLC), oral cancer, lip and oral cavity cancer, oropharyngeal cancer, ovarian cancer, pancreatic cancer, papillomatosis, paraganglioma, paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pleuropulmonary blastoma, primary central nervous system (CNS) lymphoma, prostate cancer, rectal cancer, transitional cell cancer, retinoblastoma, rhabdomyosarcoma, salivary gland
  • said method relates to the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
  • a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
  • the invention relates to methods for treatment of lung cancers, the methods comprise administering an effective amount of any of the above described compound (or a pharmaceutical composition comprising the same) to a subject in need thereof.
  • the lung cancer is a non- small cell lung carcinoma (NSCLC), for example adenocarcinoma, squamous-cell lung carcinoma or large-cell lung carcinoma.
  • the lung cancer is a small cell lung carcinoma.
  • Other lung cancers treatable with the disclosed compounds include, but are not limited to, glandular tumors, carcinoid tumors and undifferentiated carcinomas.
  • Subjects that can be treated with compounds of the invention, or pharmaceutically acceptable salt, ester, prodrug, solvate, tautomer, hydrate or derivative of said compounds, according to the methods of this invention include, for example, subjects that have been diagnosed as having acute myeloid leukemia, acute myeloid leukemia, cancer in adolescents, adrenocortical carcinoma childhood, AIDS-related cancers (e.g.
  • Lymphoma and Kaposi's Sarcoma anal cancer, appendix cancer, astrocytomas, atypical teratoid, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumors, burkitt lymphoma, carcinoid tumor, atypical teratoid, embryonal tumors, germ cell tumor, primary lymphoma, cervical cancer, childhood cancers, chordoma, cardiac tumors, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myleoproliferative disorders, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, extrahepatic ductal carcinoma in situ (DCIS), embryonal tumors, CNS cancer, endometrial cancer, ependymoma, esophageal cancer,
  • CLL chronic lymphocytic
  • esthesioneuroblastoma ewing sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor, eye cancer, fibrous histiocytoma of bone, gall bladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumors (GIST), germ cell tumor, gestational trophoblastic tumor, hairy cell leukemia, head and neck cancer, heart cancer, liver cancer, Hodgkin's lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumors, pancreatic neuroendocrine tumors, kidney cancer, laryngeal cancer, lip and oral cavity cancer, liver cancer, lobular carcinoma in situ (LCIS), lung cancer, lymphoma, metastatic squamous neck cancer with occult primary, midline tract carcinoma, mouth cancer multiple endocrine neoplasia syndromes, multiple myeloma/plasma cell neoplasm, mycosis fungoides, myel
  • subjects that are treated with the compounds of the invention include subjects that have been diagnosed as having a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e.g., benign prostatic hypertrophy (BPH)).
  • a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e.g., benign prostatic hypertrophy (BPH)).
  • Embodiments of the invention further provide methods of modulating a G12C Mutant KRAS, HRAS or RAS protein activity by contacting the protein with an effective amount of a compound of the invention. Modulation can be inhibiting or activating protein activity.
  • the invention provides methods of inhibiting protein activity by contacting the G12C Mutant KRAS, HRAS or NRAS protein with an effective amount of a compound of the invention in solution.
  • the invention provides methods of inhibiting the G12C Mutant KRAS, HRAS or NRAS protein activity by contacting a cell, tissue, organ that express the protein of interest.
  • the invention provides methods of inhibiting protein activity in subject including but not limited to rodents and mammal (e.g., human) by administering into the subject an effective amount of a compound of the invention.
  • the percentage modulation exceeds 25%, 30%, 40%, 50%), 60%), 70%), 80%), or 90%.
  • the percentage of inhibiting exceeds 25%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%.
  • the invention provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in a cell by contacting said cell with an amount of a compound of the invention sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said cell. In some embodiments, the invention provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in a tissue by contacting said tissue with an amount of a compound of the invention sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said tissue.
  • the invention provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in an organism by contacting said organism with an amount of a compound of the invention sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said organism. In some embodiments, the invention provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in an animal by contacting said animal with an amount of a compound of the invention sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said animal.
  • the invention provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in a mammal by contacting said mammal with an amount of a compound of the invention sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said mammal.
  • the invention provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in a human by contacting said human with an amount of a compound of the invention sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said human.
  • the present invention provides methods of treating a disease mediated by KRAS, HRAS or NRAS G12C activity in a subject in need of such treatment.
  • inventions provide methods for combination therapies in which an agent known to modulate other pathways, or other components of the same pathway, or even overlapping sets of target enzymes are used in combination with a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, tautomer, hydrate or derivative thereof.
  • such therapy includes but is not limited to the combination of one or more compounds of the invention with
  • chemotherapeutics are presently known in the art and can be used in combination with the compounds of the invention.
  • the chemotherapeutic is selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomeRASe inhibitors, biological response modifiers, anti- hormones, angiogenesis inhibitors, and anti-androgens.
  • Non-limiting examples are chemotherapeutic agents, cytotoxic agents, and non-peptide small molecules such as Gleevec® (Imatinib Mesylate), Velcade® (bortezomib), Casodex (bicalutamide), Iressa® (gefitinib), and Adriamycin as well as a host of chemotherapeutic agents.
  • Non-limiting examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXANTM); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and
  • methylamelamines including altretamine, triethylenemelamine,
  • trietylenephosphoramide triethylenethiophosphaoramide and trimethylolomelamine
  • nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard
  • nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine
  • antibiotics such as aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, calicheamicin, carabicin, carminomycin, carzinophilin, CasodexTM, chromomycins, dactinomycin, daunorubicin, detorub
  • dideoxyuridine, doxifluridine, enocitabine, floxuridine, androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfomithine; elliptinium acetate; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubici
  • Ara-C arabinoside
  • cyclophosphamide thiotepa
  • taxanes e.g. paclitaxel
  • chemotherapeutic cell conditioners include anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens including for example tamoxifen, (NolvadexTM), raloxifene, aromatase inhibiting 4(5)- imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY 117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as
  • the compounds or pharmaceutical composition of the present invention can be used in combination with commonly prescribed anti-cancer drugs such as Herceptin®, Avastin®, Erbitux®, Rituxan®, Taxol®, Arimidex®, Taxotere®, ABVD, AVICINE, Abagovomab, Acridine carboxamide, Adecatumumab, 17-N-Allylamino- 17-demethoxygeldanamycin,
  • Antitumorigenic herbs Apaziquone, Atiprimod, Azathioprine, Belotecan,
  • sulfoximine CBV (chemotherapy), Calyculin, cell-cycle nonspecific antineoplastic agents, Dichloroacetic acid, Discodermolide, Elsamitrucin, Enocitabine, Epothilone, Eribulin, Everolimus, Exatecan, Exisulind, Ferruginol, Forodesine, Fosfestrol, ICE chemotherapy regimen, IT-101, Imexon, Imiquimod, Indolocarbazole, Irofulven, Laniquidar, Larotaxel, Lenalidomide, Lucanthone, Lurtotecan, Mafosfamide,
  • Mitozolomide Nafoxidine, Nedaplatin, Olaparib, Ortataxel, PAC-1, Pawpaw,
  • Pixantrone Proteasome inhibitor, Rebeccamycin, Resiquimod, Rubitecan, SN-38, Salinosporamide A, Sapacitabine, Stanford V, Swainsonine, Talaporfin, Tariquidar, Tegafur-uracil, Temodar, Tesetaxel, Triplatin tetranitrate, Tris(2-chloroethyl)amine, Troxacitabine, Uramustine, Vadimezan, Vinflunine, ZD6126 or Zosuquidar.
  • Embodiments further relate to a method for using the compounds or pharmaceutical compositions provided herein, in combination with radiation therapy for inhibiting abnormal cell growth or treating the hyperproliferative disorder in the mammal.
  • Techniques for administering radiation therapy are known in the art, and these techniques can be used in the combination therapy described herein.
  • the administration of the compound of the invention in this combination therapy can be determined as described herein.
  • Radiation therapy can be administered through one of several methods, or a combination of methods, including without limitation external-beam therapy, internal radiation therapy, implant radiation, stereotactic radiosurgery, systemic radiation therapy, radiotherapy and permanent or temporary interstitial brachytherapy.
  • brachytherapy refers to radiation therapy delivered by a spatially confined radioactive material inserted into the body at or near a tumor or other proliferative tissue disease site.
  • the term is intended without limitation to include exposure to radioactive isotopes (e.g. At-211, 1-131, 1-125, Y-90, Re-186, Re-188, Sm- 153, Bi-212, P-32, and radioactive isotopes of Lu).
  • Suitable radiation sources for use as a cell conditioner of the present invention include both solids and liquids.
  • the radiation source can be a radionuclide, such as 1-125, 1-131, Yb-169, Ir-192 as a solid source, 1-125 as a solid source, or other radionuclides that emit photons, beta particles, gamma radiation, or other therapeutic rays.
  • the radioactive material can also be a fluid made from any solution of radionuclide(s), e.g., a solution of 1-125 or 1-131, or a radioactive fluid can be produced using a slurry of a suitable fluid containing small particles of solid radionuclides, such as Au-198, Y-90.
  • the radionuclide(s) can be embodied in a gel or radioactive micro spheres.
  • this invention further relates to a method for sensitizing abnormal cells in a mammal to treatment with radiation which comprises administering to the mammal an amount of a compound of the present invention or pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof, which amount is effective is sensitizing abnormal cells to treatment with radiation.
  • the amount of the compound, salt, or solvate in this method can be determined according to the means for ascertaining effective amounts of such compounds described herein.
  • the compounds or pharmaceutical compositions of the invention can be used in combination with an amount of one or more substances selected from anti- angiogenesis agents, signal transduction inhibitors, antiproliferative agents, glycolysis inhibitors, or autophagy inhibitors.
  • Anti-angiogenesis agents such as MMP-2 (matrix -metalloproteinase 2) inhibitors, MMP-9 (matrix-metalloprotienase 9) inhibitors, and COX-11 (cyclooxygenase 11) inhibitors, can be used in conjunction with a compound of the invention and pharmaceutical compositions described herein.
  • Anti-angiogenesis agents include, for example, rapamycin, temsirolimus (CCI-779), everolimus (RAD001), sorafenib, sunitinib, and bevacizumab.
  • Examples of useful COX-II inhibitors include CELEBREXTM (alecoxib), valdecoxib, and rofecoxib.
  • MMP-2 and MMP-9 inhibitors are those that have little or no activity inhibiting MMP-1. More preferred, are those that selectively inhibit MMP-2 and/or AMP-9 relative to the other matrix- metalloproteinases (i. e., MAP-1, MMP-3, MMP-4, MMP-5, MMP-6, MMP- 7, MMP- 8, MMP-10, MMP-11, MMP-12, andMMP-13).
  • MMP inhibitors useful in the invention are AG-3340, RO 32-3555, and RS 13-0830.
  • Autophagy inhibitors include, but are not limited to chloroquine, 3- methyladenine, hydroxychloroquine (PlaquenilTM), bafilomycin Al, 5-amino-4- imidazole carboxamide riboside (AICAR), okadaic acid, autophagy-suppressive algal toxins which inhibit protein phosphatases of type 2 A or type 1, analogues of cAMP, and drugs which elevate cAMP levels such as adenosine, LY204002, N6-mercaptopurine riboside, and vinblastine.
  • antisense or siRNA that inhibits expression of proteins including but not limited to ATG5 (which are implicated in autophagy), may also be used.
  • Embodiments also relate to a method of and to a pharmaceutical composition for treating a cardiovascular disease in a mammal which comprises an amount of a compound of the invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, tautomer, hydrate or derivative thereof, or an isotopically-labeled derivative thereof, and an amount of one or more therapeutic agents use for the treatment of cardiovascular diseases.
  • agents for use in cardiovascular disease applications are antithrombotic agents, e.g., prostacyclin and salicylates, thrombolytic agents, e.g., streptokinase, urokinase, tissue plasminogen activator (TP A) and anisoylated plasminogen-streptokinase activator complex (APS AC), anti -platelets agents, e.g., acetyl-salicylic acid (ASA) and clopidrogel, vasodilating agents, e.g., nitrates, calcium channel blocking drugs, anti -proliferative agents, e.g., colchicine and alkylating agents, intercalating agents, growth modulating factors such as interleukins, transformation growth factor-beta and congeners of platelet derived growth factor, monoclonal antibodies directed against growth factors, anti-inflammatory agents, both steroidal and non-steroidal, and other agents that can modulate vessel tone, function,
  • Antibiotics can also be included in combinations or coatings comprised by the invention. Moreover, a coating can be used to effect therapeutic delivery focally within the vessel wall. By incorporation of the active agent in a swellable polymer, the active agent will be released upon swelling of the polymer.
  • the compounds described herein are formulated or administered in conjunction with liquid or solid tissue barriers also known as lubricants.
  • tissue barriers include, but are not limited to, polysaccharides, polyglycans, seprafilm, interceed and hyaluronic acid.
  • medicaments which are administered in conjunction with the compounds described herein include any suitable drugs usefully delivered by inhalation for example, analgesics, e.g. codeine, dihydromorphine, ergotamine, fentanyl or morphine; anginal preparations, e.g. diltiazem; antiallergics, e.g.
  • anti-infectives e.g. cephalosporins, penicillins, streptomycin, sulphonamides, tetracyclines or pentamidine
  • antihistamines e.g. methapyrilene
  • anti-inflammatories e.g. beclomethasone, flunisolide, budesonide, tipredane, triamcinolone acetonide or fluticasone
  • antitussives e.g. noscapine
  • noscapine e.g. noscapine
  • bronchodilators e.g. ephedrine, adrenaline, fenoterol, formoterol, isoprenaline, metaproterenol, phenylephrine, phenylpropanolamine, pirbuterol, reproterol, rimiterol, salbutamol, salmeterol, terbutalin, isoetharine, tulobuterol, orciprenaline or (-)-4-amino- 3,5-dichloro-a-[[[6-[2-(2-pyridinyl)ethoxy]hexyl]-amino]methyl]benzenemethanol; diuretics, e.g.
  • the medicaments are used in the form of salts (e.g. as alkali metal or amine salts or as acid addition salts) or as esters (e.g. lower alkyl esters) or as solvates (e.g. hydrates) to optimize the activity and/or stability of the medicament.
  • salts e.g. as alkali metal or amine salts or as acid addition salts
  • esters e.g. lower alkyl esters
  • solvates e.g. hydrates
  • exemplary therapeutic agents useful for a combination therapy include but are not limited to agents as described above, radiation therapy, hormone antagonists, hormones and their releasing factors, thyroid and antithyroid drugs, estrogens and progestins, androgens, adrenocorticotropic hormone; adrenocortical steroids and their synthetic analogs; inhibitors of the synthesis and actions of adrenocortical hormones, insulin, oral hypoglycemic agents, and the pharmacology of the endocrine pancreas, agents affecting calcification and bone turnover: calcium, phosphate, parathyroid hormone, vitamin D, calcitonin, vitamins such as water-soluble vitamins, vitamin B complex, ascorbic acid, fat-soluble vitamins, vitamins A, K, and E, growth factors, cytokines, chemokines, muscarinic receptor agonists and antagonists; anticholinesteRASe agents; agents acting at the neuromuscular junction and/or autonomic ganglia; catecholamines, sympathomimetic
  • Therapeutic agents can also include agents for pain and inflammation such as histamine and histamine antagonists, bradykinin and bradykinin antagonists, 5- hydroxytryptamine (serotonin), lipid substances that are generated by biotransformation of the products of the selective hydrolysis of membrane phospholipids, eicosanoids, prostaglandins, thromboxanes, leukotrienes, aspirin, nonsteroidal anti-inflammatory agents, analgesic-antipyretic agents, agents that inhibit the synthesis of prostaglandins and thromboxanes, selective inhibitors of the inducible cyclooxygenase, selective inhibitors of the inducible cyclooxygenase-2, autacoids, paracrine hormones, somatostatin, gastrin, cytokines that mediate interactions involved in humoral and cellular immune responses, lipid-derived autacoids, eicosanoids, ⁇ -adrenergic agonists, ipratropium
  • Additional therapeutic agents contemplated herein include diuretics, vasopressin, agents affecting the renal conservation of water, rennin, angiotensin, agents useful in the treatment of myocardial ischemia, anti-hypertensive agents, angiotensin converting enzyme inhibitors, ⁇ -adrenergic receptor antagonists, agents for the treatment of hypercholesterolemia, and agents for the treatment of dyslipidemia.
  • therapeutic agents contemplated include drugs used for control of gastric acidity, agents for the treatment of peptic ulcers, agents for the treatment of gastroesophageal reflux disease, prokinetic agents, antiemetics, agents used in irritable bowel syndrome, agents used for diarrhea, agents used for constipation, agents used for inflammatory bowel disease, agents used for biliary disease, agents used for pancreatic disease.
  • Therapeutic agents used to treat protozoan infections drugs used to treat Malaria, Amebiasis, Giardiasis, Trichomoniasis, Trypanosomiasis, and/or
  • Leishmaniasis and/or drugs used in the chemotherapy of helminthiasis.
  • Other therapeutic agents include antimicrobial agents, sulfonamides, trimethoprim- sulfamethoxazole quinolones, and agents for urinary tract infections, penicillins, cephalosporins, and other, ⁇ -lactam antibiotics, an agent comprising an aminoglycoside, protein synthesis inhibitors, drugs used in the chemotherapy of tuberculosis,
  • mycobacterium avium complex disease and leprosy, antifungal agents, antiviral agents including nonretroviral agents and antiretroviral agents.
  • therapeutic agents used for immunomodulation such as immunomodulators, immunosuppressive agents, tolerogens, and immunostimulants are contemplated by the methods herein.
  • therapeutic agents acting on the blood and the blood-forming organs hematopoietic agents, growth factors, minerals, and vitamins, anticoagulant, thrombolytic, and antiplatelet drugs.
  • a compound of the present invention For treating renal carcinoma, one may combine a compound of the present invention with sorafenib and/or avastin.
  • a compound of the present invention For treating an endometrial disorder, one may combine a compound of the present invention with doxorubincin, taxotere (taxol), and/or cisplatin (carboplatin).
  • doxorubincin for treating an endometrial disorder, one may combine doxorubincin, taxotere (taxol), and/or cisplatin (carboplatin).
  • cisplatin carboxyribonitride
  • doxorubincin taxotere
  • doxorubincin taxotere
  • doxorubincin taxotere
  • doxorubincin topotecan
  • tamoxifen for treating renal carcinoma, one may combine a compound of the present invention with sorafenib and/or avastin.
  • a compound of the present invention For treating breast cancer, one may combine a compound of the present invention with taxotere (taxol), gemcitabine (capecitabine), tamoxifen, letrozole, tarceva, lapatinib, PD0325901, avastin, herceptin, OSI-906, and/or OSI-930.
  • taxotere taxotere
  • gemcitabine gemcitabine
  • tamoxifen letrozole
  • tarceva lapatinib
  • PD0325901 avastin
  • herceptin herceptin
  • OSI-906 herceptin
  • OSI-930 for treating lung cancer, one may combine a compound of the present invention with taxotere (taxol), gemcitabine, cisplatin, pemetrexed, Tarceva, PD0325901, and/or avastin.
  • agents useful in methods for combination therapy with one or more compounds of structure (I) include, but are not limited to: Erlotinib, Afatinib, Iressa, GDC0941, MLN1117, BYL719 (Alpelisib), BKM120 (Buparlisib), CYT387, GLPG0634, Baricitinib, Lestaurtinib, momelotinib, Pacritinib, Ruxolitinib, TG101348, Crizotinib, tivantinib, AMG337, cabozantinib, foretinib, onartuzumab, NVP-AEW541, Dasatinib, Ponatinib, saracatinib, bosutinib, trametinib, selumetinib, cobimetinib, PD0325901, R05126766, Axitinib,
  • the compounds described herein can be used in combination with the agents disclosed herein or other suitable agents, depending on the condition being treated. Hence, in some embodiments the one or more compounds of the invention will be co-administered with other agents as described above.
  • the compounds described herein are administered with the second agent simultaneously or separately.
  • This administration in combination can include simultaneous administration of the two agents in the same dosage form, simultaneous administration in separate dosage forms, and separate administration. That is, a compound described herein and any of the agents described above can be formulated together in the same dosage form and administered simultaneously. Alternatively, a compound of the invention and any of the agents described above can be
  • a compound of the present invention can be administered just followed by and any of the agents described above, or vice versa.
  • a compound of the invention and any of the agents described above are administered a few minutes apart, or a few hours apart, or a few days apart.
  • Compound 1 was prepared according to the above synthetic scheme as follows.
  • Test compounds were prepared as 10 mM stock solutions in DMSO
  • the MS instrument is set to positive polarity, 2 GHz resolution, and low mass (1700) mode and allowed to equilibrate for 30 minutes. The instrument is then calibrated, switched to acquisition mode and the appropriate method loaded.
  • a blank batch i.e., buffer
  • the samples are thawed at 37°C for 10 minutes, briefly centrifuged, and transfer to the bench top.
  • Wells Al and H12 are spiked with 1 uL 500 uM internal standard peptide, and the plates centrifuged at 2000 x g for 5 minutes. The method is then run and masses of each individual well recorded.
  • KRAS G12C protein species were measured using a Dionex RSLCnano system (Thermo Scientific) connected to a Q Exactive Plus mass spectrometer (Thermo Scientific).
  • the mass spectrometer was operated in profile mode at a resolution of 17500, 5 microscans, using 50 msec max injection time and an AGC target of le6, and a full mass range from 800-1850 m/z was recorded.
  • the HCD trapping gas was optimized for maximum sensitivity for intact proteins.
  • the ionization method was electrospray ionization, which used a spray voltage of 4kV, sheath gas flow set to 50 au, auxiliary gas flow set to 10 au and sweep gas flow set to 1 au.
  • the capillary ion transfer temperature was 320°C and the S-lens RF level was set to 50 voltage.
  • Deconvolution software (Thermo Scientific) was used to deconvolute the charge envelopes of protein species in samples.
  • the ability of the subject compounds to inhibit RAS-mediated cell growth is assessed and demonstrated as follows.
  • Cells expressing a wildtype or a mutant RAS are plated in white, clear bottom 96 well plates at a density of 5,000 cells per well. Cells are allowed to attach for about 2 hours after plating before a compound disclosed herein is added. After certain hours (e.g., 24 hours, 48 hours, or 72 hours of cell growth), cell proliferation is determined by measuring total ATP content using the Cell Titer Glo reagent (Promega) according to manufacturer's instructions.
  • Proliferation EC50s is determined by analyzing 8 point compound dose responses at half-log intervals decreasing from 100 ⁇ .
  • the ability of the compounds disclosed herein in inhibiting RAS- mediated signaling is assessed and demonstrated as follows.
  • Cells expressing wild type or a mutant RAS such as G12C, G12V, or G12A
  • RAS wild type or a mutant RAS
  • Inhibition of RAS signaling by one or more subject compounds is demonstrated by a decrease in the steady-state level of phosphorylated MEK, phosphorylated ERK, phosphorylated RSK, and/or Raf binding in cells treated with the one or more of the subject compounds as compared to the control cells.
  • Compound 2 was tested according to the above methods and found to covalently bind to KRAS G12C to the extent of greater than about 30% (i.e., at least about 30% of the protein present in the well was found to be covalently bound to test compound). Other compounds were tested under similar conditions. Results are presented in Table 2.
  • indicates binding activity determined at 4 hours

Abstract

Compounds having activity as inhibitors of G12C mutant KRAS protein are provided. The compounds have the following structure (I) or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof, wherein R, R1, R2a, R2b, R2c, A, B, L1 and E are as defined herein. Methods associated with preparation and use of such compounds, pharmaceutical compositions comprising such compounds and methods to modulate the activity of G12C mutant KRAS protein for treatment of disorders, such as cancer, are also provided.

Description

SUBSTITUTED QUINAZOLINE COMPOUNDS AND THEIR USE AS INHIBITORS OF G12C MUTANT KRAS, HRAS AND/OR NRAS PROTEINS
BACKGROUND
Technical Field
The present invention is generally directed to novel compounds and methods for their preparation and use as therapeutic or prophylactic agents, for example for treatment of cancer.
Description of the Related Art
RAS represents a group of closely related monomeric globular proteins of 189 amino acids (21 kDa molecular mass) which are associated with the plasma membrane and which bind either GDP or GTP. RAS acts as a molecular switch. When RAS contains bound GDP, it is in the resting or off position and is "inactive". In response to exposure of the cell to certain growth promoting stimuli, RAS is induced to exchange its bound GDP for a GTP. With GTP bound, RAS is "switched on" and is able to interact with and activate other proteins (its "downstream targets"). The RAS protein itself has a very low intrinsic ability to hydrolyze GTP back to GDP, thus turning itself into the off state. Switching RAS off requires extrinsic proteins termed GTPase-activating proteins (GAPs) that interact with RAS and greatly accelerate the conversion of GTP to GDP. Any mutation in RAS which affects its ability to interact with GAP or to convert GTP back to GDP will result in a prolonged activation of the protein and consequently a prolonged signal to the cell telling it to continue to grow and divide. Because these signals result in cell growth and division, overactive RAS signaling may ultimately lead to cancer.
Structurally, RAS proteins contain a G domain which is responsible for the enzymatic activity of RAS - the guanine nucleotide binding and the hydrolysis (GTPase reaction). It also contains a C-terminal extension, known as the CAAX box, which may be post-translationally modified and is responsible for targeting the protein to the membrane. The G domain is approximately 21-25 kDa in size and it contains a phosphate binding loop (P-loop). The P-loop represents the pocket where the nucleotides are bound in the protein, and this is the rigid part of the domain with conserved amino acid residues which are essential for nucleotide binding and hydrolysis (Glycine 12, Threonine 26 and Lysine 16). The G domain also contains the so called Switch I (residues 30-40) and Switch II (residues 60-76) regions, both of which are the dynamic parts of the protein which are often represented as the "spring- loaded" mechanism because of their ability to switch between the resting and loaded state. The key interaction is the hydrogen bonds formed by Threonine-35 and glycine- 60 with the γ-phosphate of GTP which maintain Switch 1 and Switch 2 regions respectively in their active conformation. After hydrolysis of GTP and release of phosphate, these two relax into the inactive GDP conformation.
The most notable members of the RAS subfamily are HRAS, KRAS and RAS, mainly for being implicated in many types of cancer. However, there are many other members including DIRAS1; DIRAS2; DIRAS3; ERAS; GEM; MRAS;
KIRAS1; KIRAS2; NRAS; RALA; RALB; RAPIA; RAPIB; RAP2A; RAP2B; RAP2C; RASD1; RASD2; RASL10A; RASL10B; RASL11A; RASL11B; RASL12; REM1; REM2; RERG; RERGL; RRAD; RRAS and RRAS2.
Mutations in any one of the three main isoforms of RAS (HRAS, NRAS, or KRAS) genes are among the most common events in human tumorigenesis. About 30% of all human tumors are found to carry some mutation in RAS genes.
Remarkably, KRAS mutations are detected in 25-30% of tumors. By comparison, the rates of oncogenic mutation occurring in the NRAS and HRAS family members are much lower (8% and 3% respectively). The most common KRAS mutations are found at residue G12 and G13 in the P-loop and at residue Q61.
G12C is a frequent mutation of KRAS gene (glycine-12 to cysteine). This mutation had been found in about 13% of cancer occurrences, about 43% of lung cancer occurrences, and in almost 100% of MYH-associates polyposis (familial colon cancer syndrome). However targeting this gene with small molecules is a challenge. Accordingly, while progress has been made in this field, there remains a need in the art for improved compounds and methods for treatment of cancer, for example by inhibition of KRAS, HRAS or NRAS. The present invention fulfills this need and provides further related advantages. BRIEF SUMMARY
In brief, the present invention provides compounds, including stereoisomers, pharmaceutically acceptable salts, tautomers and prodrugs thereof, which are capable of modulating G12C mutant KRAS, HRAS and/or NRAS proteins. In some instances, the compounds act as electrophiles which are capable of forming a covalent bond with the cysteine residue at position 12 of a KRAS, HRAS or NRAS G12C mutant protein. Methods for use of such compounds for treatment of various diseases or conditions, such as cancer, are also provided.
In one embodiment, compounds having the following structure (I) are provided:
(I)
or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof, wherein R, R1,
R2a R2b R2c A B Ll E
are as defined herein. Pharmaceutical compositions comprising one or more compounds of structure (I) and a pharmaceutically acceptable carrier are also provided in various other embodiments.
In other embodiments, the present invention provides a method for treatment of cancer, the method comprising administering an effective amount of a pharmaceutical composition comprising any one or more of the compounds of structure (I) to a subject in need thereof.
Other provided methods include a method for regulating activity of a
KRAS, HRAS or NRAS G12C mutant protein, the method comprising reacting the KRAS, HRAS or NRAS G12C mutant protein with any one of the compounds of structure (I). In other embodiments, a method for inhibiting proliferation of a cell population, the method comprising contacting the cell population with any one of the compounds of structure (I) is also provided.
In other embodiments, the invention is directed to a method for treating a disorder mediated by a KRAS, HRAS or NRAS G12C mutation in a subject in need thereof, the method comprising:
determining if the subject has a KRAS, HRAS or NRAS G12C mutation; and
if the subject is determined to have the KRAS, HRAS or NRAS G12C mutation, then administering to the subject a therapeutically effective amount of a pharmaceutical composition comprising any one or more compounds of structure (I).
In still more embodiments, the invention is directed to a method for preparing a labeled KRAS, HRAS or NRAS G12C mutant protein, the method comprising reacting the KRAS, HRAS or NRAS G12C mutant with a compound of structure (I), to result in the labeled KRAS, HRAS or NRAS G12C protein.
These and other aspects of the invention will be apparent upon reference to the following detailed description.
BRIEF DESCRIPTION OF THE DRAWINGS
In the figures, identical reference numbers identify similar elements. The sizes and relative positions of elements in the figures are not necessarily drawn to scale and some of these elements are arbitrarily enlarged and positioned to improve figure legibility. Further, the particular shapes of the elements as drawn are not intended to convey any information regarding the actual shape of the particular elements, and have been solely selected for ease of recognition in the figures.
Fig. 1 illustrates the enzymatic activity of RAS.
Fig. 2 depicts a signal transduction pathway for RAS.
Fig. 3 shows some common oncogenes, their respective tumor type and cumulative mutation frequencies (all tumors). DETAILED DESCRIPTION
In the following description, certain specific details are set forth in order to provide a thorough understanding of various embodiments of the invention.
However, one skilled in the art will understand that the invention may be practiced without these details.
Unless the context requires otherwise, throughout the present specification and claims, the word "comprise" and variations thereof, such as,
"comprises" and "comprising" are to be construed in an open, inclusive sense, that is as "including, but not limited to".
Reference throughout this specification to "one embodiment" or "an embodiment" means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment of the present invention. Thus, the appearances of the phrases "in one embodiment" or "in an embodiment" in various places throughout this specification are not necessarily all referring to the same embodiment. Furthermore, the particular features, structures, or characteristics may be combined in any suitable manner in one or more embodiments.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this invention belongs. As used in the specification and claims, the singular form "a", "an" and "the" include plural references unless the context clearly dictates otherwise.
"Amino" refers to the -NH2 radical.
"Carboxy" or "carboxyl" refers to the -C02H radical.
"Cyano" refers to the -CN radical.
"Hydroxy" or "hydroxyl" refers to the -OH radical.
"Imino" refers to the =NH substituent.
"Nitro" refers to the -N02 radical.
"Oxo" refers to the =0 substituent.
"Thioxo" refers to the =S substituent.
"Alkyl" refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds), having from one to twelve carbon atoms (C1-C12 alkyl), preferably one to eight carbon atoms (Ci-C8 alkyl) or one to six carbon atoms (Ci-C6 alkyl), and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, ^-propyl, 1-methylethyl (z'so-propyl), «-butyl, «-pentyl, 1, 1-dimethylethyl (t-butyl), 3-methylhexyl, 2-methylhexyl, ethenyl, prop-l-enyl, but-l-enyl, pent-l-enyl, penta-l,4-dienyl, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. In some embodiments alkyls are saturated. In other
embodiments alkyl is unsaturated and thus includes alkenyls (one or more carbon- carbon double bonds) and/or alkynyls (one or more carbon-carbon triple bonds such as ethynyl and the like). Unless stated otherwise specifically in the specification, an alkyl group is optionally substituted.
"Alkylene" or "alkylene chain" refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds), and having from one to twelve carbon atoms, e.g., methylene, ethylene, propylene, «-butylene, ethenylene, propenylene, «-butenylene, propynylene, «-butynylene, and the like. The alkylene chain is attached to the rest of the molecule through a single or double bond and to the radical group through a single or double bond. The points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, an alkylene chain is optionally substituted.
"Alkoxy" refers to a radical of the formula -ORa where Ra is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkoxy group is optionally substituted.
"Alkoxyalkyl" refers to a radical of the formula -Rt,ORa where Ra is an alkyl radical as defined above containing one to twelve carbon atoms and Rb is an alkylene radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkoxyalkyl group is optionally substituted. "Alkoxycarbonyl" refers to a radical of the formula -C(=0)ORa where Ra is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkoxycarbonyl group is optionally substituted.
"Alkylaminyl" refers to a radical of the formula - HRa or - RaRa where each Ra is, independently, an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkylaminyl group is optionally substituted.
"Aminylalkyl" refers to an alkyl group comprising at least one aminyl substituent (- RaRb wherein Ra and R are each independently H or Ci-C6 alkyl). The aminyl substituent can be on a tertiary, secondary or primary carbon. Unless stated otherwise specifically in the specification, an aminylalkyl group is optionally substituted.
"Aminylalkylaminyl" refers to a radical of the formula - RaRb wherein Ra is H or Ci-C6 alkyl and Rb is aminylalkyl. Unless stated otherwise specifically in the specification, an aminylalkylaminyl group is optionally substituted.
"Aminylalkoxy" refers to a radical of the formula -ORa H2 wherein Ra is alkylene. Unless stated otherwise specifically in the specification, an aminylalkoxy group is optionally substituted.
"Alkylaminylalkoxy" refers to a radical of the formula -ORa RbRc wherein Ra is alkylene and Rb and Rc are each independently H or Ci-C6 alkyl, provided one of Rb or Rc is Ci-C6 alkyl. Unless stated otherwise specifically in the specification, an alkylaminylalkoxy group is optionally substituted.
"Alkylcarbonylaminyl" refers to a radical of the formula -NH(C=0)Ra where Ra is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkylcarbonylaminyl group is optionally substituted. An alkenylcarbonylaminyl is an alkylcarbonylaminyl containing at least one carbon-carbon double bond. An alkenylcarbonylaminyl group is optionally substituted. "Alkylcarbonylaminylalkoxy" refers to a radical of the formula
-ORb H(C=0)Ra where Ra is an alkyl radical as defined above containing one to twelve carbon atoms and R is alkylene. Unless stated otherwise specifically in the specification, an alkylcarbonylaminylalkoxy group is optionally substituted.
"Aminylcarbonylalkyl" refers to a radical of the formula - RcC(=0) RaRb, where Ra and Rb are each independently H or alkyl and Rc is alkylene. Unless stated otherwise specifically in the specification, an aminylcarbonylalkyl group is optionally substituted.
"Aryl" refers to a carbocyclic ring system radical comprising hydrogen, 6 to 18 carbon atoms and at least one aromatic ring. For purposes of this invention, the aryl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems. Aryl radicals include, but are not limited to, aryl radicals derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, as-indacene, s-indacene, indane, indene, naphthalene, phenalene, phenanthrene, pleiadene, pyrene, and triphenylene. Unless stated otherwise specifically in the specification, the term "aryl" or the prefix "ar-" (such as in "aralkyl") is meant to include aryl radicals that are optionally substituted.
"Aralkyl" refers to a radical of the formula -Rb-Rc where Rb is an alkylene chain as defined above and Rc is one or more aryl radicals as defined above, for example, benzyl, diphenylmethyl and the like. Unless stated otherwise specifically in the specification, an aralkyl group is optionally substituted.
"Carboxyalkyl" refers to a radical of the formula -Rb-Rc where Rb is an alkylene chain as defined above and Rc is a carboxyl group as defined above. Unless stated otherwise specifically in the specification, carboxyalkyl group is optionally substituted.
"Cyanoalkyl" refers to a radical of the formula -Rb-Rc where Rb is an alkylene chain as defined above and Rc is a cyano group as defined above. Unless stated otherwise specifically in the specification, a cyanoalkyl group is optionally substituted. Carbocyclic" or "carbocycle" refers to a ring system, wherein each of the ring atoms are carbon.
"Cycloalkyl" refers to a stable non-aromatic monocyclic or polycyclic carbocyclic radical consisting solely of carbon and hydrogen atoms, which may include fused or bridged ring systems, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond. Monocyclic radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Polycyclic radicals include, for example, adamantyl, norbornyl, decalinyl,
7,7-dimethyl-bicyclo[2.2.1]heptanyl, and the like. A "cycloalkylene" is a divalent cycloalkyl, which typically connects one portion a molecule to another portion. Unless otherwise stated specifically in the specification, a cycloalkyl (or cycloalkylene) group is optionally substituted.
"Cycloalkylalkyl" refers to a radical of the formula -¾¾ where ¾ is an alkylene chain as defined above and Rd is a cycloalkyl radical as defined above. Unless stated otherwise specifically in the specification, a cycloalkylalkyl group is optionally substituted.
"Cycloalkylaminyl" refers to a radical of the formula - RaRb wherein Ra is H or alkyl and R is cycloalkyl. Unless stated otherwise specifically in the specification, an heterocyclylalkoxy group is optionally substituted.
"Fused" refers to any ring structure described herein which is fused to an existing ring structure in the compounds of the invention. When the fused ring is a heterocyclyl ring or a heteroaryl ring, any carbon atom on the existing ring structure which becomes part of the fused heterocyclyl ring or the fused heteroaryl ring is replaced with a nitrogen atom.
"Halo" or "halogen" refers to bromo, chloro, fluoro or iodo.
"Haloalkyl" refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difluorom ethyl, trichlorom ethyl, 2,2,2-trifluoroethyl, 1,2-difluoroethyl, 3-bromo-2-fluoropropyl, 1,2-dibromoethyl, and the like. Unless stated otherwise specifically in the specification, a haloalkyl group is optionally substituted.
"Heterocyclyl" or "heterocyclic ring" refers to a stable 3- to 18-membered non-aromatic ring radical having one to twelve ring carbon atoms (e.g., two to twelve) and from one to six ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. Unless stated otherwise specifically in the specification, the heterocyclyl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused, spirocyclic ("spiro-heterocyclyl") and/or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl radical is optionally oxidized; the nitrogen atom is optionally quaternized; and the heterocyclyl radical is partially or fully saturated. Examples of such heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl[l,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl,
octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl,
2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl,
tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1, 1-dioxo-thiomorpholinyl "Heterocyclylene" refers to a bivalent heterocyclyl group, which typically connects one portion of a molecule to another. Unless stated otherwise specifically in the specification, a heterocyclyl and/or heterocyclylene group is optionally substituted.
"N-heterocyclyl" refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a nitrogen atom in the heterocyclyl radical. Unless stated otherwise specifically in the specification, a N-heterocyclyl group is optionally substituted.
"Heterocyclylalkyl" refers to a radical of the formula -RbRe where ¾, is an alkylene chain as defined above and Re is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl is optionally attached to the alkyl radical at the nitrogen atom. Unless stated otherwise specifically in the specification, a heterocyclylalkyl group is optionally substituted.
"Heterocyclylalkoxy" refers to a radical of the formula -ORaRb wherein Ra is alkylene and R is heterocyclyl. Unless stated otherwise specifically in the specification, an heterocyclylalkoxy group is optionally substituted.
"Heterocyclylaminyl" refers to a radical of the formula - RaRb wherein Ra is H or alkyl and Rb is heterocyclyl. Unless stated otherwise specifically in the specification, an heterocyclylalkoxy group is optionally substituted.
"Heteroaryl" refers to a 5- to 14-membered ring system radical comprising hydrogen atoms, one to thirteen ring carbon atoms, one to six ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and at least one aromatic ring. For purposes of this invention, the heteroaryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized.
Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzothiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl,
benzothiazolyl, benzothiadiazolyl, benzo[£][l,4]dioxepinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[l,2-a]pyridinyl, carbazolyl, cinnolinyl,
dibenzofuranyl, dibenzothiophenyl, furanyl, furanonyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1- oxidopyridinyl, 1-oxidopyrimidinyl, 1-oxidopyrazinyl, 1-oxidopyridazinyl,
1 -phenyl- lH-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinazolinyl, quinoxalinyl, quinolinyl, quinuclidinyl, isoquinolinyl,
tetrahydroquinolinyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, triazinyl, and thiophenyl (i.e. thienyl). Unless stated otherwise specifically in the specification, a heteroaryl is optionally substituted.
"N-heteroaryl" refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical. Unless stated otherwise specifically in the specification, an N-heteroaryl group is optionally substituted.
"Heteroarylalkyl" refers to a radical of the formula -¾Rf where R is an alkylene chain as defined above and Rf is a heteroaryl radical as defined above. Unless stated otherwise specifically in the specification, a heteroarylalkyl group is optionally substituted.
"Hydroxylalkyl" refers to an alkyl group comprising at least one hydroxyl substituent. The -OH substituent may be on a primary, secondary or tertiary carbon. Unless stated otherwise specifically in the specification, a hydroxylalkyl group is optionally substituted. Unless stated otherwise specifically in the specification, a hydroxylalkyl group is optionally substituted.
"Phosphate" refers to the -OP(=0)(Ra)Rb group, where Ra is OH, O" or OR; and R is OH, O", ORc, or a further phosphate group (e.g., to form a di- or triphosphate), wherein Re is a counter ion (e.g., Na+ and the like).
"Phosphoalkoxy" refers to an alkoxy group, as defined herein, which is substituted with at least one phosphate group, as defined herein. Unless stated otherwise specifically in the specification, an phosphoalkoxy group is optionally substituted.
"Thioalkyl" refers to a radical of the formula -SRa where Ra is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, a thioalkyl group may be optionally substituted.
The term "substituted" used herein means any of the above groups (e.g., alkyl, alkylene, alkoxy, alkoxyalkyl, alkoxycarbonyl, aminylalkyl, aminylalkylaminyl, aminylalkoxy, alkylaminylalkoxy, alkylaminyl, alkylcarbonylaminyl, alkylcarbonylaminylalkoxy, aminylcarbonylalkyl, thioalkyl, aryl, aralkyl, carboxyalkyl, cyanoalkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclyloxy, heterocyclylaminyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N- heteroaryl, heteroarylalkyl, phosphoalkoxy, and/or hydroxylalkyl) wherein at least one hydrogen atom (e.g., 1, 2, 3 or all hydrogen atoms) is replaced by a bond to a non- hydrogen atom such as, but not limited to: a halogen atom such as F, CI, Br, and I; an oxygen atom in groups such as hydroxyl groups, alkoxy groups, and ester groups; a sulfur atom in groups such as thiol groups, thioalkyl groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, imides, and enamines; a silicon atom in groups such as trialkylsilyl groups,
dialkylarylsilyl groups, alkyldiarylsilyl groups, and triarylsilyl groups; and other heteroatoms in various other groups. "Substituted" also means any of the above groups in which one or more hydrogen atoms are replaced by a higher-order bond (e.g., a double- or triple-bond) to a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and ester groups; and nitrogen in groups such as imines, oximes, hydrazones, and nitriles. For example, "substituted" includes any of the above groups in which one or more hydrogen atoms are replaced
with - RgRh, - RgC(=0)Rh, - RgC(=0) RgRh, - RgC(=0)ORh, - RgS02Rh,
-OC(=0) RgRh, -ORg, -SRg, -SORg, -S02Rg, -OS02Rg, -S02ORg, =NS02Rg, and -S02 RgRh. "Substituted also means any of the above groups in which one or more hydrogen atoms are replaced with -C(=0)Rg, -C(=0)ORg, -C(=0) RgRh, -CH2S02Rg, -CH2S02 RgRh. In the foregoing, Rg and Rh are the same or different and independently hydrogen, alkyl, alkoxy, alkylaminyl, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl. "Substituted" further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to an aminyl, cyano, hydroxyl, imino, nitro, oxo, thioxo, halo, alkyl, alkoxy, alkylaminyl, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl group. In addition, each of the foregoing substituents may also be optionally substituted with one or more of the above substituents.
"Electrophile" or "electrophilic moiety" is any moiety capable of reacting with a nucleophile (e.g., a moiety having a lone pair of electrons, a negative charge, a partial negative charge and/or an excess of electrons, for example a -SH group). Electrophiles typically are electron poor or comprise atoms which are electron poor. In certain embodiments an electrophile contains a positive charge or partial positive charge, has a resonance structure which contains a positive charge or partial positive charge or is a moiety in which delocalization or polarization of electrons results in one or more atom which contains a positive charge or partial positive charge. In some embodiments, the electrophiles comprise conjugated double bonds, for example an α,β-unsaturated carbonyl or α,β-unsaturated thiocarbonyl compound.
The term "effective amount" or "therapeutically effective amount" refers to that amount of a compound described herein that is sufficient to effect the intended application including but not limited to disease treatment, as defined below. The therapeutically effective amount may vary depending upon the intended treatment application (in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of
administration and the like, which can readily be determined by one of ordinary skill in the art. The term also applies to a dose that will induce a particular response in target cells, e.g. reduction of platelet adhesion and/or cell migration. The specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of
administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
As used herein, "treatment" or "treating" refer to an approach for obtaining beneficial or desired results with respect to a disease, disorder or medical condition including but not limited to a therapeutic benefit and/or a prophylactic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the subject,
notwithstanding that the subject may still be afflicted with the underlying disorder. In certain embodiments, for prophylactic benefit, the compositions are administered to a subject at risk of developing a particular disease, or to a subject reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
A "therapeutic effect," as that term is used herein, encompasses a therapeutic benefit and/or a prophylactic benefit as described above. A prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
The term "co-administration," "administered in combination with," and their grammatical equivalents, as used herein, encompass administration of two or more agents to an animal, including humans, so that both agents and/or their metabolites are present in the subject at the same time. Co-administration includes simultaneous administration in separate compositions, administration at different times in separate compositions, or administration in a composition in which both agents are present.
"Pharmaceutically acceptable salt" includes both acid and base addition salts.
"Pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, camphoric acid, camphor- 10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane- 1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, gluconic acid, glucuronic acid, glutamic acid, glutaric acid, 2-oxo-glutaric acid, glycerophosphoric acid, glycolic acid, hippuric acid, isobutyric acid, lactic acid, lactobionic acid, lauric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, mucic acid, naphthalene-l,5-disulfonic acid, naphthalene-2-sulfonic acid, l-hydroxy-2-naphthoic acid, nicotinic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, propionic acid, pyroglutamic acid, pyruvic acid, salicylic acid, 4-aminosalicylic acid, sebacic acid, stearic acid, succinic acid, tartaric acid, thiocyanic acid, ^-toluenesulfonic acid, trifluoroacetic acid, undecylenic acid, and the like.
"Pharmaceutically acceptable base addition salt" refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol,
2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like. Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine,
trimethylamine, dicyclohexylamine, choline and caffeine.
The terms "antagonist" and "inhibitor" are used interchangeably, and they refer to a compound having the ability to inhibit a biological function of a target protein, whether by inhibiting the activity or expression of the protein, such as KRAS, HRAS or RAS G12C. Accordingly, the terms "antagonist" and "inhibitors" are defined in the context of the biological role of the target protein. While preferred antagonists herein specifically interact with (e.g. bind to) the target, compounds that inhibit a biological activity of the target protein by interacting with other members of the signal transduction pathway of which the target protein is a member are also specifically included within this definition. A preferred biological activity inhibited by an antagonist is associated with the development, growth, or spread of a tumor.
The term "agonist" as used herein refers to a compound having the ability to initiate or enhance a biological function of a target protein, whether by inhibiting the activity or expression of the target protein. Accordingly, the term
"agonist" is defined in the context of the biological role of the target polypeptide.
While preferred agonists herein specifically interact with (e.g. bind to) the target, compounds that initiate or enhance a biological activity of the target polypeptide by interacting with other members of the signal transduction pathway of which the target polypeptide is a member are also specifically included within this definition.
As used herein, "agent" or "biologically active agent" refers to a biological, pharmaceutical, or chemical compound or other moiety. Non-limiting examples include a simple or complex organic or inorganic molecule, a peptide, a protein, an oligonucleotide, an antibody, an antibody derivative, antibody fragment, a vitamin derivative, a carbohydrate, a toxin, or a chemotherapeutic compound. Various compounds can be synthesized, for example, small molecules and oligomers (e.g., oligopeptides and oligonucleotides), and synthetic organic compounds based on various core structures. In addition, various natural sources can provide compounds for screening, such as plant or animal extracts, and the like.
"Signal transduction" is a process during which stimulatory or inhibitory signals are transmitted into and within a cell to elicit an intracellular response. A modulator of a signal transduction pathway refers to a compound which modulates the activity of one or more cellular proteins mapped to the same specific signal transduction pathway. A modulator may augment (agonist) or suppress (antagonist) the activity of a signaling molecule.
An "anti-cancer agent", "anti-tumor agent" or "chemotherapeutic agent" refers to any agent useful in the treatment of a neoplastic condition. One class of anti- cancer agents comprises chemotherapeutic agents. "Chemotherapy" means the administration of one or more chemotherapeutic drugs and/or other agents to a cancer patient by various methods, including intravenous, oral, intramuscular, intraperitoneal, intravesical, subcutaneous, transdermal, buccal, or inhalation or in the form of a suppository.
The term "cell proliferation" refers to a phenomenon by which the cell number has changed as a result of division. This term also encompasses cell growth by which the cell morphology has changed (e.g., increased in size) consistent with a proliferative signal.
The term "selective inhibition" or "selectively inhibit" refers to a biologically active agent refers to the agent's ability to preferentially reduce the target signaling activity as compared to off-target signaling activity, via direct or indirect interaction with the target.
"Subject" refers to an animal, such as a mammal, for example a human. The methods described herein can be useful in both human therapeutics and veterinary applications. In some embodiments, the subject is a mammal, and in some
embodiments, the subject is human.
"Mammal" includes humans and both domestic animals such as laboratory animals and household pets (e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like.
"Radiation therapy" means exposing a subject, using routine methods and compositions known to the practitioner, to radiation emitters such as alpha-particle emitting radionuclides (e.g., actinium and thorium radionuclides), low linear energy transfer (LET) radiation emitters (i.e. beta emitters), conversion electron emitters (e.g. strontium-89 and samarium-153-EDTMP, or high-energy radiation, including without limitation x-rays, gamma rays, and neutrons. An "anti-cancer agent", "anti-tumor agent" or "chemotherapeutic agent" refers to any agent useful in the treatment of a neoplastic condition. One class of anticancer agents comprises chemotherapeutic agents. "Chemotherapy" means the administration of one or more chemotherapeutic drugs and/or other agents to a cancer patient by various methods, including intravenous, oral, intramuscular, intraperitoneal, intravesical, subcutaneous, transdermal, buccal, or inhalation or in the form of a suppository.
"Prodrug" is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound described herein (e.g., compound of structure (I)). Thus, the term "prodrug" refers to a precursor of a biologically active compound that is pharmaceutically acceptable. In some aspects, a prodrug is inactive when administered to a subject, but is converted in vivo to an active compound, for example, by hydrolysis. The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam). A discussion of prodrugs is provided in Higuchi, T., et al., "Pro-drugs as Novel Delivery Systems," A.C.S. Symposium Series, Vol. 14, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein. The term "prodrug" is also meant to include any covalently bonded carriers, which release the active compound in vivo when such prodrug is administered to a
mammalian subject. Prodrugs of an active compound, as described herein, are typically prepared by modifying functional groups present in the active compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent active compound. Prodrugs include compounds wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the active compound is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of a hydroxy functional group, or acetamide, formamide and benzamide derivatives of an amine functional group in the active compound and the like.
In some embodiments, prodrugs include compounds of structure (I) having a phosphate, phosphoalkoxy, ester or boronic ester substituent. Without being bound by theory, it is believed that such substituents are converted to a hydroxyl group under physiological conditions. Accordingly, embodiments include any of the compounds disclosed herein, wherein a hydroxyl group has been replaced with a phosphate, phosphoalkoxy, ester or boronic ester group, for example a phosphate or phosphoalkoxy group. For example, in some embodiments a hydroxyl group on the R1 moiety is replaced with a phosphate, phosphoalkoxy, ester or boronic ester group, for example a phosphate or alkoxy phosphate group. Exemplary prodrugs of certain embodiments thus include com ounds having one of the following R1 moieties:
wherein each R' is independently H or an optional substituent, and n is 1, 2, 3 or 4.
The term "in vivo" refers to an event that takes place in a subject's body. Embodiments of the invention disclosed herein are also meant to encompass all pharmaceutically acceptable compounds of structure (I) being
isotopically-labelled by having one or more atoms replaced by an atom having a different atomic mass or mass number. Examples of isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2H, 3H, UC, 13C, 14C, 13N, 15N, 150, 170, 180, 31P, 32P, 35S, 18F, 36C1, 123I, and 125I, respectively. These radiolabeled compounds could be useful to help determine or measure the effectiveness of the compounds, by characterizing, for example, the site or mode of action, or binding affinity to pharmacologically important site of action. Certain isotopically-labeled compounds of structure (I), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.
Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence are preferred in some circumstances.
Substitution with positron emitting isotopes, such as UC, 18F, 150 and
13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds of structure (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Preparations and Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
Certain embodiments are also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result from, for example, the oxidation, reduction, hydrolysis, amidation, esterification, and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the embodiments include compounds produced by a process comprising administering a compound of this invention to a mammal for a period of time sufficient to yield a metabolic product thereof. Such products are typically identified by administering a radiolabeled compound of the invention in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to human, allowing sufficient time for metabolism to occur, and isolating its conversion products from the urine, blood or other biological samples.
"Stable compound" and "stable structure" are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent. Often crystallizations produce a solvate of the compound of the invention. As used herein, the term "solvate" refers to an aggregate that comprises one or more molecules of a compound of the invention with one or more molecules of solvent. In some embodiments, the solvent is water, in which case the solvate is a hydrate. Alternatively, in other embodiments, the solvent is an organic solvent. Thus, the compounds of the present invention may exist as a hydrate, including a
monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding solvated forms. In some aspects, the compound of the invention is a true solvate, while in other cases, the compound of the invention merely retains adventitious water or is a mixture of water plus some adventitious solvent.
"Optional" or "optionally" means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example, "optionally substituted aryl" means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
A "pharmaceutical composition" refers to a formulation of a compound of the invention and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g. , humans. Such a medium includes all pharmaceutically acceptable carriers, diluents or excipients therefor.
"Pharmaceutically acceptable carrier, diluent or excipient" includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
The compounds of the invention (i.e., compounds of structure (I)), or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that are defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids. Embodiments thus include all such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (-), (R)- and (5)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). When the compounds described herein contain olefinic double bonds or other centres of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included.
Embodiments of the present invention include all manner of rotamers and conformationally restricted states of a compound of the invention. Atropisomers, which are stereoisomers arising because of hindered rotation about a single bond, where energy differences due to steric strain or other contributors create a barrier to rotation that is high enough to allow for isolation of individual conformers, are also included. As an example, certain compounds of the invention may exist as mixtures of atropisomers or purified or enriched for the presence of one atropisomer. Non-limiting examples of compounds which exist as atropisomers include the following compounds:
S-atropisomer In some embodiments, the compound of structure (I) is a mixture of atropisomers. In other embodiments, the compound of structure (I) is a substantially purified atropisomer. In some embodiments, the compound of structure (I) is a substantially purified R-atropisomer. In some other embodiments, the compound of structure (I) is a substantially purified R-atropisomer.
A "stereoisomer" refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present invention contemplates various stereoisomers and mixtures thereof and includes "enantiomers", which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another.
A "tautomer" refers to a proton shift from one atom of a molecule to another atom of the same molecule. Embodiments thus include tautomers of the disclosed compounds.
The chemical naming protocol and structure diagrams used herein are a modified form of the I.U.P.A.C. nomenclature system, using the ACD/Name Version 9.07 software program and/or ChemDraw Ultra Version 11.0.1 software naming program (CambridgeSoft). For complex chemical names employed herein, a substituent group is typically named before the group to which it attaches. For example, cyclopropylethyl comprises an ethyl backbone with a cyclopropyl substituent. Except as described below, all bonds are identified in the chemical structure diagrams herein, except for all bonds on some carbon atoms, which are assumed to be bonded to sufficient hydrogen atoms to complete the valency.
Compounds
In an aspect, the invention provides compounds which are capable of selectively binding to and/or modulating a G12C mutant KRAS, HRAS or RAS protein. The compounds may modulate the G12C mutant KRAS, HRAS or NRAS protein by reaction with an amino acid. While not wishing to be bound by theory, the present applicants believe that, in some embodiments, the compounds of the invention selectively react with the G12C mutant KRAS, HRAS or NRAS proteins by forming a covalent bond with the cysteine at the 12 position of a G12C mutant KRAS, HRAS or NRAS protein. By binding to the Cysteine 12, the compounds of the invention may lock the switch II of the G12C mutant KRAS, HRAS or NRAS into an inactive stage. This inactive stage may be distinct from those observed for GTP and GDP bound KRAS, HRAS or NRAS. Some compounds of the invention may also be able to perturb the switch I conformation. Some compounds of the invention may favor the binding of the bound KRAS, HRAS or NRAS to GDP rather than GTP and therefore sequester the KRAS, HRAS or NRAS into an inactive KRAS, HRAS or NRAS GDP state. Because effector binding to KRAS, HRAS or NRAS is highly sensitive to the conformation of switch I and II, the irreversible binding of these compounds may disrupt KRAS, HRAS or NRAS downstream signaling.
As noted above, in one embodiment of the present invention, compounds having activity as modulators of a G12C mutant KRAS, HRAS or NRAS protein are provided, the compounds have the followin structure (I):
(I)
or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof, wherein:
A is N or C;
B is oxo, cyano, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, heteroaryl, cycloalkylalkyl, heterocycloalkyl, heteroarylalkyl, amino, alkylamino, arylamino, -C02H, -CONH2, aminylcarbonyl, aminylcarbonylalkyl, heteroarylamino, halo, haloalkyl, alkoxy, haloalkoxy, aryl or -X-L2-Ra;
X is -NRb- or -0-;
L1 is alkylene, cycloalkylene, heterocyclylene or absent;
L2 is alkylene or absent;
R is H, cyano, amino, halo, haloalkyl, hydroxyl, cycloalkyl, heterocyclyl, heterocycloalkyl, aryl, heteroaryl, -C02H, -CONH2; aminylcarbonyl, Ci C6 alkyl, Ci-C6 alkylaminyl or Ci-C6 alkoxy; Ra is cycloalkyl, heterocyclyl, heteroaryl, -(C=0)OH, -(C=0) H2 or -(C=0) HOH;
Rb is, at each occurrence, independently H or Ci-C6 alkyl;
R1 is aryl or heteroaryl;
R2a, R2b and R2c are each independently H, amino, cyano, halo, hydroxyl, Ci-C6 alkyl, Ci-C6 alkylaminyl, -NRb(C=0)Rb, Ci-C6 haloalkyl, Ci-C6 alkoxy, C3-C8 cycloalkyl, heterocyclylalkyl, C2-C6 alkynyl, C2-C6 alkenyl, aminylalkyl,
alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aminylcarbonyl, heteroaryl or aryl;
~ is a single or double bond such that all valences are satisfied; and
E is an electrophilic moiety capable of forming a covalent bond with the cysteine residue at position 12 of a KRAS, HRAS or NRAS G12C mutant protein.
In other embodiments of the compound of structure (I):
A is N or C;
B is oxo, alkyl, cycloalkyl, heterocyclyl, heteroaryl, cycloalkylalkyl, heterocycloalkyl, heteroarylalkyl, amino, alkylamino, arylamino, -C02H, -CONH2, aminylcarbonyl, aminylcarbonylalkyl, heteroarylamino, halo, haloalkyl, alkoxy, haloalkoxy, aryl or -X-L2-Ra;
X is -NRb- or -0-;
L1 is alkylene, cycloalkylene, heterocyclylene or absent;
L2 is alkylene or absent;
R is H, cyano, amino, halo, haloalkyl, hydroxyl, cycloalkyl, heterocyclyl, heterocycloalkyl, aryl, heteroaryl, -C02H, -CONH2, aminylcarbonyl, Ci- C6 alkyl, Ci-C6 alkylaminyl or Ci-C6 alkoxy;
Ra is cycloalkyl, heterocyclyl, heteroaryl, -(C=0)OH, -(C=0)NH2 or -(C=0)NHOH;
Rb is, at each occurrence, independently H or Ci-C6 alkyl;
R1 is aryl or heteroaryl;
R2a, R2b and R2c are each independently H, amino, halo, hydroxyl, Ci-C6 alkyl, Ci-C6 alkylaminyl, -NRb(C=0)Rb, Ci-C6 haloalkyl, Ci-C6 alkoxy, C3-C8 cycloalkyl, heterocyclylalkyl, C2-C6 alkynyl, C2-C6 alkenyl, aminylalkyl,
alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aminylcarbonyl, heteroaryl or aryl;
~ is a single or double bond such that all valences are satisfied; and E is an electrophilic moiety capable of forming a covalent bond with the cysteine residue at position 12 of a KRAS, HRAS or NRAS G12C mutant protein.
In other different embodiments of the compound of structure (I):
A is N or C;
B is oxo, alkyl, cycloalkyl, heterocyclyl, heteroaryl, cycloalkylalkyl, heterocycloalkyl, heteroarylalkyl or -X-L2-Ra;
X is -NRb- or -0-;
L1 is alkylene, cycloalkylene, heterocyclylene or absent;
L2 is alkylene or absent;
R is H, cyano, amino, Ci-C6 alkyl, Ci-C6 alkylaminyl or Ci-C6 alkoxy; Ra is cycloalkyl, heterocyclyl, heteroaryl, -(C=0)OH, -(C=0)NH2 or -(C=0)NHOH;
Rb is H or Ci-C6 alkyl;
R1 is aryl or heteroaryl;
R2a, R2b and R2c are each independently H, amino, halo, hydroxyl, Ci-C6 alkyl, Ci-C6 alkylaminyl, -NRb(C=0)Rb, Ci-C6 haloalkyl, Ci-C6 alkoxy, C3-C8 cycloalkyl, heteroaryl or aryl;
~ is a single or double bond such that all valences are satisfied; and
E is an electrophilic moiety capable of forming a covalent bond with the cysteine residue at position 12 of a KRAS, HRAS or NRAS G12C mutant protein.
In some embodiments, B is cycloalkyl, heterocyclyl or heteroaryl. In other embodiments, L1 is alkylene or absent.
In some other of the foregoing embodiments, B is cycloalkyl or heterocyclyl, for example in some embodiments the compound has the following structure (IA):
(IA)
wherein:
G1 is N or CH;
G2 is RC or CHRC;
Rc is H, alkyl, alkylcarbonyl, aminocarbonyl, alkylcarbonylaminyl, aminocarbonylaminyl or heteroarylcarbonyl;
R a and R are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R3a and R3b join to form oxo a carbocyclic or heterocyclic ring; or R3a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R3b joins with R4b to form a carbocyclic or heterocyclic ring;
R a and R are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C2- C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R4a and R4b join to form oxo, a carbocyclic or heterocyclic ring; or R4a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R4b joins with R3b to form a carbocyclic or heterocyclic ring;
m1 and m2 are each independently 1, 2 or 3; and
n is an integer from 0 to 5.
In other embodiments of structure (IA): R a and R are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl,
aminylcarbonylalkyl or aminylcarbonyl; or R3a and R3b join to form a carbocyclic or heterocyclic ring; or R3a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R3b joins with R4b to form a carbocyclic or heterocyclic ring;
R a and R are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl,
aminylcarbonylalkyl or aminylcarbonyl; or R4a and R4b join to form a carbocyclic or heterocyclic ring; or R4a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R4b joins with R3b to form carbocyclic or heterocyclic ring;
In other embodiments, the compound has the following structure (IAa) or (IAb):
(IAa) (IAb)
wherein p is an integer from 0 to 3.
In still different embodiments, the compound has one of the following structures (I Ac), (IAd) or (IAe):
(IAc) (IAd) (IAe)
In other of the foregoing embodiments, Rc is alkylcarbonyl, aminocarbonyl, alkylcarbonylaminyl, aminocarbonylaminyl or heteroarylcarbonyl, for example, in some embodiments the alkylcarbonyl is substituted with aminocarbonyl, hydroxylaminocarbonyl, hydroxyl or amino.
In other embodiments, Rc has one of the following structures:
wherein p2 is an integer from 1 to 3.
In other embodiments, Rc has one of the following structures:
In some different embodiments, B is alkyl, cycloalkylalkyl, heterocycloalkyl, heteroarylalkyl or -X-L2-Ra. For example, in some embodiments B is -X-L2-Ra. In other embodiments, Ra is heterocyclyl or heteroaryl.
In various other embodiments, L1 is alkylene or absent. In some embodiments, L1 is alkylene. In other embodiments, L1 is absent.
In some embodiments, B is -X-L2-Ra and L1 is alkylene or absent, for example in some embodiments the compound has one of the following structures (IB) or (IC):
wherein:
H represents a 5 or 6-membered heteroaryl ring optionally substituted with one or more of R3a, R3b, R4a and R4b;
G1 is N or CH;
G2 is RC or CHRC;
Rc is H, alkyl, alkylcarbonyl, aminocarbonyl, alkylcarbonylaminyl, aminocarbonylaminyl or heteroaryl carbonyl;
R3a and R3b are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R3a and R3b join to form oxo a carbocyclic or heterocyclic ring; or R3a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R3b joins with R4b to form a carbocyclic or heterocyclic ring;
R4a and R4b are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R4a and R4b join to form oxo a carbocyclic or heterocyclic ring; or R4a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R4b joins with R3b to form a carbocyclic or heterocyclic ring;
m1 and m2 are each independently 1, 2 or 3; and n is an integer from 0 to 5.
In other embodiments of structure (IB) and (IC):
R3a and R3b are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl,
aminylcarbonylalkyl or aminylcarbonyl; or R3a and R3b join to form a carbocyclic or heterocyclic ring; or R3a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R3b joins with R4b to form a carbocyclic or heterocyclic ring;
R a and R are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl,
aminylcarbonylalkyl or aminylcarbonyl; or R4a and R4b join to form a carbocyclic or heterocyclic ring; or R4a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R4b joins with R3b to form a carbocyclic or heterocyclic ring;
In some embodiments, H is pyrrolidinyl or pyridinyl.
In other embodiments, the compound has one of the following structures
(IBa) or (IBb):
(IBa) (IBb)
wherein Rd is, at each occurrence, independently H, halo or hydroxyl, and p3 is an integer from 0 to 3. In still different embodiments, the compound has one of the following structures (IBc
(IBf)
In some of the foregoing embodiments, X is - H-. In some of other the foregoing embodiments, X is -0-.
In still other embodiments B has one of the following structures:
In other embodiments, B is -X-L2-Ra, and the compound has the following structure (ID):
(ID)
wherein p is an integer from
In embodiments of the foregoing, Ra is , -(C=0)OH, -(C=0) H2 or -(C=0) HOH.
In still different embodiments, the compound has the following structure
(pa):
(IDa)
In some embodiments of compounds (ID) or (IDa) X is - H-. In some other embodiments of compounds (ID) or (IDa) X is -0-.
In some of the foregoing embodiments, B is alkyl, for example in some embodiments the alkyl is substituted with -(C=0)OH, -(C=0) H2 or -(C=0) HOH.
In other different embodiments, B is heteroarylalkyl. In some of these embodiments, the heteroarylalkyl is pyrrolidinylalkyl or pyridinylalkyl.
In still other embodiments, B is oxo. In other embodiments, L1 is heterocyclylene. In further embodiments, B is oxo, and L1 is heterocyclylene. For example, in some embodiments the compound has the following structure (IE):
(IE) wherein:
G1 is CH;
G2 is N or CH;
R3a and R3b are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R3a and R3b join to form oxo a carbocyclic or heterocyclic ring; or R3a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R3b joins with R4b to form a carbocyclic or heterocyclic ring;
R4a and R4b are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R4a and R4b join to form oxo a carbocyclic or heterocyclic ring; or R4a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R4b joins with R3b to form a carbocyclic or heterocyclic ring; and m1 and m2 are each independently 1, 2 or 3.
In other embodiments of the compound of structure (IE):
R3a and R3b are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl,
aminylcarbonylalkyl or aminylcarbonyl; or R3a and R3b join to form a carbocyclic or heterocyclic ring; or R3a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R3b joins with R4b to form carbocyclic or heterocyclic ring; R4a and R4b are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl,
aminylcarbonylalkyl or aminylcarbonyl; or R4a and R4b join to form a carbocyclic or heterocyclic ring; or R4a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R4b joins with R3b to form a carbocyclic or heterocyclic ring;
In other embodiments, the compound has one of the following structures (IEa), (IEb), (IEc) or (IEd) :
(IEa) (IEb) (IEc)
(IEd)
In some different embodiments, the compound has one of the following structures IEe), (IEf), (IEg), (IEh (IEi) of (IEj):
IEh) IEi)
(IEj) (IEk)
In some different embodiments, the compound has the following structure (IEI):
(IEI)
Without wishing to be bound by theory, Applicants believe correct selection of the R1 substituent may play a part in the compounds' inhibitory activity (e.g., against KRAS, HRAS or NRAS G12C). In some embodiments, R1 is aryl or heterocyclyl (e.g., heteroaryl or aliphatic heterocyclyl), each of which is optionally substituted with one or more substituents. In some other embodiments, R1 is aryl or heteroaryl. In some embodiments, R1 is capable of reversible interaction with KRAS, HRAS or NRAS G12C mutant protein. In some embodiments R1 has high affinity towards KRAS, HRAS or NRAS and is highly specific towards G12C KRAS, HRAS or NRAS. In some embodiments R1 is capable of hydrophobic interaction with KRAS, HRAS or NRAS G12C. In some embodiments R1 is able to form hydrogen bonds with various residues of G12C KRAS, HRAS or NRAS protein.
In any of the foregoing embodiments, R1 is aryl. For example in some embodiments R1 is phenyl, and in other embodiments R1 is naphthyl. R1 is substituted or unsubstituted. In some specific embodiments, R1 is substituted with one or more substituents. In some embodiments, R1 is substituted with halo, amino, hydroxyl, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, cyano, Ci-C6 haloalkyl, Ci-C6 alkoxy, alkylaminyl, cycloalkyl, heterocyclylalkyl, heterocyclylalkoxy, heterocyclylaminyl,
cycloalkylaminyl, aryl, heteroaryl, phosphate, phosphoalkoxy, boronic acid, boronic acid ester, -OC(=0)R or Ci-C6 alkylcarbonyloxy, or combinations thereof, wherein R is Ci-C6 alkyl. For example, in some embodiments R1 is substituted with halo, amino, hydroxyl, Ci-C6 alkyl, cyano, Ci-C6 haloalkyl, Ci-C6 alkoxy, alkylaminyl, cycloalkyl, heterocyclylalkyl, aryl, heteroaryl, phosphate, phosphoalkoxy, boronic acid, boronic acid ester, -OC(=0)R or Ci-C6 alkylcarbonyloxy, or combinations thereof, wherein R is Ci-C6 alky. In other embodiments, R1 is substituted with halo, hydroxyl, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 alkoxy or Ci-C6 alkylcarbonyloxy, or combinations thereof. In different embodiments, R1 is substituted with fluoro, chloro, cyclopropyl, cyclobutyl, hydroxyl, amino, methyl, ethyl, isopropyl, trifluoromethyl or methoxy, or combinations thereof. In some even more embodiments, R1 is substituted with fluoro, hydroxyl, methyl, isopropyl, trifluoromethyl or methoxy, or combinations thereof.
In some more specific embodiments, R1 has one of the following structures:
In some different embodiments of the foregoing compounds, R1 is heteroaryl, for example a heteroaryl comprising nitrogen. In other embodiments, R1 is indazolyl or quinolinyl. In more embodiments, R1 is heteroaryl which is substituted with one or more substituents. For example, in certain embodiments, R1 is substituted with hydroxyl or Ci-C6 alkyl, or both.
In some other embodiments, R1 has one of the following structures: , for example:
In some of the foregoing embodiments R c is H. In other of any of the foregoing embodiments, R2a and R2b are each halo. For example, in some embodiments R2a is fiuoro, and in other embodiments, R2b is chloro.
In other embodiments, R2a and R2b are each independently halo, haloalkyl, alkyl, amino, hydroxyl or alkoxy. In other embodiments, R2a and R2b are each independently halo, haloalkyl, alkyl or alkoxy. In some embodiments, R2a is fluoro, chloro, CF3 or methoxy. In some embodiments, R2a is fluoro, chloro or methoxy. In different embodiments, R2b is chloro, fluoro, amino, hydroxyl or CF3. In still other different embodiments, R2b is chloro, fluoro or CF3.
In some more specific embodiments, the compounds have the following structure (IF):
(I'f)
For example, in even further different embodiments, the compounds of the following structures (IF a) or (IFb):
(IFa) (IFb)
In any of the foregoing embodiments, R is H.
In some other of the foregoing embodiments,
that all valences are satisfied. In other different embodiments, ~ is a double bond such that all valences are satisfied.
In some embodiments, A is N. In other embodiments, A is C.
The structure of E in all of the embodiments described herein is not particularly limited provided it is capable of forming a covalent bond with a
nucleophile, such as the cysteine residue at position 12 of a KRAS, HRAS or RAS
G12C mutant protein. Accordingly, E moieties which are capable of reaction with (e.g., by covalent bond formation) a nucleophile are preferred. In certain embodiments, E is capable of reacting in a conjugate addition manner (e.g., 1,4-conjugate addition) with an appropriately reactive nucleophile. In some embodiments, E comprises conjugated pi bonds such that derealization of electrons results in at least one atom (e.g., a carbon atom) having a positive charge, partial positive charge or a polarized bond. In other embodiments, E comprises one or more bonds wherein the electronegativity of the two atoms forming the bonds is sufficiently different such that a partial positive charge (e.g., by polarization of the bond) resides on one of the atoms, for example on a carbon atom. E moieties comprising carbon-halogen bonds, carbon-oxygen bonds or carbon bonds to various leaving groups known in the art are examples of such E moieties.
Accordingly, in any of the foregoing embodiments, E has the following structure:
wherein:
-C(=0)-, -C(= R8')-, - R8C(=0)-, -S(=0)2- or - R8S(=0)2- R is H, Ci-C6alkyl or hydroxylalkyl;
R8 is H, -OH, -CN or Ci-C6alkyl; and
R9 and R10 are each independently H, halo, cyano, carboxyl, Ci-C6 alkyl, alkoxycarbonyl, aminylalkyl, alkylaminylalkyl, aryl, heterocyclyl, heterocyclylalkyl, heteroaryl or hydroxylalkyl, or R9 and R10 join to form a carbocyclic, heterocyclic or heteroaryl ring.
In still other of any of the foregoing embodiments, E has the following structure:
wherein:
Q is -C(=0)-, -NR8C(=0)-, -S(=0)2- or - NR8S(=0)2-;
R8 is H, Ci-C6alkyl or hydroxylalkyl; and
R10 is H, Ci-C6alkyl, aminylalkyl, alkylaminylalkyl or hydroxylalkyl.
The Q moiety is typically selected to optimize the reactivity (i.e., electrophilicity) of E. In some of the foregoing embodiments Q is -C(=0)-,- NR8C(=0)-,
-S(=0)2- or - NR8S(=0)2-. In certain of the foregoing embodiments, Q is -C(=0)-. In other embodiments, Q is -S(=0)2-. In still more embodiments, Q is -NR8C(=0)-. In still more different embodiments, Q is - NR8S(=0)2-.
In some other of the foregoing embodiments, Q is -C(=NR8 )-, wherein R8 is H, -OH, -CN or Ci-C6alkyl. For example, in some embodiments R8 is H. In other embodiments, R8 is -CN. In other embodiments, R8 is -OH.
In some of the foregoing embodiments, R8 is H. In other of these embodiments, R8 is hydroxylalkyl, for example in some embodiments the hydroxylalkyl is 2-hydroxylalkyl.
In some of any one of the foregoing embodiments, at least one of R9 or R10 is H. For example, in some embodiments each of R9 and R10 are H.
In other of the foregoing embodiments, R10 is alkylaminylalkyl. In some of these embodiments, R10 has the following structure:
In other embodiments, R10 is hydroxylalkyl, such as 2-hydroxylalkyl.
In some other different embodiments of the foregoing embodiments, R9 and R10 join to form a carbocyclic ring. For example, in some of these embodiments the carbocyclic ring is a cyclopentene, cyclohexene or phenyl ring. In other embodiments, the carbocyclic ring is a cyclopentene or cyclohexene ring. In other embodiments, the carbocyclic ring is a phenyl ring, for example a phenyl ring having the following structure: In some of any of the foregoing embodiments E is an electrophile capable of bonding with a KRAS, HRAS or NRAS protein comprising G12C mutation. In some embodiments, the electrophile E is capable of forming an irreversible covalent bond with a G12C mutant KRAS, HRAS or NRAS protein. In some cases, the electrophile E may bind with the cysteine residue at the position 12 of a G12C mutant KRAS, HRAS or NRAS protein. In various embodiments of any of the foregoing, E has one of the followin structures:
In some embodiments, E has one of the following structures:
In different embodiments, E has one of the following structures:
In some cases E has one of the following structures:
wherein:
R is H or Ci-Cealkyl;
R9 is H, cyano or Ci-C6alkyl, or R9 joins with R10 to form a carbocycle; R10 is H or Ci-C6alkyl or R10 joins with R9 to form a carbocycle and R10a is H or Ci-C6alkyl.
O . In some embodiments E is
In certain embodiments, R a and R are, at each occurrence, independently H, -OH, - H2, -C02H, halo, cyano, hydroxylalkly, aminylalkyl, cyanoalkyl, carboxyalkyl or aminylcarbonyl, and R4a and R4b are, at each occurrence, independently H, -OH, - H2, -C02H, halo, cyano, hydroxylalkly, aminylalkyl, cyanoalkyl, carboxyalkyl or aminylcarbonyl.
In other of the foregoing embodiments, R3a and R4a are, at each occurrence, independently H, -OH, hydroxylalkly, cyano, or aminylcarbonyl and R3b and R4b are H. In certain other embodiments, R a and R a are H and R and R are, at each occurrence, independently H, -OH, - H2, -C02H, halo, cyano, hydroxylalkly, aminylalkyl, cyanoalkyl, carboxyalkyl or aminylcarbonyl.
In any of the foregoing embodiments, at least one of R3a, R3b, R4a or R4b is H. In some embodiments, each of R3a, R3b, R4a and R4b are H.
In other of the foregoing embodiments, R3a and R4a are, at each occurrence, independently H or Ci-C6 alkyl, provided at least one of R3a or R4a is Ci-C6 alkyl. In some embodiments, at least one of R3a, R4a, R3b and R4b is independently Ci- C6 alkyl, such as methyl. In some embodiments, one occurrence of R3a is Ci-C6 alkyl, such as methyl, and the remaining R3a and each R4a is H. In some other embodiments, two occurrences of R3a are Ci-C6 alkyl, such as methyl, and the remaining R3a and each R4a is H. In some other embodiments, one occurrence of R3a and one occurrence of R4a is independently Ci-C6 alkyl, such as methyl, and the remaining R3a and R4a are each H.
In some embodiments, R3a is -OH, - H2, -C02H, halo, cyano, hydroxylalkly, aminylalkyl, cyanoalkyl, carboxyalkyl or aminylcarbonyl, and R3b, R4a and R4b are H.
In other embodiments, R4a is -OH, - H2, -C02H, halo, cyano, hydroxylalkly, aminylalkyl, cyanoalkyl, carboxyalkyl or aminylcarbonyl, and R3a, R3b and R4b are H.
In other embodiments, R3a is H, -OH, - H2, -C02H, halo, cyano, hydroxylalkly, aminylalkyl, cyanoalkyl, carboxyalkyl or aminylcarbonyl, and R3b joins with R4b to form a carbocyclic or heterocyclic ring;
In still more embodiments, R4a is H, -OH, - H2, -C02H, halo, cyano, hydroxylalkly, aminylalkyl, cyanoalkyl, carboxyalkyl or aminylcarbonyl, and R4b joins with R3b to form a carbocyclic or heterocyclic ring.
In other embodiments, R3a and R3b join to form a carbocyclic or heterocyclic ring. In other embodiments, R4a and R4b join to form a carbocyclic or heterocyclic ring. In still other embodiments, R a or R a is aminylcarbonyl. For example,
O in certain embodiments, the aminylcarbonyl is . In other embodiments, R3a or
R4a is cyano. In other embodiments, R3a or R4a is -OH. In other embodiments, R3a or R4a is hydroxylalkyl, for example hydroxylmethyl.
In some embodiments of any of the foregoing compounds, R1 is aryl or heteroaryl and R2a, R2b and R2c are independently selected from H and halo, for example in some further embodiments R1 is aryl or heteroaryl and R2a and R2b are independently selected from halo, such as chloro and fluoro, and R2c is H. In some embodiments, R1 is aryl or heteroaryl, R2a is chloro, R2b is fluoro and R2c is H. In other embodiments R1 is aryl or heteroaryl, one of R2a or R2b is halo, such as chloro or fluoro, and the other one of R2a or R2b is H.
In some embodiments 1. In other embodiments m1 is 2. In still more embodiments, m1 is 3. In different embodiments, m2 is 1. In some other embodiments, m2 is 2. In yet still more embodiments, m2 is 3.
In some other particular embodiments of any of the foregoing compounds, 1, and m2 is 1. In other embodiments, m1 is 1 and, m2 is 2. In still other embodiments and m2 is 2. In more embodiments, 1, and m2 is 3.
Some embodiments of the compounds include more than one
stereoisomer. Other embodiments are directed to a single stereoisomer. In some embodiments the compounds are racemic (e.g., mixture of atropisomers), while in other embodiments the compounds are substantially a single isomer, for example a substantially purified atropisomer.
In various different embodiments, the compound has one of the structures set forth in Table 1 below. The compounds in Table 1 were each prepared and analyzed by mass spectrometry and/or 1H MR. Experimental mass spectrometry data is included in Table 1. Exemplary synthetic procedures are described in more detail below and in the Examples. General methods by which the compounds may be prepared are provided below and indicated in Table 1. Table 1
Representative Compounds
4(3H)-one No. Structure Name Method [M+H]+
0
3-(l- acryloylazetidin-3- yl)-7-(3-
17 A 398.1 hydroxynaphthale
n-l-yl)quinazolin- 4(3H)-one
It is understood that in the present description, combinations of substituents and/or variables of the depicted formulae are permissible only if such contributions result in stable compounds.
Furthermore, all compounds of the invention which exist in free base or acid form can be converted to their pharmaceutically acceptable salts by treatment with the appropriate inorganic or organic base or acid by methods known to one skilled in the art. Salts of the compounds of the invention can be converted to their free base or acid form by standard techniques.
The following General Reaction Schemes illustrate exemplary methods of making compounds of compounds of structure I):
(I)
or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof, wherein R, R1,
R2a R2b R2c A B Ll E
are as defined herein. It is understood that one skilled in the art may be able to make these compounds by similar methods or by combining other methods known to one skilled in the art. It is also understood that one skilled in the art would be able to make, in a similar manner as described below, other compounds of structure (I) not specifically illustrated below by using the appropriate starting components and modifying the parameters of the synthesis as needed. In general, starting components may be obtained from sources such as Sigma Aldrich, Lancaster Synthesis, Inc., Maybridge, Matrix Scientific, TCI, and Fluorochem USA, etc. or synthesized according to sources known to those skilled in the art (see, for example, Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5th edition (Wiley, December 2000)) or prepared as described in this invention.
General Reaction Scheme 1
Embodiments of the compound of structure (I) (e.g., compound A-6) can be prepared according to General Reaction Scheme 1 ("Method A"), wherein R1, R2a, R2b, R2c, R3a, R3b, R4a, R4b, R9, R10, Q, m1 and m2 are as defined herein. Referring to General Reaction Scheme 1, compounds of structure A-1 are purchased from commercial sources or prepared according to techniques known in the art. The desired R1 substituent added to A-1 by way of Suzuki coupling to yield A-2. Cyclization of A- 2 with a reagent such as formamidine acetate provides quinazolinone A-3. Treatment of A-3 with an appropriately substituted cyclic group and an activating agent, such as HATU, provides A-4 which can then be deprotected by treatment with acid. The "E" moiety is then installed under conditions known in the art to yield A-6.
General Reaction Scheme 2
Embodiments of the compound of structure (I) (e.g., compound B-3) can be prepared according to General Reaction Scheme 2 ("Method B"), wherein R1, R2a, R2b, R2c, R3a, R3b, R4a, R4b, R9, R10, m1 and m2 are as defined herein. Referring to General Reaction Scheme 2, compounds of structure A-3 are prepared as described in General Scheme 1. Treatment of A-3 with an appropriately substituted cyclic group and an activating agent, such as HATU, provides B-1. B-1 is deprotected and the "E" moiety installed to yield B-3 in a manner analogous to that described above with regard to General Reaction Scheme 1.
General Reaction Scheme 3
C-9
Embodiments of the compound of structure (I) (e.g., compound C-9) can be prepared according to General Reaction Scheme 3 ("Method C"), wherein R1, R2a, R2b, R2c, R3a, R3b, R4a, R4b, R9, R10, m1 and m2 are as defined herein. Referring to General Reaction Scheme 3, compounds of structure C-1 are purchased from commercial sources or prepared according to methods known in the art. C-1 is reacted with diethyl 2-(ethoxymethylene)malonate to yield C-2. C-2 can then be cyclized by heating in an appropriate high-boiling solvent (e.g., Ph20) to yield quinolone C-3. Chlorination of C-3 yields C-4 which is reacted with an appropriate heterocyclic moiety to yield C-5. The desired R1 moiety is installed using Suzuki chemistry as described above. The protecting group of C-6 is removed, and the free amine is optionally functionalized to yield C-7. Saponification of C-7 followed by decarboxylation and amination yields C-8. C-8 is then treated in a manner analogous to that described for Method A to yield C-9.
Additional general synthetic methods are provided in the Examples. It will be apparent to one of ordinary skill in the art that all compounds of structure (I) can be prepared according to one or more of the methods described herein or otherwise known in the art. It will also be apparent that in some instances it will be necessary to use a differently substituted starting material and/or protecting groups to arrive at the desired compound when following the general procedures described herein. Various substituents may also be added at various points in the synthetic scheme to prepare the desired compound.
Further, one skilled in the art will recognize that certain modifications to the above schemes and those provided in the examples are possible to prepare different embodiments of compounds of structure (I). For example, for ease of illustration the General Reaction Schemes above depict preparation of compounds of structure (I) wherein R2a, R2b and R2c are present in the starting material. However, it will be apparent to one of ordinary skill in the art that these substituents may be added at any point during the synthetic scheme or obtained by using differently substituted starting materials and/or adding the desired substituent using methods known in the art.
It will also be appreciated by those skilled in the art that in the processes for preparing the compounds described herein the functional groups of intermediate compounds may need to be protected by suitable protecting groups. Such functional groups include, but are not limited to, hydroxy, amino, mercapto and carboxylic acid. Suitable protecting groups for hydroxy include trialkylsilyl or diarylalkylsilyl (for example, t-butyldimethylsilyl, t-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable protecting groups for amino, amidino and guanidino include t-butoxycarbonyl, benzyloxycarbonyl, and the like. Suitable protecting groups for mercapto include -C(0)-R" (where R" is alkyl, aryl or arylalkyl), /?-methoxybenzyl, trityl and the like. Suitable protecting groups for carboxylic acid include alkyl, aryl or arylalkyl esters. Protecting groups are optionally added or removed in accordance with standard techniques, which are known to one skilled in the art and as described herein. The use of protecting groups is described in detail in Green, T.W. and P.G.M. Wutz, Protective Groups in Organic Synthesis (1999), 3rd Ed., Wiley. As one of skill in the art would appreciate, the protecting group may also be a polymer resin such as a Wang resin, Rink resin or a 2-chlorotrityl-chloride resin.
It will also be appreciated by those skilled in the art, although such protected derivatives of compounds of this invention may not possess pharmacological activity as such, they may be administered to a mammal and thereafter metabolized in the body to form compounds of the invention which are pharmacologically active. Such derivatives may therefore be described as "prodrugs". All prodrugs of compounds of this invention are included within the scope of the invention.
Pharmaceutical Compositions
Other embodiments are directed to pharmaceutical compositions. The pharmaceutical composition comprises any one (or more) of the foregoing compounds and a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical composition is formulated for oral administration. In other embodiments, the pharmaceutical composition is formulated for injection. In still more embodiments, the pharmaceutical compositions comprise a compound as disclosed herein and an additional therapeutic agent (e.g., anticancer agent). Non-limiting examples of such therapeutic agents are described herein below.
Suitable routes of administration include, but are not limited to, oral, intravenous, rectal, aerosol, parenteral, ophthalmic, pulmonary, transmucosal, transdermal, vaginal, otic, nasal, and topical administration. In addition, by way of example only, parenteral delivery includes intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intralymphatic, and intranasal injections.
In certain embodiments, a compound as described herein is administered in a local rather than systemic manner, for example, via injection of the compound directly into an organ, often in a depot preparation or sustained release formulation. In specific embodiments, long acting formulations are administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
Furthermore, in other embodiments, the drug is delivered in a targeted drug delivery system, for example, in a liposome coated with organ-specific antibody. In such embodiments, the liposomes are targeted to and taken up selectively by the organ. In yet other embodiments, the compound as described herein is provided in the form of a rapid release formulation, in the form of an extended release formulation, or in the form of an intermediate release formulation. In yet other embodiments, the compound described herein is administered topically.
The compounds according to the invention are effective over a wide dosage range. For example, in the treatment of adult humans, dosages from 0.01 to 1000 mg, from 0.5 to 100 mg, from 1 to 50 mg per day, and from 5 to 40 mg per day are examples of dosages that are used in some embodiments. An exemplary dosage is 10 to 30 mg per day. The exact dosage will depend upon the route of administration, the form in which the compound is administered, the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician.
In some embodiments, a compound of the invention is administered in a single dose. Typically, such administration will be by injection, e.g., intravenous injection, in order to introduce the agent quickly. However, other routes are used as appropriate. A single dose of a compound of the invention may also be used for treatment of an acute condition.
In some embodiments, a compound of the invention is administered in multiple doses. In some embodiments, dosing is about once, twice, three times, four times, five times, six times, or more than six times per day. In other embodiments, dosing is about once a month, once every two weeks, once a week, or once every other day. In another embodiment a compound of the invention and another agent are administered together about once per day to about 6 times per day. In another embodiment the administration of a compound of the invention and an agent continues for less than about 7 days. In yet another embodiment the administration continues for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, continuous dosing is achieved and maintained as long as necessary.
Administration of the compounds of the invention may continue as long as necessary. In some embodiments, a compound of the invention is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, or 28 days. In some embodiments, a compound of the invention is administered for less than 28, 14, 7, 6, 5, 4, 3, 2, or 1 day. In some embodiments, a compound of the invention is administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
In some embodiments, the compounds of the invention are administered in dosages. It is known in the art that due to intersubject variability in compound pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy. Dosing for a compound of the invention may be found by routine experimentation in light of the instant disclosure.
In some embodiments, the compounds described herein are formulated into pharmaceutical compositions. In specific embodiments, pharmaceutical compositions are formulated in a conventional manner using one or more
physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any pharmaceutically acceptable techniques, carriers, and excipients are used as suitable to formulate the pharmaceutical compositions described herein: Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa. : Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975; Liberman, H.A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and
Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkinsl999).
Provided herein are pharmaceutical compositions comprising a compound of structure (I) and a pharmaceutically acceptable diluent(s), excipient(s), or carrier(s). In certain embodiments, the compounds described are administered as pharmaceutical compositions in which compounds of structure (I) are mixed with other active ingredients, as in combination therapy. Encompassed herein are all combinations of actives set forth in the combination therapies section below and throughout this disclosure. In specific embodiments, the pharmaceutical compositions include one or more compounds of structure (I).
A pharmaceutical composition, as used herein, refers to a mixture of a compound of structure (I) with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients. In certain embodiments, the pharmaceutical composition facilitates administration of the compound to an organism. In some embodiments, practicing the methods of treatment or use provided herein, therapeutically effective amounts of compounds of structure (I) provided herein are administered in a pharmaceutical composition to a mammal having a disease, disorder or medical condition to be treated. In specific embodiments, the mammal is a human. In certain embodiments, therapeutically effective amounts vary depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors. The compounds described herein are used singly or in combination with one or more therapeutic agents as components of mixtures.
In one embodiment, one or more compounds of structure (I) is formulated in an aqueous solutions. In specific embodiments, the aqueous solution is selected from, by way of example only, a physiologically compatible buffer, such as Hank's solution, Ringer's solution, or physiological saline buffer. In other
embodiments, one or more compound of structure (I) is/are formulated for transmucosal administration. In specific embodiments, transmucosal formulations include penetrants that are appropriate to the barrier to be permeated. In still other embodiments wherein the compounds described herein are formulated for other parenteral injections, appropriate formulations include aqueous or nonaqueous solutions. In specific embodiments, such solutions include physiologically compatible buffers and/or excipients. In another embodiment, compounds described herein are formulated for oral administration. Compounds described herein are formulated by combining the active compounds with, e.g., pharmaceutically acceptable carriers or excipients. In various embodiments, the compounds described herein are formulated in oral dosage forms that include, by way of example only, tablets, powders, pills, dragees, capsules, liquids, gels, syrups, elixirs, slurries, suspensions and the like.
In certain embodiments, pharmaceutical preparations for oral use are obtained by mixing one or more solid excipient with one or more of the compounds described herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as: for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methylcellulose,
microcrystalline cellulose, hydroxypropylmethylcellulose, sodium
carboxymethylcellulose; or others such as: polyvinylpyrrolidone (PVP or povidone) or calcium phosphate. In specific embodiments, disintegrating agents are optionally added. Disintegrating agents include, by way of example only, cross-linked
croscarmellose sodium, polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
In one embodiment, dosage forms, such as dragee cores and tablets, are provided with one or more suitable coating. In specific embodiments, concentrated sugar solutions are used for coating the dosage form. The sugar solutions, optionally contain additional components, such as by way of example only, gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs and/or pigments are also optionally added to the coatings for identification purposes.
Additionally, the dyestuffs and/or pigments are optionally utilized to characterize different combinations of active compound doses.
In certain embodiments, therapeutically effective amounts of at least one of the compounds described herein are formulated into other oral dosage forms. Oral dosage forms include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. In specific embodiments, push-fit capsules contain the active ingredients in admixture with one or more filler. Fillers include, by way of example only, lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In other embodiments, soft capsules, contain one or more active compound that is dissolved or suspended in a suitable liquid. Suitable liquids include, by way of example only, one or more fatty oil, liquid paraffin, or liquid polyethylene glycol. In addition, stabilizers are optionally added.
In other embodiments, therapeutically effective amounts of at least one of the compounds described herein are formulated for buccal or sublingual
administration. Formulations suitable for buccal or sublingual administration include, by way of example only, tablets, lozenges, or gels. In still other embodiments, the compounds described herein are formulated for parental injection, including
formulations suitable for bolus injection or continuous infusion. In specific
embodiments, formulations for injection are presented in unit dosage form (e.g., in ampoules) or in multi-dose containers. Preservatives are, optionally, added to the injection formulations. In still other embodiments, the pharmaceutical compositions are formulated in a form suitable for parenteral injection as sterile suspensions, solutions or emulsions in oily or aqueous vehicles. Parenteral injection formulations optionally contain formulatory agents such as suspending, stabilizing and/or dispersing agents. In specific embodiments, pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. In additional embodiments, suspensions of the active compounds (e.g., compounds of structure (I)) are prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles for use in the pharmaceutical compositions described herein include, by way of example only, fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. In certain specific embodiments, aqueous injection suspensions contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension contains suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Alternatively, in other embodiments, the active ingredient is in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
In still other embodiments, the compounds of structure (I) are administered topically. The compounds described herein are formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, pastes, medicated sticks, balms, creams or ointments. Such pharmaceutical
compositions optionally contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.
In yet other embodiments, the compounds of structure (I) are formulated for transdermal administration. In specific embodiments, transdermal formulations employ transdermal delivery devices and transdermal delivery patches and can be lipophilic emulsions or buffered, aqueous solutions, dissolved and/or dispersed in a polymer or an adhesive. In various embodiments, such patches are constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents. In additional embodiments, the transdermal delivery of the compounds of structure (I) is
accomplished by means of iontophoretic patches and the like. In certain embodiments, transdermal patches provide controlled delivery of the compounds of structure (I). In specific embodiments, the rate of absorption is slowed by using rate-controlling membranes or by trapping the compound within a polymer matrix or gel. In alternative embodiments, absorption enhancers are used to increase absorption. Absorption enhancers or carriers include absorbable pharmaceutically acceptable solvents that assist passage through the skin. For example, in one embodiment, transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
In other embodiments, the compounds of structure (I) are formulated for administration by inhalation. Various forms suitable for administration by inhalation include, but are not limited to, aerosols, mists or powders. Pharmaceutical compositions of any of compound of structure (I) are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant (e.g., dichlorodifluoromethane, trichlorofluoromethane,
dichlorotetrafluoroethane, carbon dioxide or other suitable gas). In specific
embodiments, the dosage unit of a pressurized aerosol is determined by providing a valve to deliver a metered amount. In certain embodiments, capsules and cartridges of, such as, by way of example only, gelatin for use in an inhaler or insufflator are formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
In still other embodiments, the compounds of structure (I) are formulated in rectal compositions such as enemas, rectal gels, rectal foams, rectal aerosols, suppositories, jelly suppositories, or retention enemas, containing conventional suppository bases such as cocoa butter or other glycerides, as well as synthetic polymers such as polyvinylpyrrolidone, PEG, and the like. In suppository forms of the compositions, a low-melting wax such as, but not limited to, a mixture of fatty acid glycerides, optionally in combination with cocoa butter is first melted.
In certain embodiments, pharmaceutical compositions are formulated in any conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any pharmaceutically acceptable techniques, carriers, and excipients are optionally used as suitable. Pharmaceutical compositions comprising a compound of structure (I) are manufactured in a
conventional manner, such as, by way of example only, by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes.
Pharmaceutical compositions include at least one pharmaceutically acceptable carrier, diluent or excipient and at least one compound of structure (I), described herein as an active ingredient. The active ingredient is in free-acid or free- base form, or in a pharmaceutically acceptable salt form. In addition, the methods and pharmaceutical compositions described herein include the use of N-oxides, crystalline forms (also known as polymorphs), as well as active metabolites of these compounds having the same type of activity. All tautomers of the compounds described herein are included within the scope of the compounds presented herein. Additionally, the compounds described herein encompass unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. The solvated forms of the compounds presented herein are also considered to be disclosed herein. In addition, the pharmaceutical compositions optionally include other medicinal or pharmaceutical agents, carriers, adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure, buffers, and/or other therapeutically valuable substances.
Methods for the preparation of compositions comprising the compounds described herein include formulating the compounds with one or more inert, pharmaceutically acceptable excipients or carriers to form a solid, semi-solid or liquid. Solid compositions include, but are not limited to, powders, tablets, dispersible granules, capsules, cachets, and suppositories. Liquid compositions include solutions in which a compound is dissolved, emulsions comprising a compound, or a solution containing liposomes, micelles, or nanoparticles comprising a compound as disclosed herein. Semi-solid compositions include, but are not limited to, gels, suspensions and creams. The form of the pharmaceutical compositions described herein include liquid solutions or suspensions, solid forms suitable for solution or suspension in a liquid prior to use, or as emulsions. These compositions also optionally contain minor amounts of nontoxic, auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, and so forth.
In some embodiments, pharmaceutical composition comprising at least one compound of structure (I) illustratively takes the form of a liquid where the agents are present in solution, in suspension or both. Typically when the composition is administered as a solution or suspension a first portion of the agent is present in solution and a second portion of the agent is present in particulate form, in suspension in a liquid matrix. In some embodiments, a liquid composition includes a gel formulation. In other embodiments, the liquid composition is aqueous.
In certain embodiments, useful aqueous suspensions contain one or more polymers as suspending agents. Useful polymers include water-soluble polymers such as cellulosic polymers, e.g., hydroxypropyl methylcellulose, and water-insoluble polymers such as cross-linked carboxyl-containing polymers. Certain pharmaceutical compositions described herein comprise a mucoadhesive polymer, selected for example from carboxymethylcellulose, carbomer (acrylic acid polymer),
poly(methylmethacrylate), polyacrylamide, polycarbophil, acrylic acid/butyl acrylate copolymer, sodium alginate and dextran.
Useful pharmaceutical compositions also, optionally, include solubilizing agents to aid in the solubility of a compound of structure (I). The term "solubilizing agent" generally includes agents that result in formation of a micellar solution or a true solution of the agent. Certain acceptable nonionic surfactants, for example polysorbate 80, are useful as solubilizing agents, as can ophthalmically acceptable glycols, polyglycols, e.g., polyethylene glycol 400, and glycol ethers.
Furthermore, useful pharmaceutical compositions optionally include one or more pH adjusting agents or buffering agents, including acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane; and buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride. Such acids, bases and buffers are included in an amount required to maintain pH of the composition in an acceptable range.
Additionally, useful compositions also, optionally, include one or more salts in an amount required to bring osmolality of the composition into an acceptable range. Such salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions; suitable salts include sodium chloride, potassium chloride, sodium thiosulfate, sodium bisulfite and ammonium sulfate. Other useful pharmaceutical compositions optionally include one or more preservatives to inhibit microbial activity. Suitable preservatives include mercury-containing substances such as merfen and thiomersal; stabilized chlorine dioxide; and quaternary ammonium compounds such as benzalkonium chloride, cetyltrimethylammonium bromide and cetylpyridinium chloride.
Still other useful compositions include one or more surfactants to enhance physical stability or for other purposes. Suitable nonionic surfactants include polyoxyethylene fatty acid glycerides and vegetable oils, e.g., polyoxyethylene (60) hydrogenated castor oil; and polyoxyethylene alkylethers and alkylphenyl ethers, e.g., octoxynol 10, octoxynol 40.
Still other useful compositions include one or more antioxidants to enhance chemical stability where required. Suitable antioxidants include, by way of example only, ascorbic acid and sodium metabisulfite.
In certain embodiments, aqueous suspension compositions are packaged in single-dose non-reclosable containers. Alternatively, multiple-dose reclosable containers are used, in which case it is typical to include a preservative in the composition.
In alternative embodiments, other delivery systems for hydrophobic pharmaceutical compounds are employed. Liposomes and emulsions are examples of delivery vehicles or carriers useful herein. In certain embodiments, organic solvents such as N-methylpyrrolidone are also employed. In additional embodiments, the compounds described herein are delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials are useful herein. In some embodiments, sustained-release capsules release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization are employed.
In certain embodiments, the formulations described herein comprise one or more antioxidants, metal chelating agents, thiol containing compounds and/or other general stabilizing agents. Examples of such stabilizing agents, include, but are not limited to: (a) about 0.5% to about 2% w/v glycerol, (b) about 0.1% to about 1% w/v methionine, (c) about 0.1% to about 2% w/v monothioglycerol, (d) about 1 mM to about 10 mM EDTA, (e) about 0.01% to about 2% w/v ascorbic acid, (f) 0.003% to about 0.02% w/v polysorbate 80, (g) 0.001% to about 0.05% w/v. polysorbate 20, (h) arginine, (i) heparin, (j) dextran sulfate, (k) cyclodextrins, (1) pentosan poly sulfate and other heparinoids, (m) divalent cations such as magnesium and zinc; or (n)
combinations thereof.
In some embodiments, the concentration of one or more compounds provided in the pharmaceutical compositions of the present invention is less than 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 1 1%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002%, or 0.0001% w/w, w/v or v/v.
In some embodiments, the concentration of one or more compounds of the invention is greater than 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19.75%, 19.50%, 19.25% 19%, 18.75%, 18.50%, 18.25% 18%, 17.75%, 17.50%, 17.25% 17%, 16.75%, 16.50%, 16.25% 16%, 15.75%, 15.50%, 15.25% 15%, 14.75%, 14.50%, 14.25% 14%, 13.75%, 13.50%, 13.25% 13%, 12.75%, 12.50%, 12.25% 12%, 1 1.75%, 1 1.50%, 1 1.25% 1 1%, 10.75%, 10.50%, 10.25% 10%, 9.75%, 9.50%, 9.25% 9%, 8.75%, 8.50%, 8.25% 8%, 7.75%, 7.50%, 7.25% 7%, 6.75%, 6.50%, 6.25% 6%, 5.75%, 5.50%, 5.25% 5%, 4.75%, 4.50%, 4.25%, 4%, 3.75%, 3.50%, 3.25%, 3%, 2.75%, 2.50%, 2.25%, 2%, 1.75%, 1.50%, 125% , 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002%, or 0.0001% w/w, w/v, or v/v.
In some embodiments, the concentration of one or more compounds of the invention is in the range from approximately 0.0001%) to approximately 50%, approximately 0.001 ) to approximately 40 %, approximately 0.01% to approximately 30%, approximately 0.02% to approximately 29%, approximately 0.03%> to
approximately 28%, approximately 0.04% to approximately 27%, approximately 0.05% to approximately 26%, approximately 0.06% to approximately 25%, approximately 0.07%) to approximately 24%, approximately 0.08% to approximately 23%,
approximately 0.09% to approximately 22%, approximately 0.1% to approximately
21%), approximately 0.2% to approximately 20%, approximately 0.3% to approximately 19%), approximately 0.4% to approximately 18%, approximately 0.5% to approximately 17%), approximately 0.6% to approximately 16%, approximately 0.7% to approximately 15%), approximately 0.8% to approximately 14%, approximately 0.9% to approximately 12%), approximately 1% to approximately 10% w/w, w/v or v/v.
In some embodiments, the concentration of one or more compounds of the invention is in the range from approximately 0.001%) to approximately 10%, approximately 0.01% to approximately 5%, approximately 0.02% to approximately 4.5%o, approximately 0.03% to approximately 4%, approximately 0.04% to
approximately 3.5%, approximately 0.05% to approximately 3%, approximately 0.06% to approximately 2.5%, approximately 0.07% to approximately 2%, approximately 0.08%) to approximately 1.5%, approximately 0.09% to approximately 1%,
approximately 0.1% to approximately 0.9% w/w, w/v or v/v.
In some embodiments, the amount of one or more compounds of the invention is equal to or less than 10 g, 9.5 g, 9.0 g, 8.5 g, 8.0 g, 7.5 g, 7.0 g, 6.5 g, 6.0 g, 5.5 g, 5.0 g, 4.5 g, 4.0 g, 3.5 g, 3.0 g, 2.5 g, 2.0 g, 1.5 g, 1.0 g, 0.95 g, 0.9 g, 0.85 g, 0.8 g, 0.75 g, 0.7 g, 0.65 g, 0.6 g, 0.55 g, 0.5 g, 0.45 g, 0.4 g, 0.35 g, 0.3 g, 0.25 g, 0.2 g, 0.15 g, 0.1 g, 0.09 g, 0.08 g, 0.07 g, 0.06 g, 0.05 g, 0.04 g, 0.03 g, 0.02 g, 0.01 g, 0.009 g, 0.008 g, 0.007 g, 0.006 g, 0.005 g, 0.004 g, 0.003 g, 0.002 g, 0.001 g, 0.0009 g, 0.0008 g, 0.0007 g, 0.0006 g, 0.0005 g, 0.0004 g, 0.0003 g, 0.0002 g, or 0.0001 g.
In some embodiments, the amount of one or more compounds of the invention is more than 0.0001 g, 0.0002 g, 0.0003 g, 0.0004 g, 0.0005 g, 0.0006 g, 0.0007 g, 0.0008 g, 0.0009 g, 0.001 g, 0.0015 g, 0.002 g, 0.0025 g, 0.003 g, 0.0035 g, 0.004 g, 0.0045 g, 0.005 g, 0.0055 g, 0.006 g, 0.0065 g, 0.007 g, 0.0075 g, 0.008 g, 0.0085 g, 0.009 g, 0.0095 g, 0.01 g, 0.015 g, 0.02 g, 0.025 g, 0.03 g, 0.035 g, 0.04 g, 0.045 g, 0.05 g, 0.055 g, 0.06 g, 0.065 g, 0.07 g, 0.075 g, 0.08 g, 0.085 g, 0.09 g, 0.095 g, 0.1 g, , 0.15 g, 0.2 g, , 0.25 g, 0.3 g, , 0.35 g, 0.4 g, , 0.45 g, 0.5 g, 0.55 g, 0.6 g, , 0.65 g, 0.7 g, 0.75 g, 0.8 g, 0.85 g, 0.9 g, 0.95 g, 1 g, 1.5 g, 2 g, 2.5, 3 g, 3.5, 4 g, 4.5 g, 5 g, 5.5 g, 6 g, 6.5g, 7 g, 7.5g, 8 g, 8.5 g, 9 g, 9.5 g, or 10 g.
In some embodiments, the amount of one or more compounds of the invention is in the range of 0.0001-10 g, 0.0005-9 g, 0.001-8 g, 0.005-7 g, 0.01-6 g, 0.05-5 g, 0.1-4 g, 0.5-4 g, or 1-3 g.
Kits/ Articles of Manufacture
For use in the therapeutic applications described herein, kits and articles of manufacture are also provided. In some embodiments, such kits comprise a carrier, package, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in a method described herein. Suitable containers include, for example, bottles, vials, syringes, and test tubes. The containers are formed from a variety of materials such as glass or plastic.
The articles of manufacture provided herein contain packaging materials. Packaging materials for use in packaging pharmaceutical products include those found in, e.g., U.S. Pat. Nos. 5,323,907, 5,052,558 and 5,033,252. Examples of
pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of
administration and treatment. For example, the container(s) includes one or more compounds described herein, optionally in a composition or in combination with another agent as disclosed herein. The container(s) optionally have a sterile access port (for example the container is an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). Such kits optionally comprising a compound with an identifying description or label or instructions relating to its use in the methods described herein. For example, a kit typically includes one or more additional containers, each with one or more of various materials (such as reagents, optionally in concentrated form, and/or devices) desirable from a commercial and user standpoint for use of a compound described herein. Non-limiting examples of such materials include, but not limited to, buffers, diluents, filters, needles, syringes; carrier, package, container, vial and/or tube labels listing contents and/or instructions for use, and package inserts with instructions for use. A set of instructions will also typically be included. A label is optionally on or associated with the container. For example, a label is on a container when letters, numbers or other characters forming the label are attached, molded or etched into the container itself, a label is associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert. In addition, a label is used to indicate that the contents are to be used for a specific therapeutic application. In addition, the label indicates directions for use of the contents, such as in the methods described herein. In certain embodiments, the pharmaceutical compositions is presented in a pack or dispenser device which contains one or more unit dosage forms containing a compound provided herein. The pack for example contains metal or plastic foil, such as a blister pack. Or, the pack or dispenser device is accompanied by instructions for administration. Or, the pack or dispenser is
accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, is the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert. In some embodiments, compositions containing a compound provided herein formulated in a compatible pharmaceutical carrier are prepared, placed in an
appropriate container, and labeled for treatment of an indicated condition.
Methods
Embodiments of the present invention provide a method of inhibiting RAS-mediated cell signaling comprising contacting a cell with an effective amount of one or more compounds disclosed herein. Inhibition of RAS-mediated signal transduction can be assessed and demonstrated by a wide variety of ways known in the art. Non-limiting examples include a showing of (a) a decrease in GTPase activity of RAS; (b) a decrease in GTP binding affinity or an increase in GDP binding affinity; (c) an increase in K off of GTP or a decrease in K off of GDP; (d) a decrease in the levels of signaling transduction molecules downstream in the RAS pathway, such as a decrease in pMEK level; and/or (e) a decrease in binding of RAS complex to downstream signaling molecules including but not limited to Raf. Kits and
commercially available assays can be utilized for determining one or more of the above.
Embodiments also provide methods of using the compounds or pharmaceutical compositions of the present invention to treat disease conditions, including but not limited to conditions implicated by G12C KRAS, HRAS or NRAS mutation, G12C HRAS mutation and/or G12C NRAS mutation (e.g., cancer).
In some embodiments, a method for treatment of cancer is provided, the method comprising administering an effective amount of any of the foregoing pharmaceutical compositions comprising a compound of structure (I) to a subject in need thereof. In some embodiments, the cancer is mediated by a KRAS, HRAS or NRAS G12C mutation. In other embodiments, the cancer is pancreatic cancer, colon cancer, MYH associated polyposis, colorectal cancer or lung cancer.
In some embodiments the invention provides method of treating a disorder in a subject in need thereof, wherein the said method comprises determining if the subject has a KRAS, HRAS or NRAS G12C mutation and if the subject is determined to have the KRAS, HRAS or NRAS G12C mutation, then administering to the subject a therapeutically effective dose of at least one compound of structure (I) or a pharmaceutically acceptable salt, ester, prodrug, tautomer, solvate, hydrate or derivative thereof.
The disclosed compounds strongly inhibit anchorage-independent cell growth and therefore have the potential to inhibit tumor metastasis. Accordingly, in another embodiment the disclosure provides a method for inhibiting tumor metastasis, the method comprising administering an effective amount a pharmaceutical composition of comprising any of the compounds disclosed herein and a
pharmaceutically acceptable carrier to a subject in need thereof.
KRAS, HRAS or RAS G12C mutations have also been identified in hematological malignancies (e.g., cancers that affect blood, bone marrow and/or lymph nodes). Accordingly, certain embodiments are directed to administration of a disclosed compounds (e.g., in the form of a pharmaceutical composition) to a patient in need of treatment of a hematological malignancy. Such malignancies include, but are not limited to leukemias and lymphomas. For example, the presently disclosed compounds can be used for treatment of diseases such as Acute lymphoblastic leukemia (ALL), Acute myelogenous leukemia (AML), Chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), Chronic myelogenous leukemia (CML), Acute monocytic leukemia (AMoL) and/ or other leukemias. In other embodiments, the compounds are useful for treatment of lymphomas such as all subtypes of Hodgkin's lymphoma or non-Hodgkin's lymphoma.
Determining whether a tumor or cancer comprises a G12C KRAS,
HRAS or NRAS mutation can be undertaken by assessing the nucleotide sequence encoding the KRAS, HRAS or NRAS protein, by assessing the amino acid sequence of the KRAS, HRAS or NRAS protein, or by assessing the characteristics of a putative KRAS, HRAS or NRAS mutant protein. The sequence of wild-type human KRAS, HRAS or NRAS is known in the art, (e.g. Accession No. NP203524).
Methods for detecting a mutation in a KRAS, HRAS or NRAS nucleotide sequence are known by those of skill in the art. These methods include, but are not limited to, polymeRASe chain reaction-restriction fragment length
polymorphism (PCR-RFLP) assays, polymeRASe chain reaction-single strand conformation polymorphism (PCR-SSCP) assays, real-time PCR assays, PCR sequencing, mutant allele-specific PCR amplification (MASA) assays, direct sequencing, primer extension reactions, electrophoresis, oligonucleotide ligation assays, hybridization assays, TaqMan assays, SNP genotyping assays, high resolution melting assays and microarray analyses. In some embodiments, samples are evaluated for G12C KRAS, HRAS or NRAS mutations by real-time PCR. In real-time PCR, fluorescent probes specific for the KRAS, HRAS or RAS G12C mutation are used. When a mutation is present, the probe binds and fluorescence is detected. In some embodiments, the KRAS, HRAS or NRAS G12C mutation is identified using a direct sequencing method of specific regions (e.g., exon 2 and/or exon 3) in the KRAS, HRAS or NRAS gene. This technique will identify all possible mutations in the region sequenced.
Methods for detecting a mutation in a KRAS, HRAS or NRAS protein are known by those of skill in the art. These methods include, but are not limited to, detection of a KRAS, HRAS or NRAS mutant using a binding agent (e.g., an antibody) specific for the mutant protein, protein electrophoresis and Western blotting, and direct peptide sequencing.
Methods for determining whether a tumor or cancer comprises a G12C KRAS, HRAS or NRAS mutation can use a variety of samples. In some embodiments, the sample is taken from a subject having a tumor or cancer. In some embodiments, the sample is taken from a subject having a cancer or tumor. In some embodiments, the sample is a fresh tumor/cancer sample. In some embodiments, the sample is a frozen tumor/cancer sample. In some embodiments, the sample is a formalin-fixed paraffin- embedded sample. In some embodiments, the sample is processed to a cell lysate. In some embodiments, the sample is processed to DNA or RNA.
Embodiments of the invention also relate to a method of treating a hyperproliferative disorder in a mammal that comprises administering to said mammal a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof. In some embodiments, said method relates to the treatment of cancer such as acute myeloid leukemia, cancer in adolescents, adrenocortical carcinoma childhood, AIDS- related cancers (e.g. Lymphoma and Kaposi's Sarcoma), anal cancer, appendix cancer, astrocytomas, atypical teratoid, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumors, burkitt lymphoma, carcinoid tumor, atypical teratoid, embryonal tumors, germ cell tumor, primary lymphoma, cervical cancer, childhood cancers, chordoma, cardiac tumors, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myleoproliferative disorders, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, extrahepatic ductal carcinoma in situ (DCIS), embryonal tumors, CNS cancer, endometrial cancer, ependymoma, esophageal cancer, esthesioneuroblastoma, ewing sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor, eye cancer, fibrous histiocytoma of bone, gall bladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumors (GIST), germ cell tumor, gestational trophoblastic tumor, hairy cell leukemia, head and neck cancer, heart cancer, liver cancer, Hodgkin's lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumors, pancreatic neuroendocrine tumors, kidney cancer, laryngeal cancer, lip and oral cavity cancer, liver cancer, lobular carcinoma in situ (LCIS), lung cancer, lymphoma, metastatic squamous neck cancer with occult primary, midline tract carcinoma, mouth cancer multiple endocrine neoplasia syndromes, multiple myeloma/plasma cell neoplasm, mycosis fungoides,
myelodysplastic syndromes, my elodysplastic/my el oproliferative neoplasms, multiple myeloma, merkel cell carcinoma, malignant mesothelioma, malignant fibrous histiocytoma of bone and osteosarcoma, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-Hodgkin's lymphoma, non-small cell lung cancer (NSCLC), oral cancer, lip and oral cavity cancer, oropharyngeal cancer, ovarian cancer, pancreatic cancer, papillomatosis, paraganglioma, paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pleuropulmonary blastoma, primary central nervous system (CNS) lymphoma, prostate cancer, rectal cancer, transitional cell cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, skin cancer, stomach (gastric) cancer, small cell lung cancer, small intestine cancer, soft tissue sarcoma, T-Cell lymphoma, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer, transitional cell cancer of the renal pelvis and ureter, trophoblastic tumor, unusual cancers of childhood, urethral cancer, uterine sarcoma, vaginal cancer, vulvar cancer, or Viral-Induced cancer. In some embodiments, said method relates to the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
In certain particular embodiments, the invention relates to methods for treatment of lung cancers, the methods comprise administering an effective amount of any of the above described compound (or a pharmaceutical composition comprising the same) to a subject in need thereof. In certain embodiments the lung cancer is a non- small cell lung carcinoma (NSCLC), for example adenocarcinoma, squamous-cell lung carcinoma or large-cell lung carcinoma. In other embodiments, the lung cancer is a small cell lung carcinoma. Other lung cancers treatable with the disclosed compounds include, but are not limited to, glandular tumors, carcinoid tumors and undifferentiated carcinomas.
Subjects that can be treated with compounds of the invention, or pharmaceutically acceptable salt, ester, prodrug, solvate, tautomer, hydrate or derivative of said compounds, according to the methods of this invention include, for example, subjects that have been diagnosed as having acute myeloid leukemia, acute myeloid leukemia, cancer in adolescents, adrenocortical carcinoma childhood, AIDS-related cancers (e.g. Lymphoma and Kaposi's Sarcoma), anal cancer, appendix cancer, astrocytomas, atypical teratoid, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumors, burkitt lymphoma, carcinoid tumor, atypical teratoid, embryonal tumors, germ cell tumor, primary lymphoma, cervical cancer, childhood cancers, chordoma, cardiac tumors, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myleoproliferative disorders, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, extrahepatic ductal carcinoma in situ (DCIS), embryonal tumors, CNS cancer, endometrial cancer, ependymoma, esophageal cancer,
esthesioneuroblastoma, ewing sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor, eye cancer, fibrous histiocytoma of bone, gall bladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumors (GIST), germ cell tumor, gestational trophoblastic tumor, hairy cell leukemia, head and neck cancer, heart cancer, liver cancer, Hodgkin's lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumors, pancreatic neuroendocrine tumors, kidney cancer, laryngeal cancer, lip and oral cavity cancer, liver cancer, lobular carcinoma in situ (LCIS), lung cancer, lymphoma, metastatic squamous neck cancer with occult primary, midline tract carcinoma, mouth cancer multiple endocrine neoplasia syndromes, multiple myeloma/plasma cell neoplasm, mycosis fungoides, myelodysplastic syndromes, myelodysplastic/myeloproliferative neoplasms, multiple myeloma, merkel cell carcinoma, malignant mesothelioma, malignant fibrous histiocytoma of bone and osteosarcoma, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-Hodgkin's lymphoma, non-small cell lung cancer (NSCLC), oral cancer, lip and oral cavity cancer, oropharyngeal cancer, ovarian cancer, pancreatic cancer, papillomatosis, paraganglioma, paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pleuropulmonary blastoma, primary central nervous system (CNS) lymphoma, prostate cancer, rectal cancer, transitional cell cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, skin cancer, stomach (gastric) cancer, small cell lung cancer, small intestine cancer, soft tissue sarcoma, T-Cell lymphoma, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer, transitional cell cancer of the renal pelvis and ureter, trophoblastic tumor, unusual cancers of childhood, urethral cancer, uterine sarcoma, vaginal cancer, vulvar cancer, or Viral-Induced cancer. In some embodiments subjects that are treated with the compounds of the invention include subjects that have been diagnosed as having a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e.g., benign prostatic hypertrophy (BPH)).
Embodiments of the invention further provide methods of modulating a G12C Mutant KRAS, HRAS or RAS protein activity by contacting the protein with an effective amount of a compound of the invention. Modulation can be inhibiting or activating protein activity. In some embodiments, the invention provides methods of inhibiting protein activity by contacting the G12C Mutant KRAS, HRAS or NRAS protein with an effective amount of a compound of the invention in solution. In some embodiments, the invention provides methods of inhibiting the G12C Mutant KRAS, HRAS or NRAS protein activity by contacting a cell, tissue, organ that express the protein of interest. In some embodiments, the invention provides methods of inhibiting protein activity in subject including but not limited to rodents and mammal (e.g., human) by administering into the subject an effective amount of a compound of the invention. In some embodiments, the percentage modulation exceeds 25%, 30%, 40%, 50%), 60%), 70%), 80%), or 90%. In some embodiments, the percentage of inhibiting exceeds 25%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%.
In some embodiments, the invention provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in a cell by contacting said cell with an amount of a compound of the invention sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said cell. In some embodiments, the invention provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in a tissue by contacting said tissue with an amount of a compound of the invention sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said tissue. In some embodiments, the invention provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in an organism by contacting said organism with an amount of a compound of the invention sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said organism. In some embodiments, the invention provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in an animal by contacting said animal with an amount of a compound of the invention sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said animal. In some embodiments, the invention provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in a mammal by contacting said mammal with an amount of a compound of the invention sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said mammal. In some embodiments, the invention provides methods of inhibiting KRAS, HRAS or NRAS G12C activity in a human by contacting said human with an amount of a compound of the invention sufficient to inhibit the activity of KRAS, HRAS or NRAS G12C in said human. In other embodiments, the present invention provides methods of treating a disease mediated by KRAS, HRAS or NRAS G12C activity in a subject in need of such treatment. Other embodiments provide methods for combination therapies in which an agent known to modulate other pathways, or other components of the same pathway, or even overlapping sets of target enzymes are used in combination with a compound of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, tautomer, hydrate or derivative thereof. In one aspect, such therapy includes but is not limited to the combination of one or more compounds of the invention with
chemotherapeutic agents, therapeutic antibodies, and radiation treatment, to provide a synergistic or additive therapeutic effect.
Many chemotherapeutics are presently known in the art and can be used in combination with the compounds of the invention. In some embodiments, the chemotherapeutic is selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomeRASe inhibitors, biological response modifiers, anti- hormones, angiogenesis inhibitors, and anti-androgens.
Non-limiting examples are chemotherapeutic agents, cytotoxic agents, and non-peptide small molecules such as Gleevec® (Imatinib Mesylate), Velcade® (bortezomib), Casodex (bicalutamide), Iressa® (gefitinib), and Adriamycin as well as a host of chemotherapeutic agents. Non-limiting examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXANTM); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and
methylamelamines including altretamine, triethylenemelamine,
trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamine; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, calicheamicin, carabicin, carminomycin, carzinophilin, CasodexTM, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo- L-norleucine, doxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine,
dideoxyuridine, doxifluridine, enocitabine, floxuridine, androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfomithine; elliptinium acetate; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK.RTM.; razoxane; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; urethan; vindesine;
dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine;
arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxanes, e.g. paclitaxel
(TAXOLTM, Bristol-Myers Squibb Oncology, Princeton, N.J.) and docetaxel
(TAXOTERETM, Rhone-Poulenc Rorer, Antony, France); retinoic acid; esperamicins; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. Also included as suitable chemotherapeutic cell conditioners are anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens including for example tamoxifen, (NolvadexTM), raloxifene, aromatase inhibiting 4(5)- imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY 117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP- 16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; camptothecin-11 (CPT-11); topoisomeRASe inhibitor RFS 2000;
difluoromethylormthine (DMFO). Where desired, the compounds or pharmaceutical composition of the present invention can be used in combination with commonly prescribed anti-cancer drugs such as Herceptin®, Avastin®, Erbitux®, Rituxan®, Taxol®, Arimidex®, Taxotere®, ABVD, AVICINE, Abagovomab, Acridine carboxamide, Adecatumumab, 17-N-Allylamino- 17-demethoxygeldanamycin,
Alpharadin, Alvocidib, 3-Aminopyridine-2-carboxaldehyde thiosemicarbazone, Amonafide, Anthracenedione, Anti-CD22 immunotoxins, Antineoplastic,
Antitumorigenic herbs, Apaziquone, Atiprimod, Azathioprine, Belotecan,
Bendamustine, BIBW 2992, Biricodar, Brostallicin, Bryostatin, Buthionine
sulfoximine, CBV (chemotherapy), Calyculin, cell-cycle nonspecific antineoplastic agents, Dichloroacetic acid, Discodermolide, Elsamitrucin, Enocitabine, Epothilone, Eribulin, Everolimus, Exatecan, Exisulind, Ferruginol, Forodesine, Fosfestrol, ICE chemotherapy regimen, IT-101, Imexon, Imiquimod, Indolocarbazole, Irofulven, Laniquidar, Larotaxel, Lenalidomide, Lucanthone, Lurtotecan, Mafosfamide,
Mitozolomide, Nafoxidine, Nedaplatin, Olaparib, Ortataxel, PAC-1, Pawpaw,
Pixantrone, Proteasome inhibitor, Rebeccamycin, Resiquimod, Rubitecan, SN-38, Salinosporamide A, Sapacitabine, Stanford V, Swainsonine, Talaporfin, Tariquidar, Tegafur-uracil, Temodar, Tesetaxel, Triplatin tetranitrate, Tris(2-chloroethyl)amine, Troxacitabine, Uramustine, Vadimezan, Vinflunine, ZD6126 or Zosuquidar.
Embodiments further relate to a method for using the compounds or pharmaceutical compositions provided herein, in combination with radiation therapy for inhibiting abnormal cell growth or treating the hyperproliferative disorder in the mammal. Techniques for administering radiation therapy are known in the art, and these techniques can be used in the combination therapy described herein. The administration of the compound of the invention in this combination therapy can be determined as described herein.
Radiation therapy can be administered through one of several methods, or a combination of methods, including without limitation external-beam therapy, internal radiation therapy, implant radiation, stereotactic radiosurgery, systemic radiation therapy, radiotherapy and permanent or temporary interstitial brachytherapy. The term "brachytherapy," as used herein, refers to radiation therapy delivered by a spatially confined radioactive material inserted into the body at or near a tumor or other proliferative tissue disease site. The term is intended without limitation to include exposure to radioactive isotopes (e.g. At-211, 1-131, 1-125, Y-90, Re-186, Re-188, Sm- 153, Bi-212, P-32, and radioactive isotopes of Lu). Suitable radiation sources for use as a cell conditioner of the present invention include both solids and liquids. By way of non-limiting example, the radiation source can be a radionuclide, such as 1-125, 1-131, Yb-169, Ir-192 as a solid source, 1-125 as a solid source, or other radionuclides that emit photons, beta particles, gamma radiation, or other therapeutic rays. The radioactive material can also be a fluid made from any solution of radionuclide(s), e.g., a solution of 1-125 or 1-131, or a radioactive fluid can be produced using a slurry of a suitable fluid containing small particles of solid radionuclides, such as Au-198, Y-90. Moreover, the radionuclide(s) can be embodied in a gel or radioactive micro spheres.
Without being limited by any theory, the compounds of the present invention can render abnormal cells more sensitive to treatment with radiation for purposes of killing and/or inhibiting the growth of such cells. Accordingly, this invention further relates to a method for sensitizing abnormal cells in a mammal to treatment with radiation which comprises administering to the mammal an amount of a compound of the present invention or pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof, which amount is effective is sensitizing abnormal cells to treatment with radiation. The amount of the compound, salt, or solvate in this method can be determined according to the means for ascertaining effective amounts of such compounds described herein.
The compounds or pharmaceutical compositions of the invention can be used in combination with an amount of one or more substances selected from anti- angiogenesis agents, signal transduction inhibitors, antiproliferative agents, glycolysis inhibitors, or autophagy inhibitors.
Anti-angiogenesis agents, such as MMP-2 (matrix -metalloproteinase 2) inhibitors, MMP-9 (matrix-metalloprotienase 9) inhibitors, and COX-11 (cyclooxygenase 11) inhibitors, can be used in conjunction with a compound of the invention and pharmaceutical compositions described herein. Anti-angiogenesis agents include, for example, rapamycin, temsirolimus (CCI-779), everolimus (RAD001), sorafenib, sunitinib, and bevacizumab. Examples of useful COX-II inhibitors include CELEBREX™ (alecoxib), valdecoxib, and rofecoxib. Examples of useful matrix metalloproteinase inhibitors are described in WO 96/33172 (published October 24, 1996), WO 96/27583 (published March 7, 1996), European Patent Application No. 97304971.1 (filed July 8,1997), European Patent Application No. 99308617.2 (filed October 29, 1999), WO 98/07697 (published February 26,1998), WO 98/03516 (published January 29, 1998), WO 98/34918 (published August 13, 1998), WO 98/34915 (published August 13,1998), WO 98/33768 (published August 6, 1998), WO 98/30566 (published July 16, 1998), European Patent Publication 606,046 (published July 13, 1994), European Patent Publication 931, 788 (published July 28, 1999), WO
90/05719 (published May 31, 1990), WO 99/52910 (published October 21,1999), WO 99/52889 (published October 21, 1999), WO 99/29667 (published June 17,1999), PCT International Application No. PCT/IB98/01113 (filed July 21, 1998), European Patent Application No. 99302232.1 (filed March 25, 1999), Great Britain Patent Application No. 9912961.1 (filed June 3, 1999), United States Provisional Application No.
60/148,464 (filed August 12,1999), United States Patent 5,863, 949 (issued January 26, 1999), United States Patent 5,861, 510 (issued January 19,1999), and European Patent Publication 780,386 (published June 25, 1997), all of which are incorporated herein in their entireties by reference. Preferred MMP-2 and MMP-9 inhibitors are those that have little or no activity inhibiting MMP-1. More preferred, are those that selectively inhibit MMP-2 and/or AMP-9 relative to the other matrix- metalloproteinases (i. e., MAP-1, MMP-3, MMP-4, MMP-5, MMP-6, MMP- 7, MMP- 8, MMP-10, MMP-11, MMP-12, andMMP-13). Some specific examples of MMP inhibitors useful in the invention are AG-3340, RO 32-3555, and RS 13-0830.
Autophagy inhibitors include, but are not limited to chloroquine, 3- methyladenine, hydroxychloroquine (Plaquenil™), bafilomycin Al, 5-amino-4- imidazole carboxamide riboside (AICAR), okadaic acid, autophagy-suppressive algal toxins which inhibit protein phosphatases of type 2 A or type 1, analogues of cAMP, and drugs which elevate cAMP levels such as adenosine, LY204002, N6-mercaptopurine riboside, and vinblastine. In addition, antisense or siRNA that inhibits expression of proteins including but not limited to ATG5 (which are implicated in autophagy), may also be used.
Embodiments also relate to a method of and to a pharmaceutical composition for treating a cardiovascular disease in a mammal which comprises an amount of a compound of the invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, tautomer, hydrate or derivative thereof, or an isotopically-labeled derivative thereof, and an amount of one or more therapeutic agents use for the treatment of cardiovascular diseases.
Exemplary agents for use in cardiovascular disease applications are antithrombotic agents, e.g., prostacyclin and salicylates, thrombolytic agents, e.g., streptokinase, urokinase, tissue plasminogen activator (TP A) and anisoylated plasminogen-streptokinase activator complex (APS AC), anti -platelets agents, e.g., acetyl-salicylic acid (ASA) and clopidrogel, vasodilating agents, e.g., nitrates, calcium channel blocking drugs, anti -proliferative agents, e.g., colchicine and alkylating agents, intercalating agents, growth modulating factors such as interleukins, transformation growth factor-beta and congeners of platelet derived growth factor, monoclonal antibodies directed against growth factors, anti-inflammatory agents, both steroidal and non-steroidal, and other agents that can modulate vessel tone, function, arteriosclerosis, and the healing response to vessel or organ injury post intervention. Antibiotics can also be included in combinations or coatings comprised by the invention. Moreover, a coating can be used to effect therapeutic delivery focally within the vessel wall. By incorporation of the active agent in a swellable polymer, the active agent will be released upon swelling of the polymer.
In some embodiments, the compounds described herein are formulated or administered in conjunction with liquid or solid tissue barriers also known as lubricants. Examples of tissue barriers include, but are not limited to, polysaccharides, polyglycans, seprafilm, interceed and hyaluronic acid. In some embodiments, medicaments which are administered in conjunction with the compounds described herein include any suitable drugs usefully delivered by inhalation for example, analgesics, e.g. codeine, dihydromorphine, ergotamine, fentanyl or morphine; anginal preparations, e.g. diltiazem; antiallergics, e.g. cromoglycate, ketotifen or nedocromil; anti-infectives, e.g. cephalosporins, penicillins, streptomycin, sulphonamides, tetracyclines or pentamidine; antihistamines, e.g. methapyrilene; anti-inflammatories, e.g. beclomethasone, flunisolide, budesonide, tipredane, triamcinolone acetonide or fluticasone; antitussives, e.g. noscapine;
bronchodilators, e.g. ephedrine, adrenaline, fenoterol, formoterol, isoprenaline, metaproterenol, phenylephrine, phenylpropanolamine, pirbuterol, reproterol, rimiterol, salbutamol, salmeterol, terbutalin, isoetharine, tulobuterol, orciprenaline or (-)-4-amino- 3,5-dichloro-a-[[[6-[2-(2-pyridinyl)ethoxy]hexyl]-amino]methyl]benzenemethanol; diuretics, e.g. amiloride; anticholinergics e.g. ipratropium, atropine or oxitropium; hormones, e.g. cortisone, hydrocortisone or prednisolone; xanthines e.g. aminophylline, choline theophyllinate, lysine theophyllinate or theophylline; and therapeutic proteins and peptides, e.g. insulin or glucagon. It will be clear to a person skilled in the art that, where appropriate, the medicaments are used in the form of salts (e.g. as alkali metal or amine salts or as acid addition salts) or as esters (e.g. lower alkyl esters) or as solvates (e.g. hydrates) to optimize the activity and/or stability of the medicament.
Other exemplary therapeutic agents useful for a combination therapy include but are not limited to agents as described above, radiation therapy, hormone antagonists, hormones and their releasing factors, thyroid and antithyroid drugs, estrogens and progestins, androgens, adrenocorticotropic hormone; adrenocortical steroids and their synthetic analogs; inhibitors of the synthesis and actions of adrenocortical hormones, insulin, oral hypoglycemic agents, and the pharmacology of the endocrine pancreas, agents affecting calcification and bone turnover: calcium, phosphate, parathyroid hormone, vitamin D, calcitonin, vitamins such as water-soluble vitamins, vitamin B complex, ascorbic acid, fat-soluble vitamins, vitamins A, K, and E, growth factors, cytokines, chemokines, muscarinic receptor agonists and antagonists; anticholinesteRASe agents; agents acting at the neuromuscular junction and/or autonomic ganglia; catecholamines, sympathomimetic drugs, and adrenergic receptor agonists or antagonists; and 5 -hydroxytryptamine (5-HT, serotonin) receptor agonists and antagonists.
Therapeutic agents can also include agents for pain and inflammation such as histamine and histamine antagonists, bradykinin and bradykinin antagonists, 5- hydroxytryptamine (serotonin), lipid substances that are generated by biotransformation of the products of the selective hydrolysis of membrane phospholipids, eicosanoids, prostaglandins, thromboxanes, leukotrienes, aspirin, nonsteroidal anti-inflammatory agents, analgesic-antipyretic agents, agents that inhibit the synthesis of prostaglandins and thromboxanes, selective inhibitors of the inducible cyclooxygenase, selective inhibitors of the inducible cyclooxygenase-2, autacoids, paracrine hormones, somatostatin, gastrin, cytokines that mediate interactions involved in humoral and cellular immune responses, lipid-derived autacoids, eicosanoids, β-adrenergic agonists, ipratropium, glucocorticoids, methylxanthines, sodium channel blockers, opioid receptor agonists, calcium channel blockers, membrane stabilizers and leukotriene inhibitors.
Additional therapeutic agents contemplated herein include diuretics, vasopressin, agents affecting the renal conservation of water, rennin, angiotensin, agents useful in the treatment of myocardial ischemia, anti-hypertensive agents, angiotensin converting enzyme inhibitors, β-adrenergic receptor antagonists, agents for the treatment of hypercholesterolemia, and agents for the treatment of dyslipidemia.
Other therapeutic agents contemplated include drugs used for control of gastric acidity, agents for the treatment of peptic ulcers, agents for the treatment of gastroesophageal reflux disease, prokinetic agents, antiemetics, agents used in irritable bowel syndrome, agents used for diarrhea, agents used for constipation, agents used for inflammatory bowel disease, agents used for biliary disease, agents used for pancreatic disease. Therapeutic agents used to treat protozoan infections, drugs used to treat Malaria, Amebiasis, Giardiasis, Trichomoniasis, Trypanosomiasis, and/or
Leishmaniasis, and/or drugs used in the chemotherapy of helminthiasis. Other therapeutic agents include antimicrobial agents, sulfonamides, trimethoprim- sulfamethoxazole quinolones, and agents for urinary tract infections, penicillins, cephalosporins, and other, β-lactam antibiotics, an agent comprising an aminoglycoside, protein synthesis inhibitors, drugs used in the chemotherapy of tuberculosis,
mycobacterium avium complex disease, and leprosy, antifungal agents, antiviral agents including nonretroviral agents and antiretroviral agents.
Examples of therapeutic antibodies that can be combined with a compound of the invention include but are not limited to anti-receptor tyrosine kinase antibodies (cetuximab, panitumumab, tRAStuzumab), anti CD20 antibodies (rituximab, tositumomab), and other antibodies such as alemtuzumab, bevacizumab, and
gemtuzumab.
Moreover, therapeutic agents used for immunomodulation, such as immunomodulators, immunosuppressive agents, tolerogens, and immunostimulants are contemplated by the methods herein. In addition, therapeutic agents acting on the blood and the blood-forming organs, hematopoietic agents, growth factors, minerals, and vitamins, anticoagulant, thrombolytic, and antiplatelet drugs.
For treating renal carcinoma, one may combine a compound of the present invention with sorafenib and/or avastin. For treating an endometrial disorder, one may combine a compound of the present invention with doxorubincin, taxotere (taxol), and/or cisplatin (carboplatin). For treating ovarian cancer, one may combine a compound of the present invention with cisplatin (carboplatin), taxotere, doxorubincin, topotecan, and/or tamoxifen. For treating breast cancer, one may combine a compound of the present invention with taxotere (taxol), gemcitabine (capecitabine), tamoxifen, letrozole, tarceva, lapatinib, PD0325901, avastin, herceptin, OSI-906, and/or OSI-930. For treating lung cancer, one may combine a compound of the present invention with taxotere (taxol), gemcitabine, cisplatin, pemetrexed, Tarceva, PD0325901, and/or avastin.
In other embodiments, agents useful in methods for combination therapy with one or more compounds of structure (I) include, but are not limited to: Erlotinib, Afatinib, Iressa, GDC0941, MLN1117, BYL719 (Alpelisib), BKM120 (Buparlisib), CYT387, GLPG0634, Baricitinib, Lestaurtinib, momelotinib, Pacritinib, Ruxolitinib, TG101348, Crizotinib, tivantinib, AMG337, cabozantinib, foretinib, onartuzumab, NVP-AEW541, Dasatinib, Ponatinib, saracatinib, bosutinib, trametinib, selumetinib, cobimetinib, PD0325901, R05126766, Axitinib, Bevacizumab, Bostutinib, Cetuximab, Crizotinib, Fostamatinib, Gefitinib, Imatinib, Lapatinib, Lenvatinib, Ibrutinib, Nilotinib, Panitumumab, Pazopanib, Pegaptanib, Ranibizumab, Ruxolitinib, Sorafenib, Sunitinib, SU6656, Trastuzumab, Tofacitinib, Vandetanib, Vemurafenib, Irinotecan, Taxol, Docetaxel, Rapamycin or MLN0128.
Further therapeutic agents that can be combined with a compound of the invention are found in Goodman and Gilman's "The Pharmacological Basis of
Therapeutics" Tenth Edition edited by Hardman, Limbird and Gilman or the
Physician's Desk Reference, both of which are incorporated herein by reference in their entirety.
The compounds described herein can be used in combination with the agents disclosed herein or other suitable agents, depending on the condition being treated. Hence, in some embodiments the one or more compounds of the invention will be co-administered with other agents as described above. When used in combination therapy, the compounds described herein are administered with the second agent simultaneously or separately. This administration in combination can include simultaneous administration of the two agents in the same dosage form, simultaneous administration in separate dosage forms, and separate administration. That is, a compound described herein and any of the agents described above can be formulated together in the same dosage form and administered simultaneously. Alternatively, a compound of the invention and any of the agents described above can be
simultaneously administered, wherein both the agents are present in separate
formulations. In another alternative, a compound of the present invention can be administered just followed by and any of the agents described above, or vice versa. In some embodiments of the separate administration protocol, a compound of the invention and any of the agents described above are administered a few minutes apart, or a few hours apart, or a few days apart. The examples and preparations provided below further illustrate and exemplify the compounds of the present invention and methods of preparing such compounds. It is to be understood that the scope of the present invention is not limited in any way by the scope of the following examples and preparations. In the following examples, and throughout the specification and claims, molecules with a single chiral center, unless otherwise noted, exist as a racemic mixture. Those molecules with two or more chiral centers, unless otherwise noted, exist as a racemic mixture of diastereomers. Single enantiomers/diastereomers may be obtained by methods known to those skilled in the art.
EXAMPLES
The following examples are provided for exemplary purposes. Other compounds of structure (I) were prepared according to the following general procedures as indicated in Table 1. EXAMPLE 1
SYNTHESIS OF 3 -( 1 - ACRYLOYLPIPERIDIN-3 -YL)-6-CHLORO-8-FLUORO-7-(2-FLUORO-6- HYDROXYPHENYL UINAZOLIN-4(3H)-ONE (1)
Compound 1 was prepared according to the above synthetic scheme as follows.
3-Amino-2,2'-difluoro-6'-methoxy-[l,l'-biphenyl]-4-carboxylic acid
To a stirred solution of 2-amino-4-bromo-3-fluorobenzoic acid (10 g, 43 mmol) in 1,4-dioxane (400 mL) and H20 (100 mL), 2-fluoro-6-methoxyphenylboronic acid (36 g, 213 mmol), Tetrakis(triphenylphosphine)palladium (2.5 g, 2.15 mmol) and Na2C03 (27g,258mmol) were added. The mixture was degassed and back-filled with N2 several times, and stirred at 100 °C overnight. The mixture was partitioned between water (500 mL) and extracted with ethyl acetate (200 mL X 2). The organic layer was discarded, and 1M HC1 solution was added to the aqueous phase to adjust pH<3. The aqueous phase was extracted with ethyl acetate (200 mL x 2), washed with brine, dried over Na2S04 and concentrated to afford the desired product (1 lg, 92% yield) as a white solid. ESI-MS w/z: 280.1 [M+H]+. 3-Amino-6-chloro-2,2'-difluoro-6'-methoxy-[l,l'-biphenyl]-4-carboxylic acid
To a solution of 3-amino-2,2'-difluoro-6'-methoxy-[l, -biphenyl]-4- carboxylic acid (11 g, 39.6 mmol) in N,N-dimethylformamide (100 mL) at RT, N- chlorosuccinimde (5.27 g, 39.6 mmol) was added. The resulting mixture was stirred at 100 °C for 1 h. The mixture was allowed to cool to RT, and the reaction mixture was slowly added to water (300 mL), the mixture was filtered and the cake was dried to afford the desired product (11.5 g, 93.1% yield) as a brown solid.
6-Chloro-8-fluoro-7-(2-fluoro-6-methoxyphenyl)quinazolin-4-ol
A mixture of 3-amino-6-chloro-2,2'-difluoro-6'-methoxy-[l, -biphenyl]- 4-carboxylic acid (8.1 g, 25.8 mmol) in EtOH (150 mL), formimidamide acetate (35 g, 336.4 mmol) was added. The mixture was stirred at 100 °C overnight. The reaction mixture was concentrated and water was added. The mixture was filtered and the cake was dried to afford the desired product (7.5 g, 90.3% yield) as a light yellow solid. tert-Butyl3-(6-chloro-8-fluoro-7-(2-fluoro-6-methoxyphenyl)-4-oxoquinazolin- 3(4H)-yl)piperidine-l-carboxylate
To a solution of 6-chloro-8-fluoro-7-(2-fluoro-6- methoxyphenyl)quinazolin-4(3H)-one (1.0 g, 3.1 mmol) in MeCN (20 mL), HATU (4.7 g, 12.4 mmol), DBU (1.9 g, 12.4 mmol) and tert-butyl3-aminopiperidine-l-carboxylate (2.3 g, 12.4 mmol) were added and the resulting mixture was stirred at RT for 48 h. The mixture was partitioned between ethyl acetate and water. The organic layer was washed with brine, dried over with Na2S04, filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel (ethyl acetate/ petroleum ether = 1 :5 to 1 :3) to give the product (670 mg, 42.7% yield). ESI-MS m/z: 506.2 [M+H]+. 6-Chloro-8-fluoro-7-(2-fluoro-6-methoxyphenyl)-3-(piperidin-3-yl)quinazolin- 4(3H)-one
To a solution of tert-butyl 3-(6-chloro-8-fluoro-7-(2-fluoro-6- methoxyphenyl)-4-oxoquinazolin-3(4H)-yl)piperidine-l-carboxylate (230 mg, 0.57 mmol) in dichlorom ethane (10 mL), TFA (4 mL) was added and the resulting mixture was stirred at RT for 1 h. The mixture was concentrated in vacuo. The residue was partitioned between water and ethyl acetate. The organic layer was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue (230 mg) was dissolved in DCM (10 mL), BBr3 (1.4 g, 5.7 mmol) was added at -78 °C and the mixture was stirred at RT for 3 h. The mixture was quenched with saturated NaHC03 solution and extracted with ethyl acetate. The organic layer was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel (MeOH/DCM = 1 :30) to give the product (150 mg, 67% yield). ESI-MS m/z: 392.1 [M+H]+. 3-(l-Acryloylpiperidin-3-yl)-6-chloro-8-fluoro-7-(2-fluoro-6- hydroxyphenyl)quinazolin-4(3H)-one
To a solution of 6-chloro-8-fluoro-7-(2-fluoro-6-methoxyphenyl)-3- (piperidin-3-yl)quinazolin-4(3H)-one (150 mg, 0.38 mmol) and triethylamine (0.3 mL, 1.9 mmol) in DCM (10 mL) at 0 °C, acryloyl chloride (70 mg, 0.77 mmol) was added and the resulting mixture was stirred for 20 min. The mixture was quenched with saturated NaHC03 solution and partitioned between water and dichloromethane. The organic layer was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was dissolved in THF (7 mL) and H20 (7 mL). Lithium hydroxide hydrate (70 mg, 1.67 mmol) was added and the mixture was stirred at RT for 40 min. The pH of mixture was adjusted to 7 and extracted with ethyl acetate. The organic layer was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by Pre-TLC plate (MeOH/DCM =1 :20) to give the product (42 mg, 22.6 % yield). ESI-MS m/z: 446.3 [M+H]+. 1H- MR (400 MHz, DMSO^d) δ: 10.36 (s, 1H), 8.59 (s, 1H), 8.10 (s, 1H), 7.41-7.35 (m, 1H), 6.90-6.75 (m, 3H), 6.16 (d, J= 16.7 Hz, 1H), 5.73 (dd, J = 2.1, 10.6 Hz, 1H), 4.61-4.48 (m, 2H), 4.19 (dd, J= 12.2, 71.5 Hz, 1H), 3.54-2.65 (m, 1H), 3.20-3.09 (m, 1H), 2.28-2.16 (m, 1H), 2.04-1.88 (m, 2H), 1.59-1.53 (m, 1H).
EXAMPLE 2
SYNTHESIS OF N- 3-(6-CHLORO-8-FLUORO-7-(2-FLUORO-6-HYDROXYPHENYL)-4-
Compound 2 was prepared according to the above synthetic scheme as follows. tert-Butyl 3-(6-chloro-8-fluoro-7-(2-fluoro-6-methoxyphenyl)-4-oxoquinazolin- 3(4H)-yl)cyclobutylcarbamate
To a solution of 6-chloro-8-fluoro-7-(2-fluoro-6- methoxyphenyl)quinazolin-4(3H)-one (0.6 g, 1.86 mmol) in MeCN (30 mL), HATU (1.4 g, 3.72 mmol), DBU (1.7 g, 11.16 mmol) and tert-butyl 3- aminocyclobutylcarbamate (0.415 g, 2.23 mmol) were added and the resulting mixture was stirred at RT for 48 h. The mixture was partitioned between ethyl acetate and water. The organic layer was washed with brine, dried over with Na2S04, filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel (ethyl acetate/ petroleum ether = 1 :3) to give the product (200 mg, 22% yield). ESI-MS m/z: 491.1 [M+H]+. N-(3-(6-Chloro-8-fluoro-7-(2-fluoro-6-hydroxyphenyl)-4-oxoquinazolin-3(4H)- yl)cyclobutyl)acrylamide
To a solution of tert-butyl 3-(6-chloro-8-fluoro-7-(2-fluoro-6- methoxyphenyl)-4-oxoquinazolin-3(4H)-yl)cyclobutylcarbamate (200 mg, 0.40 mmol) in DCM (5 mL), TFA (2 mL) was added and the resulting mixture was stirred at RT for 1 h. The mixture was concentrated in vacuo. The residue was dissolved in DCM (3 mL), BBr3 (1 mL, 10.79 mmol) was added at -78°C and the mixture was stirred at RT for 2 h. The mixture was quenched at -78°C with MeOH and saturated NaHC03 solution, and then was extracted with mixture solvent (DCM:MeOH= 10: 1). The organic layer was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was dissolved in DCM (2 mL), Et3N (162 mg, 1.6 mmol) and acryloyl chloride (55 mg, 0.6 mmol) were added at 0 °C and the resulting mixture was stirred for 20 min. The mixture was quenched with saturated NaHC03 solution and partitioned between water and DCM. The organic layer was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was dissolved in THF (3 mL) and H20 (2 mL).
Lithium hydroxide hydrate (42 mg, 1.2 mmol) was added and the mixture was stirred at RT for 40 min. The pH of mixture was adjusted to 6 and extracted with ethyl acetate. The organic layer was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by Pre-TLC plate (MeOH/dichloromethane = 1 :20) to give the product (31 mg, 18 % yield for 4steps). ESI-MS m/z: 431.1 [M+H]+. 1H NMR (400
MHz, DMSO-i¾) 5: 10.35 (s, 1H), 8.75 (d, J= 6.7 Hz, 1H), 8.61 (s, 1H), 8.07 (d, J= 1.4 Hz, 1H), 7.40-7.34 (m, 1H), 6.87-6.79 (m, 2H), 6.30-6.23 (m, 1H), 6.15-6.10 (m, 1H), 5.63 (dd, J= 2.1, 9.9Hz, 1H), 5.30-5.22 (m, 1H), 4.39-4.33 (m, 1H), 2.91-2.84 (m, 2H), 2.48-2.44 (m, 2H).
EXAMPLE 3
SYNTHESIS OF N-(4-(4-ACETYLPIPERAZIN- 1 -YL)-6-CHLORO-8-FLUORO-7-(2-FLUORO-6- HYDROXYPHENYL)QUINOLIN-3 -YL) ACRYL AMIDE (3)
follows.
3-bromo-2-fluorobenzenamine
To a mixture of l-bromo-2-fluoro-3 -nitrobenzene (13.75 g, 62.76 mmol), HO Ac (26.36 g, 439 mmol), EtOH (150 mL) and H20 (60 mL) at RT, iron powder (9.14 g, 163 mmol) was added in portions. The resulting mixture was stirred at RT for 16 h and then was neutralized with NaOH (5 N) solution. The mixture was extracted with ethyl acetate. The organic layer was washed with brine, dried over Na2S04 and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel (petroleum ether / ethyl acetate = 10: 1) to afford the desired product (7.77 g, 65% yield) as a brown oil.
3-Bromo-4-chloro-2-fluorobenzenamine
To a solution of 3-bromo-2-fluorobenzenamine (1.9 g, 10 mmol) in
DMF (10 mL) at RT, was added NCS (1.4 g, 10.5 mmol) and the resulting mixture was stirred at RT for 16 h. The mixture was poured into ice water and extracted with ethyl acetate. The organic layer was washed with brine, dried over Na2S04 and concentrated in vacuo. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate = 30: 1) to afford the desired product (1.15 g, 51% yield). ESI- MSm/z: 225.9 [M + H]+.
Diethyl 2-((3-bromo-4-chloro-2-fluorophenylamino)methylene)malonate
A mixture of 3-bromo-4-chloro-2-fluorobenzenamine (2.3 g, 10.2 mmol) and diethyl2-(ethoxymethylene)malonate (2.42 g, 11.22 mmol) was stirred at 120 °C for 3 h. The mixture was allowed to cool to RT. Petroleum was added and stirred at RT for 1 h. The precipitate was filtered and dried to afford the desired product (2.76 g, 68.7% yield). ESI-MS m/z: 395.9[M+H]+.
Ethyl 7-bromo-6-chloro-8-fluoro-4-hydroxyquinoline-3-carboxylate
Diethyl 2-((3-bromo-4-chloro-2-fluorophenylamino)methylene)malonate (2.76 g, 6.99 mmol) was suspended in Ph20 (20 mL).The mixture was stirred at 250 °C for 2 h. The mixture was allowed to cool to RT and then 100 mL of petroleum ether was added. The white solid was collected by filtration and rinsed with petroleum ether (100 mL) to afford the desired product (1.85 g, 76% yield). ESI-MS m/z: 349.9 [M + H]+. Ethyl 7-bromo-4,6-dichloro-8-fluoroquinoline-3-carboxylate
A mixture of ethyl 7-bromo-6-chloro-8-fluoro-4-hydroxyquinoline-3- carboxylate (1.85 g, 5.31 mmol) and POCl3 (10 mL) was stirred at reflux for 4 h. The mixture was allowed to cool to RT and concentrated in vacuo to afford the crude product (1.41 g) which was used directly in the next step.
Ethyl4-(4-(tert-butoxycarbonyl)piperazin-l-yl)-7-bromo-6-chloro-8- fluoroquinoline-3-carboxylate
A mixture of ethyl 7-bromo-4,6-dichloro-8-fluoroquinoline-3- carboxylate (1.41 g, 3.84 mmol), tert-butyl piperazine-l-carboxylate (1.43 g, 7.68 mmol), Et3N (1.55 g, 15.36 mmol) in DMSO (20 mL) was stirred at 80 °C under argon for 2 h. The mixture was cooled to RT, poured into ice-water, and extracted with ethyl acetate. The combined organic layer was washed with brine, dried over Na2S04, and concentrated in vacuo. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate = 3 : 1) to afford the desired product (1.96 g, 98% yield). ESI-MS m/z: 518.1 [M+H]+.
Ethyl 4-(4-(tert-butoxycarbonyl)piperazin-l-yl)-6-chloro-8-fluoro-7-(2-fluoro-6- methoxyphenyl)quinoline-3-carboxylate
A mixture of ethyl4-(4-(tert-butoxycarbonyl)piperazin-l-yl)-7-bromo-6- chloro-8-fluoroquinoline-3-carboxylate (1.13 g, 2.19 mmol) and (2-fluoro-6- methoxyphenyl)boronic acid (1.86 g, 10.93 mmol) in Na2C03 aqueous solution (3.64 mL, 1.5 M) and Dioxane (80 mL), was added Pd(PPh)4 under Ar. The mixture was stirred at 90°C for 16 h. The solvent was removed and the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate = 6: 1) to afford the desired product (851 mg, 69% yield). ESI-MS m/z: 562.2 [M+H]+.
Ethyl 4-(4-acetylpiperazin-l-yl)-6-chloro-8-fluoro-7-(2-fluoro-6- methoxyphenyl)quinoline-3-carboxylate
A mixture of ethyl 4-(4-(tert-butoxycarbonyl)piperazin-l-yl)-6-chloro-8- fluoro-7-(2-fluoro-6-methoxyphenyl)quinoline-3-carboxylate (851 mg, 1.52 mmol) in DCM (20 mL) at RT, was added TFA (4 mL) and the resulting mixture was stirred for 2 h. The mixture was added to a stirred mixture of 2M NaOH (40mL) and ethyl acetate (40 mL). Then acetyl chloride (4 mL) was slowly added to the reaction. The mixture was extracted with ethyl acetate. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate = 1 : 1) to afford the desired product (575 mg, 75% yield). ESI-MS m/z: 504.3 [M+H]+.
4-(4-Acetylpiperazin-l-yl)-6-chloro-8-fluoro-7-(2-fluoro-6- methoxyphenyl)quinoline-3-carboxylic acid
A mixture of ethyl 4-(4-acetylpiperazin-l-yl)-6-chloro-8-fluoro-7-(2- fluoro-6-methoxyphenyl)quinoline-3-carboxylate (267 mg, 053 mmol) in THF (20 mL) and water (20 mL), was added lithium hydroxide (111 mg, 2.651 mmol) and the resulting mixture was stirred at RT for 16 h. The mixture was diluted with 2M NaOH (30 mL), and extracted with 50% ethyl acetate/petroleum ether. The water layer was acidified by 1M HCl and extracted with ethyl acetate. The solvent was removed to afford the product (277 mg, crude). ESI-MS m/z: 476.2 [M+H]+. l-(4-(3-Amino-6-chloro-8-fluoro-7-(2-fluoro-6-methoxyphenyl)quinolin-4- yl)piperazin-l-yl)ethanone
A mixture of 4-(4-acetylpiperazin-l-yl)-6-chloro-8-fluoro-7-(2-fluoro-6- methoxyphenyl)quinoline-3-carboxylic acid (277 mg, 0.58 mmol) and Et3N (235 mg, 2.33 mmol) in DMF (20 mL) and t-BuOH (3 mL), was added DPPA (401 mg, 1.456 mmol) and the resulting mixture was stirred at 120 °C for 2.5 h. The mixture was allowed to cool to RT and quenched with water (50 mL). The mixture was extracted with ethyl acetate and concentrated in vacuo. The residue was dissolved in a mixture of DCM (10 mL) and TFA (3 mL), and then stirred for 1 h. The mixture was added to 2M NaOH (30 mL) and extracted with ethyl acetate. The combined organic layer was washed with brine, dried over Na2SC>4, and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM/MeOH = 30: 1) to afford the desired product (120 mg, 46% yield). ESI-MS m/z: 447.2 [M+H]+. l-(4-(3-Amino-6-chloro-8-fluoro-7-(2-fluoro-6-hydroxyphenyl)quinolin-4- yl)piperazin-l-yl)ethanone
A mixture of l-(4-(3-amino-6-chloro-8-fluoro-7-(2-fluoro-6- methoxyphenyl)quinolin-4-yl)piperazin-l-yl)ethanone (120 mg, 0.27 mmol) in DCM (10 mL) at -78 °C, was added BBr3 (674 mg, 2.69 mmol) and the resulting mixture was stirred at RT for 2 h. The mixture was poured to ice-water, and partitioned between ethyl acetate and NaHC03 solution. The organic layer was dried over Na2S04 and concentrated in vacuo to afford the desired product (194 mg, crude). ESI-MS m/z: 433.2 [M+H]+. N-(4-(4-Acetylpiperazin-l-yl)-6-chloro-8-fluoro-7-(2-fluoro-6- hydroxyphenyl)quinolin-3-yl)acrylamide
A mixture of l-(4-(3-amino-6-chloro-8-fluoro-7-(2-fluoro-6- hydroxyphenyl)quinolin-4-yl)piperazin-l-yl)ethanone (194 mg, 0.45 mmol) and Et3N (181 mg, 1.80 mmol) in DCM (20 mL) and THF (20 mL), was added acryloyl chloride (162 mg, 1.796 mmol) and the resulting mixture was stirred at 0 °C for 30 min. The mixture was partitioned between DCM and NaHC03 solution. The organic layer was dried over Na2S04 and concentrated in vacuo. The residue was dissolved in THF (10 mL) and water (10 mL), and lithium hydroxide (75 mg, 1.794 mmol) was added. The mixture was stirred at RT for 1 h. The mixture was adjusted pH to 8 with IN HCl and NaHC03 solution, and then extracted with ethyl acetate. The organic layer was dried over Na2S04 and concentrated in vacuo. The residue was purified by flash
chromatography on silica gel (DCM/MeOH = 30: 1) to afford the desired product (32 mg, 24% yield for two steps). ESI-MS m/z: 487.3 [M+H]+. 1H-NMR (400 MHz, OMSO-d6) δ: 10.26 (s, 1H), 10.06 (s, 1H), 8.70 (s, 1H), 8.04 (s, 1H), 7.39 (t, J7 = 7.6 Hz, J2 = 15.2 Hz, 1H), 6.88 (m, 2H), 6.61 (m, 1H), 6.36 (m, 1H), 5.88 (m, 1H), 3.69 (m, 4H), 3.29 (m, 4H), 2.07 (s, 3H).
EXAMPLE 4
SYNTHESIS OF 3 -( 1 - ACRYLOYL AZETIDIN-3 -YL)-6-CHLORO-8-FLUORO-7-(2-FLUORO-6- HYDROXYPHENYL QUINAZOLIN-4(3H)-ONE (4)
Compound 4 was prepared according to the above synthetic scheme as follows. tert-Butyl 3-(6-chloro-8-fluoro-7-(2-fluoro-6-methoxyphenyl)-4-oxoquinazolin- 3(4H)-yl)azetidine-l-carboxylate
To a solution of 6-chloro-8-fluoro-7-(2-fluoro-6- methoxyphenyl)quinazolin-4(3H)-one (1 g, 3.1 mmol) in MeCN (20 mL), HATU (3.53 g, 9.32 mmol), DBU (4.73 g, 31.06 mmol) and tert-butyl 3-aminoazetidine-l- carboxylate (2.14 g, 12.42 mmol) were added and the resulting mixture was stirred at RT for 16 h. The mixture was partitioned between ethyl acetate and water. The organic layer was washed with brine, dried over with Na2SC"4, filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel (ethyl acetate/ petroleum ether = 1 : 10 to 1 :2) to give the product (500 mg, 34% yield). ESI- MS m/z: 478.2 [M+H]+.
3-(Azetidin-3-yl)-6-chloro-8-fluoro-7-(2-fluoro-6-methoxyphenyl)quinazolin-4(3H)- one
To a solution of tert-butyl 3-(6-chloro-8-fluoro-7-(2-fluoro-6- methoxyphenyl)-4-oxoquinazolin-3(4H)-yl)azetidine-l-carboxylate (400 mg, 0.84 mmol) in DCM (10 mL), TFA (4 mL) was added and the resulting mixture was stirred at RT for 1 h. The mixture was concentrated in vacuo. The residue was partitioned between water and ethyl acetate. The organic layer was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue (220 mg) was dissolved in DCM (20 mL), BBr3 (0.2 mL, 2.16 mmol) was added at -78 °C and the resulting mixture was stirred at RT for 1.5 h. The mixture was quenched with saturated NaHC03 solution and extracted with ethyl acetate. The organic layer was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by Pre-TLC plate on silica gel (MeOH/DCM = 1 : 10) to give the product (42 mg, 39% yield for 2 steps). ESI-MS m/z: 364.1 [M+H]+. 3-(l-Acryloylazetidin-3-yl)-6-chloro-8-fluoro-7-(2-fluoro-6- hydroxyphenyl)quinazolin-4(3H)-one
To a solution of 3-(azetidin-3-yl)-6-chloro-8-fluoro-7-(2-fluoro-6- methoxyphenyl)quinazolin-4(3H)-one (130 mg, 0.36 mmol) and triethylamine (217 mg, 2.15 mmol) in DCM (10 mL) at -78 °C, acryloyl chloride (65 mg, 0.72 mmol) was added and the mixture was stirred -60 °C for 20 min. The mixture was quenched with saturated NaHC03 solution and partitioned between water and DCM. The organic layer was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was dissolved in THF (7 mL) and H20 (7 mL). Lithium hydroxide hydrate (360 mg, 8.6 mmol) was added and the resulting mixture was stirred at RT for 40 min. The pH of mixture was adjusted to 6 and then extracted with ethyl acetate. The organic layer was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by Pre-TLC plate (MeOH/DCM = 1 : 15) to afford the product (42 mg, 29 % yield). ESI-MS m/z: 418.1 [M+H]+. 1HNMR (400MHz, DMSO-i¾) 5: 10.36 (s, 1H), 8.53 (s, 1H), 8.09 (s, 1Η),7.40-7.34 (m , 1H), 6.87-6.80 (m ,2H), 6.40-6.33 (m, 1H), 6.18-6.13 (m,lH), 5.76-5.71(m, 1H), 5.34-5.30 (m, 1H), 4.68-4.64 (m, 2H), 4.47-4.44 (m, lH), 4.43-4.3 l(m,lH). EXAMPLE 5
SYNTHESIS OF 3-(1-ACRYLOYLAZETIDIN-3-YL)-6-AMINO-8-CHLORO-7-
HYDROXYNAPHTHALEN- 1 -YL QUINAZ0LIN-4(3H)-0NE (12)
2-Bromo-6-nitroaniline
A mixture of l-bromo-2-fluoro-3 -nitrobenzene (20.0 g, 91.37 mmol) and H3 in CH3OH (7M, 60 mL) was stirred at 100 °C in a sealed tube for 16 h. The solvent was removed and the residue was dissolved in H20, extracted with ethyl acetate. The organic layer was washed with brine, dried over Na2S04 and concentrated in vacuo. The residue was purified by column chromatography eluting with (ethyl acetate/petroleum ether = 1 : 100) to afford the desired product as a yellow solid (16.0 g, 81.2% yield). l-Bromo-2-chloro-3-nitrobenzene
A mixture of 2-bromo-6-nitroaniline (16.0 g, 74.10 mmol), tert-butyl nitrite (11.40 g 111.15mmol) and CuCl2 (12.0 g, 88.90 mmol) in CH3CN (160 mL) was stirred at 60 °C under argon for 1 h. The mixture was allowed to cool to RT, quenched with H20, and extracted with ethyl acetate. The organic layer was washed with brine, dried over Na2S04 and concentrated in vacuo. The residue was purified by column chromatography on silica (ethyl acetate/petroleum ether = 1 : 100) to yield the product (15.0 g, 86.2% yield). 3-Bromo-2-chlorobenzenamine
To a mixture of l-bromo-2-chloro-3 -nitrobenzene (15.0 g, 63.60 mmol), HOAc (20 mL), EtOH (120 mL) and H20 (40 mL) at RT, iron powder (10.7 g, 190.70 mmol) was added in portions. The resulting mixture was stirred at RT for 16 h and then was neutralized with NaOH (5 N) solution. The mixture was extracted with ethyl acetate. The organic layer was washed with brine, dried over Na2S04 and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel (petroleum ether/ethyl acetate = 10: 1) to afford the desired product (14.0 g, 100% yield). (E)-N-(3-Bromo-2-chlorophenyl)-2-(hydroxyimino)acetamide
A mixture of 2,2,2-trichloroethane-l,l-diol (13.5 g, 82.00 mmol) and Na2S04 (87.3 g, 614.70 mmol) in water was warmed to 35°C. 3-bromo-2- chlorobenzenamine (14.0 g, 68.30 mmol) in water was added, followed by 35% aqueous HC1 (30 mL) and hydroxylamine hydrochloride (14.2 g, 204.90 mmol). The resulting mixture was stirred at 90°C for 16 h and yellow precipitate was formed. The mixture was allowed to cool to RT. The solid was filtered, washed with water and dried in the air to afford the desired product (14.5 g, 76.3% yield).
6-Bromo-7-chloroindoline-2,3-dione
To the concentrated sulfuric acid (120 mL) at 60 °C, was added (E)-N- (3-bromo-2-chlorophenyl)-2-(hydroxyimino)acetamide (14.5 g, 52.25 mmol). The temperature was raised to 90 °C and maintained for 3 h. The reaction mixture was allowed to cool to RT and poured into ice to get yellow precipitate. The precipitate was collected by filtration and dried to afford the desired product (7.4 g, 54.4% yield).
2-Amino-4-bromo-3-chlorobenzoic acid
To a solution of 6-bromo-7-chloroindoline-2,3-dione (7.4 g, 28.57 mmol) in 2 N NaOH (200 mL) at 0 °C, was added H202 (30%, 20 mL) and the resulting mixture was stirred at 0 °C for 30 min and then at RT for 16 h. The mixture was poured into ice-water and the solution was acidified with cone. HC1. The precipitate was collected by filtration and dried in the air to afford the desired product (2.5 g, 35.2% yield).
7-Bromo-8-chloroquinazolin-4(3H)-one
To a solution of 2-amino-4-bromo-3-chlorobenzoic acid (2.5 g, 10.00 mmol) in 2-methoxyethanol (50 mL) at RT, was added formamidine acetate (6.2 g, 60.00 mmol) and the resulting mixture was stirred at reflux for 16 h. The mixture was concentrated in vacuo. The residue was partitioned between water and ethyl acetate. The organic layer was washed with brine, dried over Na2S04 and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM/methanol = 100: 1 to 50: 1) to afford the desired product (1.8 g, 69.2% yield). ESI-MSm/z: 258.8 [M+H]+.
7-Bromo-8-chloro-6-nitroquinazolin-4(3H)-one
To a solution of 7-bromo-8-chloroquinazolin-4(3H)-one (1.8 g, 6.95 mmol) in H2S04 (20 mL) at RT, was added KN03 (1.1 g, 10.42mmol) and the mixture was stirred at 100 °C for 16 h. The mixture was poured into ice-water. The precipitate was collected by filtration, washed with water and dried to afford the desired product (1.5 g, 71.4% yield). ESI-MS m/z: 303.9 [M+H]+. tert-Butyl3-(7-bromo-8-chloro-6-nitro-4-oxoquinazolin-3(4H)-yl)azetidine-l- carboxylate
To a solution of 7-bromo-8-chloro-6-nitroquinazolin-4(3H)-one (1.5 g,
4.93 mmol) in MeCN (20 mL), HATU (3.75 g, 9.86 mmol), DBU (4.5 g, 29.58 mmol) and tert-butyl 3-aminoazetidine-l-carboxylate (1.3 g, 7.40 mmol) were added and the resulting mixture was stirred at RT for 2 h. The mixture was partitioned between ethyl acetate and water. The organic layer was washed with brine, dried over with Na2S04, filtered and concentrated in vacuo. The residue was purified by flash column
chromatography on silica gel (ethyl acetate/ petroleum ether = 1 :5 to 1 :3) to afford the product (1.0 g, 43.5% yield). ESI-MS m/z: 459.0 [M+H]+. tert-Butyl3-(6-amino-7-bromo-8-chloro-4-oxoquinazolin-3(4H)-yl)azetidine-l- carboxylate
To a mixture of tert-butyl3-(7-bromo-8-chloro-6-nitro-4-oxoquinazolin- 3(4H)-yl)azetidine-l-carboxylate (300 mg, 0.66 mmol), HOAc(2 mL), EtOH (12 mL) and H20 (4 mL) at RT, iron powder (183.4 mg, 3.28 mmol) was added in portions and the resulting mixture was stirred at RT for 16 h. The mixture was neutralized with NaOH (2 N) solution and then was extracted with ethyl acetate. The organic layer was washed with brine, dried over Na2S04 and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel (petroleum ether/ethyl acetate = 10: 1) to afford the desired product (283 mg, 100% yield). tert-Butyl3-(6-amino-8-chloro-7-(2-hydroxynaphthalen-4-yl)-4-oxoquinazolin- 3(4H)-yl)azetidine-l-carboxylate
A mixture of tert-butyl3-(6-amino-7-bromo-8-chloro-4-oxoquinazolin- 3(4H)-yl)azetidine-l-carboxylate (283 mg, 0.66 mmol), 3-hydroxynaphthalen-l-yl-l- boronic acid (142.1 mg, 0.66 mmol), Pd(PPh3)4 (76.2 mg, 0.1 mmol) and Na2C03
(209.9 mg, 1.98 mmol ) in l,4-dioxane/H20 (16 mL/4 mL) was stirred at 100 °C under argon for 16 h. The mixture was allowed to cool to RT and concentrated in vacuo. The residue was purified by column chromatography on silica gel to afford the desired product as a white solid (150 mg, 46.2% yield). 6-Amino-3-(azetidin-3-yl)-8-chloro-7-(2-hydroxynaphthalen-4-yl)quinazolin- 4(3H)-one
To a solution of tert-butyl3-(6-amino-8-chloro-7-(2-hydroxynaphthalen- 4-yl)-4-oxoquinazolin-3(4H)-yl)azetidine-l-carboxylate (150 mg, 0.30 mmol) in DCM (16 mL), TFA (4 mL) was added and the resulting mixture was stirred at RT for 1 h. The mixture was concentrated in vacuo. The residue was partitioned between water and ethyl acetate. The organic layer was dried over anhydrous Na2S04, filtered and concentrated in vacuo to afford the desired product (80 mg). 3-(l-Acryloylazetidin-3-yl)-6-amino-8-chloro-7-(2-hydroxynaphthalen-4- yl)quinazolin-4(3H)-one
A mixture of 6-amino-3-(azetidin-3-yl)-8-chloro-7-(2- hydroxynaphthalen-4-yl)quinazolin-4(3H)-one (80 mg, 0.20 mmol) and NaOH (5 mL, 2N) in THF (20 mL) at 0 °C, acryloyl chloride (18 mg, 0.20 mmol) was added and the resulting mixture was stirred at 0 °C for 30 min. The mixture was partitioned between ethyl acetate and NaHC03 solution. The organic layer was dried over Na2S04 and concentrated in vacuo. The residue was purified by flash chromatography on silica gel (DCM/MeOH = 30: 1) to afford the desired product (18 mg, 20.2% yield). ESI-MS m/z: 447.1 [M+H]+. 1H NMR (400 MHz, DMSO-i¾) δ: 8.31 (s, 1H), 8.02(d, J= 8.0 Hz,
1H), 7.78 (d, J= 8.0 Hz, 1H), 7.47-7.43 (m, 1H), 7.35-7.33 (m, 1H), 7.26-7.21 (m, 2H), 7.10 (d, J= 2.8 Hz, 1H), 6.44 (dd, J= 1.6, 17.2 Hz, 1H), 6.24 (dd, J= 10.4, 17.2 Hz, 1H), 5.79 (dd, J= 1.2, 10.4 Hz, 1H), 5.43-5.30 (m, 1H), 4.78-4.56 (m, 4H).
EXAMPLE 6
SYNTHESIS OF 3-(1-ACRYLOYLAZETIDIN-3-YL)-8-CHLORO-5-HYDROXY-7-(3- HYDROXYNAPHTHALEN-1-YL UINAZOLIN-4(3H)-ONE (14)
2- Bromo-4-methoxy-6-nitrobenzenamine
To a solution of 4-methoxy-2-nitrobenzenamine (6.72 g, 40 mmol) in DCM (70 mL) at -20 °C, bromine (2.50 mL, 48 mmol) was added dropwise. The mixture was stirred at -20 °C for 30 min. The mixture was poured into ice-water. The pH was adjusted to 8 with saturated sodium bicarbonate aqueous solution and then extracted with DCM. The organic layer was washed with brine, dried over Na2S04 and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM/petroleum ether = 1 :2) to yield the product (6.42 g, 65% yield). ESI-MS m/z: 247.0 [M +H]+. l-Bromo-2-chloro-5-methoxy-3-nitrobenzene
To a solution of t-Butyl nitrite (3.75 g, 36.4 mmol) and CuCl2 (3.92 g, 29.2 mmol) in acetonitrile (120 mL) at 60 °C, was added 2-bromo-4-methoxy-6- nitrobenzenamine (6.0 g, 24.3 mmol) and the resulting mixture was stirred at 60 °C overnight. The mixture was concentrated in vacuo. The residue was dissolved in DCM (100 mL), washed with brine, dried over Na2S04 and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM/petroleum ether = 1 :3) to yield the product (5.01 g, 77% yield).
3- Bromo-2-chloro-5-methoxybenzenamine
To a mixture of l-bromo-2-chloro-5-methoxy-3-nitrobenzene (5.01 g, 18.8 mmol) and HO Ac (6 mL) in EtOH (36 mL) and H20 (12 mL) at RT, iron powder (3.14 g, 56.3mmol) was added portion-wise. The resulting mixture was stirred at RT for 16 h and then was neutralized with NaOH (5 N) solution. The mixture was extracted with ethyl acetate. The organic layer was washed with brine, dried over Na2S04 and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel (DCM/petroleum ether = 1 :3) to afford the desired product (3.90 g, 88% yield) as a brown oil. ESI-MSw/z: 237.9 [M+H]+.
6-Bromo-7-chloro-4-methoxyindoline-2,3-dione
To a solution of 3-bromo-2-chloro-5-methoxybenzenamine (3.9 g, 16.5 mmol) in 1,2-dichlorom ethane (50 mL) at 0 °C, oxalyl chloride (2.7 g, 21.4 mmol) was added and the resulting mixture was allowed to warm to RT and stirred at RT for 4 h. The mixture was stirred at 55 °C for 30 min and cooled back down to 0 °C. A1C13 (2.8 g, 21.4 mmol) was added in portions. The mixture was stirred at 55 °C overnight. The mixture was diluted with DCM (50 mL), washed with brine, dried over Na2SC"4 and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM/petroleum ether = 1 :3) to yield the product (3.1g, 65% yield). ESI-MS m/z: 291.9 [M+H]+.
2-Amino-4-bromo-3-chloro-6-methoxybenzoic acid
To a solution of 6-bromo-7-chloro-4-methoxyindoline-2,3-dione (3.1 g, 10.7 mmol) in 2 N NaOH (80 mL) was added H202 (30%, 8 mL) at 0°C, the mixture was stirred at 0°C for 30 min. After stirring at room temperature for 16 h, the mixture was poured into ice water, the solution was acidified with Cone. HC1, the precipitate was filtered and dried in the air to afford the desired product as a white solid (1.6 g, 53% yield). ESI-MSw/z: 278.9 [M +H]+. 7-Bromo-8-chloro-5-methoxyquinazolin-4(3H)-one
To a solution of 2-amino-4-bromo-3-chloro-6-methoxybenzoic acid (1.6 g, 5.7 mmol) in ethanol (25 mL) at RT, formamidine acetate (3.6 g, 34.3 mmol) was added and the resulting mixture was stirred at reflux for 16 h. The mixture was concentrated in vacuo and the residue was diluted with water. The aqueous solution was extracted with ethyl acetate. The organic layer was washed with brine, dried over Na2SC"4 and concentrated in vacuo. The residue was purified by column
chromatography on silica gel (DCM/methanol = 100: 1 to 50: 1) to afford the desired product (700 mg, 42% yield). tert-Butyl 3-(7-bromo-8-chloro-5-methoxy-4-oxoquinazolin-3(4H)-yl)azetidine-l- carboxylate
To a solution of 7-bromo-8-chloro-5-methoxyquinazolin-4(3H)-one (630 mg, 2.18 mmol) in MeCN (15 mL), HATU (1.66 g, 4.36 mmol), DBU (1.99 g, 13.08 mmol) and tert-butyl 3 -aminoazetidine-1 -carboxylate (562 mg, 3.27 mmol) were added and the resulting mixture was stirred at 80 °C overnight. The mixture was partitioned between ethyl acetate and water. The organic layer was washed with brine, dried over with Na2S04, filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel (ethyl acetate/ petroleum ether = 1 : 1) to afford the product (360 mg, 37% yield). ESI-MSw/z: 446.0 [M +H]+. tert-Butyl 3-(8-chloro-7-(2-hydroxynaphthalen-4-yl)-5-methoxy-4-oxoquinazolin- 3(4H)-yl)azetidine-l-carboxylate
A mixture of tert-butyl 3-(7-bromo-8-chloro-5-methoxy-4- oxoquinazolin-3(4H)-yl)azetidine-l-carboxylate (300 mg, 0.67 mmol), 3- hydroxynaphthalen-l-yl-l-boronic acid (127 mg, 0.67 mmol), Pd(PPh3)4 (81 mg, 0.07 mmol) and Na2C03 (142 mg, 1.34 mmol ) in l,4-dioxane/H20 (8 mL/2 mL) was stirred under argon at 100 °C overnight. The mixture was allowed to cool to RT and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel (ethyl acetate/ petroleum ether = 1 :2) to afford the product (221 mg, 65% yield). ESI-MS m/z: 508.2 [M+H]+. 3-(Azetidin-3-yl)-8-chloro-7-(2-hydroxynaphthalen-4-yl)-5-methoxyquinazolin- 4(3H)-one
To a solution of tert-butyl 3-(8-chloro-7-(2-hydroxynaphthalen-4-yl)-5- methoxy-4-oxoquinazolin-3(4H)-yl)azetidine-l-carboxylate (160 mg, 0.44 mmol) in DCM (6 mL), TF A (2 mL) was added and the resulting mixture was stirred at RT for 1 h. The mixture was concentrated in vacuo. The residue was purified by flash column chromatography on silica gel (DCM/MeOH/NH3 H20= 5: 1 :0.2) to afford the product (160 mg, 90% yield). ESI-MS m/z: 408.1 [M+H]+.
3-(Azetidin-3-yl)-8-chloro-5-hydroxy-7-(2-hydroxynaphthalen-4-yl)quinazolin- 4(3H)-one
To a mixture of 3-(azetidin-3-yl)-8-chloro-7-(2-hydroxynaphthalen-4- yl)-5-methoxyquinazolin-4(3H)-one (160 mg, 0.39 mmol) in DCM (5 mL) at -78 °C, BBr3 (490 mg, 1.96 mmol) was added slowly. The mixture was stirred at RT for 3 h and poured into ice-water. K2C03 was added. The mixture was concentrated in vacuo. The residue was purified by flash chromatography on silica gel (DCM/MeOH/NH3 H20 = 5: 1 :0.2) to afford the desired product (92 mg, 60% yield). ESI-MS m/z: 394.1 [M+H]+.
3-(l-Acryloylazetidin-3-yl)-8-chloro-5-hydroxy-7-(2-hydroxynaphthalen-4- yl)quinazolin-4(3H)-one
To a mixture of 3-(azetidin-3-yl)-8-chloro-5-hydroxy-7-(2- hydroxynaphthalen-4-yl)quinazolin-4(3H)-one (92 mg, 0.23 mmol) and DIPEA (59 mg, 0.46 mmol) in DCM (5 mL) at -78 °C, was added dropwise a solution of acryloyl chloride (21 mg, 0.23 mmol) in DCM (2 mL). The mixture was stirred at -78 °C for 10 min. The mixture was concentrated in vacuo and the residue was purified by prep-TLC plate (DCM/MeOH = 8: 1) to afford the desired product (8 mg, 8% yield). ESI-MS m/z: 448.1 [M+H]+. 1HNMR (400MHz, CDC13) 5: 11.36 (s, 1H), 8.28 (s, 1H), 7.75 (d, J = 8.0 Hz, 1H), 7.45-7.41 (m, 1H), 7.35-7.32 (m, 1H), 7.28 (d, J= 2.4 Hz, 1H), 7.24-7.21 (m, 1H), 7.06 (d, J= 2.4 Hz, 1H), 7.03 (s, 1H), 6.44 (dd, J= 1.6, 17.2 Hz, 1H), 6.24 (dd, J= 10.4, 16.8 Hz, 1H), 5.80 (dd, J = 1.6, 10.8 Hz, 1H), 5.41-5.34 (m, 1H), 4.77- 4.57 (m, 4H).
EXAMPLE 7
BIOCHEMICAL ASSAY OF THE COMPOUNDS
Test compounds were prepared as 10 mM stock solutions in DMSO
(Fisher cat# BP-231-100). KRAS G12C 1-169, his-tagged protein, GDP-loaded was diluted to 2 μιη in buffer (20mM Hepes, 150mM NaCl, ImM MgCl2). Compounds were tested for activity as follows:
Compounds were diluted to 50X final test concentration in DMSO in 96- well storage plates. Compound stock solutions were vortexed before use and observed carefully for any sign of precipitation. Dilutions were as follow:
1. For ΙΟΟμΜ final compound concentration, compounds were diluted to 5000μΜ (5μ1 lOmM compound stock + 5μ1 DMSO and mixed well by pipetting. 2. For 30μΜ final compound concentration, compounds were diluted to 1500μΜ (3μ1 lOmM compound stock + 17μ1 DMSO) and mixed well by pipetting.
3. For ΙΟμΜ final compound concentration, compounds were diluted to 500μΜ (2μ1 lOmM compound stock + 38μ1 DMSO) and mixed well by pipetting.
49μ1 of the stock protein solution was added to each well of a 96-well PCR plate (Fisher cat# 1423027). Ιμΐ of the diluted 50X compounds were added to appropriate wells in the PCR plate using 12-channel pipettor. Reactions were mixed carefully and thoroughly by pipetting up/down with a 200μ1 multi-channel pipettor. The plate was sealed well with aluminum plate seal, and stored in drawer at room temperature for 30 min, 2 hour or 24hrs. 5μ1 of 2% formic acid (Fisher cat# Al 17) in DI H20 was then added to each well followed by mixing with a pipette. The plate was then resealed with aluminum seal and stored on dry ice until analyzed as described below.
The above described assays were analyzed by mass spectrometry according to one of the following two procedures:
RapidFire/TOF Assay:
The MS instrument is set to positive polarity, 2 GHz resolution, and low mass (1700) mode and allowed to equilibrate for 30 minutes. The instrument is then calibrated, switched to acquisition mode and the appropriate method loaded.
After another 30 minute equilibration time, a blank batch (i.e., buffer) is run to ensure equipment is operating properly. The samples are thawed at 37°C for 10 minutes, briefly centrifuged, and transfer to the bench top. Wells Al and H12 are spiked with 1 uL 500 uM internal standard peptide, and the plates centrifuged at 2000 x g for 5 minutes. The method is then run and masses of each individual well recorded.
The masses (for which integration data is desired) for each well are pasted into the platemap and exported from the analysis. Masses for the internal standards are exported as well. The data at 50 ppm is extracted for the +19 charge state, and identity of well Al is assigned using the internal standard spike and integrated. Peak data is exported as a TOF list and the above steps are repeated individually, for the +20, 21, 22, 23, 24, and 25 charge states.
O-Exactive Assay:
The masses and peak intensities of KRAS G12C protein species were measured using a Dionex RSLCnano system (Thermo Scientific) connected to a Q Exactive Plus mass spectrometer (Thermo Scientific).
20 mL of sample was each loaded onto a Aeris™ 3.6 μπι WIDEPORE C4 200 A, LC Column 50 x 2.1 mm column maintained at 40°C at a flow rate of 600 μΐ min"1 with 20% Solvent A (0.1% formic acid in H20) and 80% Solvent B (0.1% formic acid in acetonitrile). The liquid chromatography conditions were 20% solvent B for 1 min, 20% to 60% solvent B for 1.5 min, 60% to 90% solvent for 0.5 min, 90% solvent B for 0.2 min, 90% to 20% solvent B for 0.2 min, and then equilibrated for 1.6 min before the following sample injection. The flow rate was maintained at 600 μΐ min"1 throughout the sampl e analy si s .
The mass spectrometer was operated in profile mode at a resolution of 17500, 5 microscans, using 50 msec max injection time and an AGC target of le6, and a full mass range from 800-1850 m/z was recorded. The HCD trapping gas was optimized for maximum sensitivity for intact proteins. The ionization method was electrospray ionization, which used a spray voltage of 4kV, sheath gas flow set to 50 au, auxiliary gas flow set to 10 au and sweep gas flow set to 1 au. The capillary ion transfer temperature was 320°C and the S-lens RF level was set to 50 voltage. Protein
Deconvolution software (Thermo Scientific) was used to deconvolute the charge envelopes of protein species in samples.
Data was analyzed using the Thermo protein deconvolution package. Briefly the charge envelope for each observed species was quantitatively deconvoluted to determine the mass and intensity of each parent species (modified or unmodified protein). % modification was calculated based on the deconvoluted peak intensities. Other in vitro analyses are as follows:
Inhibition of Cell Growth:
The ability of the subject compounds to inhibit RAS-mediated cell growth is assessed and demonstrated as follows. Cells expressing a wildtype or a mutant RAS are plated in white, clear bottom 96 well plates at a density of 5,000 cells per well. Cells are allowed to attach for about 2 hours after plating before a compound disclosed herein is added. After certain hours (e.g., 24 hours, 48 hours, or 72 hours of cell growth), cell proliferation is determined by measuring total ATP content using the Cell Titer Glo reagent (Promega) according to manufacturer's instructions.
Proliferation EC50s is determined by analyzing 8 point compound dose responses at half-log intervals decreasing from 100 μΜ.
Inhibition of RAS-mediated signaling transduction:
The ability of the compounds disclosed herein in inhibiting RAS- mediated signaling is assessed and demonstrated as follows. Cells expressing wild type or a mutant RAS (such as G12C, G12V, or G12A) are treated with or without (control cells) a subject compound. Inhibition of RAS signaling by one or more subject compounds is demonstrated by a decrease in the steady-state level of phosphorylated MEK, phosphorylated ERK, phosphorylated RSK, and/or Raf binding in cells treated with the one or more of the subject compounds as compared to the control cells.
Compound 2 was tested according to the above methods and found to covalently bind to KRAS G12C to the extent of greater than about 30% (i.e., at least about 30% of the protein present in the well was found to be covalently bound to test compound). Other compounds were tested under similar conditions. Results are presented in Table 2.
I l l Table 2
Activity of Representative Compounds*
* binding activity determined at 2 hours unless otherwise noted
+ indicates binding activity up to 5%
++ indicates binding activity from 5% up to 50%
+++ indicates binding activity 50% or greater
† indicates binding activity determined at 4 hours
1 indicates binding activity determined at 6 hours
TBD = to be determined
All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification or the attached Application Data Sheet are incorporated herein by reference, in their entirety to the extent not inconsistent with the present description.
U.S. provisional patent application Serial No. 62/195,636 filed July 22, 2015, U.S. provisional patent application Serial No. 62/342,078 filed May 26, 2016, the GCC patent application Serial No. 31750, filed July 21, 2016 and Jordanian patent application Serial No. 150/2016, filed on July 21, 2016 are incorporated herein by reference, in their entirety.
From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. Accordingly, the invention is not limited except as by the appended claims.

Claims

A compound having the following structure (I):
(I)
or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof, wherein:
A is N or C;
B is oxo, cyano, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, heteroaryl, cycloalkylalkyl, heterocycloalkyl, heteroaiylalkyl, amino, alkylamino, arylamino, -C02H, -CO H2, aminylcarbonyl, aminylcarbonylalkyl, heteroarylamino, halo, haloalkyl, alkoxy, haloalkoxy, aryl or -X-L2-Ra;
X is - Rb- or -0-;
L1 is alkylene, cycloalkylene, heterocyclylene or absent;
L2 is alkylene or absent;
R is H, cyano, amino, halo, haloalkyl, hydroxyl, cycloalkyl, heterocyclyl, heterocycloalkyl, aryl, heteroaryl, -C02H, -CO H2, aminylcarbonyl, Ci- C6 alkyl, Ci-C6 alkylaminyl or Ci-C6 alkoxy;
Ra is cycloalkyl, heterocyclyl, heteroaryl, -(C=0)OH, -(C=0) H2 or -(C=0) HOH;
Rb is, at each occurrence, independently H or Ci-C6 alkyl;
R1 is aryl or heteroaryl;
R2a, R2b and R2c are each independently H, amino, cyano, halo, hydroxyl, Ci-C6 alkyl, Ci-C6 alkylaminyl, -NRb(C=0)Rb, Ci-C6 haloalkyl, Ci-C6 alkoxy, C3-C8 cycloalkyl, heterocyclylalkyl, C2-C6 alkynyl, C2-C6 alkenyl, aminylalkyl,
alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aminylcarbonyl, heteroaryl or aryl;
~ is a single or double bond such that all valences are satisfied; and E is an electrophilic moiety capable of forming a covalent bond with the cysteine residue at position 12 of a KRAS, HRAS or NRAS G12C mutant protein.
2. The compound of claim 1, wherein B is cycloalkyl, heterocyclyl or heteroaryl.
3. The compound of any one of claims 1 or 2, wherein L1 is alkylene or absent.
4. The compound of any one of claims 1-3, wherein the compound has the following structure (IA):
(IA)
wherein:
G1 is N or CH;
G2 is NRC or CHRC;
Rc is H, alkyl, alkylcarbonyl, aminocarbonyl, alkylcarbonylaminyl, aminocarbonylaminyl or heteroaryl carbonyl;
R3a and R3b are, at each occurrence, independently
H, -OH, -NH2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C2- C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R3a and R3b join to form oxo, a carbocyclic or heterocyclic ring; or R3a is H, -OH, -NH2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R3b joins with R4b to form a carbocyclic or heterocyclic ring; R a and R are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, C C6 haloalkyl, Ci-C6 haloalkoxy, C2- C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R4a and R4b join to form oxo, a carbocyclic or heterocyclic ring; or R4a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R4b joins with R3b to form a carbocyclic or heterocyclic ring;
m1 and m2 are each independently 1, 2 or 3; and
n is an integer from 0 to 5.
5. The compound of claim 4, wherein the compound has the following structure (IAa or (IAb):
(IAa) (IAb)
wherein p is an integer from
6. The compound of any one of claims 4 or 5, wherein the compound has one of the followin structures (IAc), (IAd) or (IAe):
(IAc) (IAd) (IAe)
7. The compound of any one of claims 4-6, wherein Rc is alkylcarbonyl, aminocarbonyl, alkylcarbonylaminyl, aminocarbonylaminyl or heteroary 1 carb ony 1.
8. The compound of claim 7, wherein alkylcarbonyl is substituted with aminocarbonyl, hydroxylaminocarbonyl, hydroxyl or amino.
9. The compound of any one of claims 4-8, wherein Rc has the following structures:
wherein p2 is an integer from
10. The compound of claim 1, wherein B is alkyl, cycloalkylalkyl, heterocycloalkyl, heteroarylalkyl or -X-L2-Ra.
11. The compound of claim 10, wherein B is -X-L2-Ra.
12. The compound of claim 11, wherein Ra is heterocyclyl or heteroaryl.
13. The compound of any one of claims 10-12, wherein L1 is alkylene or absent.
14. The compound of any one of claims 10-13, wherein the compound has one of the following structures (IB) or (IC):
wherein:
H represents a 5 or 6-membered heteroaryl ring optionally substituted with one or more of R3a, R3b, R4a and R4b;
G1 is N or CH;
G2 is RC or CHRC;
Rc is H, alkyl, alkylcarbonyl, aminocarbonyl, alkylcarbonylaminyl, aminocarbonylaminyl or heteroaryl carbonyl;
R3a and R3b are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C2- C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R3a and R3b join to form oxo, a carbocyclic or heterocyclic ring; or R3a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R3b joins with R4b to form a carbocyclic or heterocyclic ring;
R4a and R4b are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, C C6 haloalkyl, Ci-C6 haloalkoxy, C2- C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R4a and R4b join to form oxo, a carbocyclic or heterocyclic ring; or R4a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R4b joins with R3b to form a carbocyclic or heterocyclic ring;
m1 and m2 are each independently 1, 2 or 3; and
n is an integer from 0 to 5.
The compound of claim 14, wherein H is pyrrolidinyl pyridinyl.
16. The compound of claim 14, wherein the compound has one of the following structures (IBa or (IBb):
(IBa) (IBb)
wherein Rd is, at each occurrence, independently H, halo or hydroxyl, and p3 is an integer from 0 to 3.
17. The compound of any one of claims 14-16, wherein the compound has one of the followin structures (IBc), (IBd), (IBe) or (IBf):
(IBc) (ICa) (IBe)
(IBf)
18. The compound of any one of claims 11-17, wherein X is NH.
The compound of any one of claims 11-18, wherein L2 is absent.
20. The compound of any one of claims 11-18, wherein L2 is alkylene.
21. The compound of any one of claims 11-20, wherein B has the following structures:
22. The compound of claim 11, wherein the compound has the following structure (ID):
(ID)
wherein p is an integer from
23. The compound of claim 22, wherein Ra is
, -(C=0)OH, -(C=0) H2 or -(C=0) HOH.
24. The compound of any one of claims 22 or 23, wherein the compound has the following structure IDa):
25. The compound of any one of claims 22-24, wherein X is NH.
26. The compound of claim 10, wherein B is alkyl.
27. The compound of claim 26, wherein alkyl is substituted with -(C=0)OH, -(C=0)NH2 or -(C=0)NHOH.
28. The compound of claim 10, wherein B is heteroarylalkyl.
29. The compound of claim 28, wherein the heteroarylalkyl is pyrrolidinylalkyl or pyridinylalkyl.
30. The compound of claim 1, wherein B is oxo.
31. The compound of claim 30, wherein the compound has the following structure (IE):
(IE)
wherein:
G1 is CH;
G2 is N or CH;
R3a and R3b are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, C C6 haloalkyl, Ci-C6 haloalkoxy, C2- C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R3a and R3b join to form oxo, a carbocyclic or heterocyclic ring; or R3a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R3b joins with R4b to form a carbocyclic or heterocyclic ring;
R4a and R4b are, at each occurrence, independently
H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C2- C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl; or R4a and R4b join to form oxo, a carbocyclic or heterocyclic ring; or R4a is H, -OH, - H2, -C02H, halo, cyano, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 haloalkoxy, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl or aminylcarbonyl, and R4b joins with R3b to form a carbocyclic or heterocyclic ring; and m1 and m2 are each independently 1, 2 or 3.
32. The compound of claim 31, wherein the compound has one of the following structures (IEa), (IEb), (IEc) or (IEd):
(IEa) (IEb) (IEc)
(IEd)
33. The compound of any one of claims 31 or 32, wherein the compound has one of the following structures (IEe), (IEf), (IEg), (IEh), (IEi), (IEj), (IEk) or (IEI):
(IEe) (IEf) (IEg)
(IEh) (IEi)
(IEj) (IEk) (IEI)
34. The compound of any one of claims 1-33, wherein R1 is aryl.
35. The compound of claim 34, wherein R1 is phenyl.
36. The compound of claim 34, wherein R1 is naphthyl.
37. The compound of any one of claims 34-36, wherein R1 is substituted with one or more substituents.
38. The compound of claim 37, wherein R1 is substituted with halo, amino, hydroxyl, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, cyano, Ci-C6 haloalkyl, Ci- C6 alkoxy, alkylaminyl, cycloalkyl, heterocyclylalkyl, heterocyclylalkoxy,
heterocyclylaminyl, cycloalkylaminyl, aryl, heteroaryl, phosphate, phosphoalkoxy, boronic acid, boronic acid ester, -OC(=0)R or Ci-C6 alkylcarbonyloxy, or combinations thereof, wherein R is Ci-C6 alkyl.
39. The compound of claim 38, wherein R1 is substituted with fluoro, chloro, cyclopropyl, cyclobutyl, hydroxyl, amino, methyl, ethyl, isopropyl,
trifluoromethyl or methoxy, or combinations thereof.
40. The compound of any one of claims 34-39, wherein R1 has one of the followin structures:
41. The compound of any one of claims 1-33, wherein R1 is heteroaryl.
42. The compound of claim 41, wherein R1 has one of the following
43. The compound of any one of claims 1-42, wherein R c is H.
44. The compound of any one of claims 1-43, wherein R a and R are each independently halo, haloalkyl, alkyl, amino, hydroxyl or alkoxy.
45. The compound of any one of claims 1-44, wherein R2a is fluoro, chloro or methoxy.
46. The compound of any one of claims 1-45, wherein R2b is chloro, fluoro, amino, hydroxyl or CF3.
47. The compound of any one of claims 1-46, wherein R is H.
48. The compound of any one of claims 1-47, wherein E has the following structure:
¾-Q ^ R10
R9
wherein:
= represents a double or triple bond;
Q is -C(=0)-, -C(= R8')-, - R8C(=0)-, -S(=0)2- or - R8S(=0)2-;
R8 is H, Ci-C6 alkyl, hydroxylalkyl, aminoalkyl, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, C3-C8 cycloalkyl or heterocycloalkyl;
R8' is H, -OH, -CN or Ci-C6 alkyl; when = is a double bond then R9 and R10 are each independently H, halo, cyano, carboxyl, Ci-C6 alkyl, alkoxycarbonyl, aminylalkyl, alkylaminylalkyl, aryl, heterocyclyl, heterocyclylalkyl, heteroaryl or hydroxylalkyl, or R9 and R10 join to form a carbocyclic, heterocyclic or heteroaryl ring; and
when = is a triple bond then R9 is absent and R10 is H, Ci-C6 alkyl, aminylalkyl, alkylaminylalkyl or hydroxylalkyl.
49. The compound of claim 48, wherein Q is -C(=0)-.
50. The compound of claim 48, wherein Q is -S(=0)2-.
51. The compound of claim 48, wherein Q is - R8C(=0)-.
52. The compound of claim 48, wherein Q is - R8S(=0)2-.
53. The compound of any one of claims 48-52, wherein each of R9 and R10 are H.
54. The compound of any one of claims 1-53, wherein E has one of the followin structures:
55. The compound of claim 1, wherein the compound is selected from a compound in Table 1.
56. A substantially purified atropisomer of the compound according to any one of claims 1-55.
57. A pharmaceutical composition comprising a compound of any one of claims 1-56 and a pharmaceutically acceptable carrier.
58. The pharmaceutical composition of claim 57, wherein the pharmaceutical composition is formulated for oral administration.
59. The pharmaceutical composition of claim 57, wherein the pharmaceutical composition is formulated for injection.
60. A method for treatment of cancer, the method comprising administering an effective amount of the pharmaceutical composition of claim 57 to a subject in need thereof.
61. The method of claim 60, wherein the cancer is mediated by a KRAS G12C, HRAS G12C or RAS G12C mutation.
62. The method of claim 60, wherein the cancer is a hematological cancer, pancreatic cancer, MYH associated polyposis, colorectal cancer or lung cancer.
63. A method for regulating activity of a KRAS, HRAS or NRAS G12C mutant protein, the method comprising reacting the KRAS G12C mutant protein with the compound of any one of claims 1-56.
64. A method for inhibiting proliferation of a cell population, the method comprising contacting the cell population with the compound of any one of claims 1-56.
65. The method of claim 64, wherein inhibition of proliferation is measured as a decrease in cell viability of the cell population.
66. A method for treating a disorder mediated by a KRAS G12C, HRAS G12C or NRAS G12C mutation in a subject in need thereof, the method comprising:
determining if the subject has a KRAS, HRAS or NRAS G12C mutation; and
if the subject is determined to have the KRAS, HRAS or NRAS G12C mutation, then administering to the subject a therapeutically effective amount of the pharmaceutical composition of claim 57.
67. The method of claim 66, wherein the disorder is a cancer.
68. The method of claim 67, wherein the cancer is a hematological cancer, pancreatic cancer, MYH associated polyposis, colorectal cancer or lung cancer.
69. A method for preparing a labeled KRAS, HRAS or NRAS G12C mutant protein, the method comprising reacting the KRAS, HRAS or NRAS G12C mutant with a compound of any one of claims 1-56, to result in the labeled KRAS, HRAS or NRAS G12C protein.
70. A method for inhibiting tumor metastasis, the method comprising administering an effective amount of the pharmaceutical composition of claim 57 to a subject in need thereof.
EP16747678.7A 2015-07-22 2016-07-22 Substituted quinazoline compounds and their use as inhibitors of g12c mutant kras, hras and/or nras proteins Withdrawn EP3325447A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201562195636P 2015-07-22 2015-07-22
US201662342078P 2016-05-26 2016-05-26
GC201631750 2016-07-21
JOP/2016/0150A JOP20160150B1 (en) 2015-07-22 2016-07-21 Substituted quinazoline compounds and methods of use thereof
PCT/US2016/043568 WO2017015562A1 (en) 2015-07-22 2016-07-22 Substituted quinazoline compounds and their use as inhibitors of g12c mutant kras, hras and/or nras proteins

Publications (1)

Publication Number Publication Date
EP3325447A1 true EP3325447A1 (en) 2018-05-30

Family

ID=57821530

Family Applications (1)

Application Number Title Priority Date Filing Date
EP16747678.7A Withdrawn EP3325447A1 (en) 2015-07-22 2016-07-22 Substituted quinazoline compounds and their use as inhibitors of g12c mutant kras, hras and/or nras proteins

Country Status (5)

Country Link
EP (1) EP3325447A1 (en)
JP (1) JP6869947B2 (en)
CA (1) CA2993013A1 (en)
MX (1) MX2018000777A (en)
WO (1) WO2017015562A1 (en)

Families Citing this family (78)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
UY35464A (en) 2013-03-15 2014-10-31 Araxes Pharma Llc KRAS G12C COVALENT INHIBITORS.
TWI659021B (en) 2013-10-10 2019-05-11 亞瑞克西斯製藥公司 Inhibitors of kras g12c
JO3556B1 (en) 2014-09-18 2020-07-05 Araxes Pharma Llc Combination therapies for treatment of cancer
WO2016049524A1 (en) 2014-09-25 2016-03-31 Araxes Pharma Llc Inhibitors of kras g12c mutant proteins
US10011600B2 (en) 2014-09-25 2018-07-03 Araxes Pharma Llc Methods and compositions for inhibition of Ras
EA201792214A1 (en) 2015-04-10 2018-01-31 Араксис Фарма Ллк COMPOUNDS OF SUBSTITUTE QUINAZOLINE
JP6789239B2 (en) 2015-04-15 2020-11-25 アラクセス ファーマ エルエルシー Condensation tricyclic inhibitor of KRAS and method of its use
US10144724B2 (en) 2015-07-22 2018-12-04 Araxes Pharma Llc Substituted quinazoline compounds and methods of use thereof
US10975071B2 (en) 2015-09-28 2021-04-13 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
WO2017058902A1 (en) 2015-09-28 2017-04-06 Araxes Pharma Llc Inhibitors of kras g12c mutant proteins
WO2017058768A1 (en) 2015-09-28 2017-04-06 Araxes Pharma Llc Inhibitors of kras g12c mutant proteins
US10858343B2 (en) 2015-09-28 2020-12-08 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
EP3356354A1 (en) 2015-09-28 2018-08-08 Araxes Pharma LLC Inhibitors of kras g12c mutant proteins
WO2017058792A1 (en) 2015-09-28 2017-04-06 Araxes Pharma Llc Inhibitors of kras g12c mutant proteins
US10875842B2 (en) 2015-09-28 2020-12-29 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
WO2017070256A2 (en) 2015-10-19 2017-04-27 Araxes Pharma Llc Method for screening inhibitors of ras
AU2016355433C1 (en) 2015-11-16 2021-12-16 Araxes Pharma Llc 2-substituted quinazoline compounds comprising a substituted heterocyclic group and methods of use thereof
US9988357B2 (en) 2015-12-09 2018-06-05 Araxes Pharma Llc Methods for preparation of quinazoline derivatives
CN109311868B (en) 2015-12-22 2022-04-01 尚医治疗有限责任公司 Compounds for the treatment of cancer and inflammatory diseases
US10822312B2 (en) 2016-03-30 2020-11-03 Araxes Pharma Llc Substituted quinazoline compounds and methods of use
US10646488B2 (en) 2016-07-13 2020-05-12 Araxes Pharma Llc Conjugates of cereblon binding compounds and G12C mutant KRAS, HRAS or NRAS protein modulating compounds and methods of use thereof
US10280172B2 (en) 2016-09-29 2019-05-07 Araxes Pharma Llc Inhibitors of KRAS G12C mutant proteins
JP2019534260A (en) 2016-10-07 2019-11-28 アラクセス ファーマ エルエルシー Heterocyclic compounds as inhibitors of RAS and methods of use thereof
BR112019012976A2 (en) 2016-12-22 2019-12-31 Amgen Inc kras g12c inhibitors and methods of using them
JOP20190272A1 (en) 2017-05-22 2019-11-21 Amgen Inc Kras g12c inhibitors and methods of using the same
JP2020521741A (en) * 2017-05-25 2020-07-27 アラクセス ファーマ エルエルシー Compounds for the treatment of cancer and methods of their use
KR20200010306A (en) 2017-05-25 2020-01-30 아락세스 파마 엘엘씨 Covalent Inhibitors of KRAS
IL271230B1 (en) 2017-06-21 2024-02-01 SHY Therapeutics LLC Compounds that interact with the ras superfamily for the treatment of cancers, inflammatory diseases, rasopathies, and fibrotic disease
SG11202001499WA (en) 2017-09-08 2020-03-30 Amgen Inc Inhibitors of kras g12c and methods of using the same
KR20200051804A (en) * 2017-09-20 2020-05-13 더 유나이티드 스테이츠 오브 어메리카, 애즈 리프리젠티드 바이 더 세크러테리, 디파트먼트 오브 헬쓰 앤드 휴먼 서비씨즈 HLA class II-restricted T cell receptor for mutated RAS
TW201938561A (en) * 2017-12-08 2019-10-01 瑞典商阿斯特捷利康公司 Chemical compounds
TW201942115A (en) * 2018-02-01 2019-11-01 美商輝瑞股份有限公司 Substituted quinazoline and pyridopyrimidine derivatives useful as anticancer agents
EP3788053A1 (en) 2018-05-04 2021-03-10 Amgen Inc. Kras g12c inhibitors and methods of using the same
AU2019262589B2 (en) 2018-05-04 2022-07-07 Amgen Inc. KRAS G12C inhibitors and methods of using the same
US11932633B2 (en) * 2018-05-07 2024-03-19 Mirati Therapeutics, Inc. KRas G12C inhibitors
TW202012415A (en) 2018-05-08 2020-04-01 瑞典商阿斯特捷利康公司 Chemical compounds
JP7361720B2 (en) 2018-05-10 2023-10-16 アムジエン・インコーポレーテツド KRAS G12C inhibitors for the treatment of cancer
US11096939B2 (en) 2018-06-01 2021-08-24 Amgen Inc. KRAS G12C inhibitors and methods of using the same
MX2020012261A (en) 2018-06-12 2021-03-31 Amgen Inc Kras g12c inhibitors encompassing a piperazine ring and use thereof in the treatment of cancer.
JOP20200338A1 (en) * 2018-06-27 2020-12-24 Reborna Biosciences Inc Prophylactic or therapeutic agent for spinal muscular atrophy
MX2021003158A (en) 2018-09-18 2021-07-16 Nikang Therapeutics Inc Fused tricyclic ring derivatives as src homology-2 phosphatase inhibitors.
JP2020090482A (en) 2018-11-16 2020-06-11 アムジエン・インコーポレーテツド Improved synthesis of key intermediate of kras g12c inhibitor compound
JP7377679B2 (en) 2018-11-19 2023-11-10 アムジエン・インコーポレーテツド Combination therapy comprising a KRASG12C inhibitor and one or more additional pharmaceutically active agents for the treatment of cancer
MA55136A (en) 2018-11-19 2022-02-23 Amgen Inc G12C KRAS INHIBITORS AND METHODS OF USE THEREOF
WO2020165732A1 (en) 2019-02-12 2020-08-20 Novartis Ag Pharmaceutical combination comprising tno155 and a krasg12c inhibitor
EP3931188B1 (en) 2019-02-26 2023-08-30 Boehringer Ingelheim International GmbH New isoindolinone substituted indoles and derivatives as ras inhibitors
EP3738593A1 (en) 2019-05-14 2020-11-18 Amgen, Inc Dosing of kras inhibitor for treatment of cancers
CR20210665A (en) 2019-05-21 2022-01-25 Amgen Inc Solid state forms
US20220227729A1 (en) 2019-05-21 2022-07-21 Bayer Aktiengesellschaft Identification and use of kras inhibitors
AU2020372881A1 (en) 2019-10-28 2022-06-09 Merck Sharp & Dohme Llc Small molecule inhibitors of KRAS G12C mutant
AU2020379731A1 (en) 2019-11-04 2022-05-05 Revolution Medicines, Inc. Ras inhibitors
WO2021091982A1 (en) 2019-11-04 2021-05-14 Revolution Medicines, Inc. Ras inhibitors
MX2022005360A (en) 2019-11-04 2022-06-02 Revolution Medicines Inc Ras inhibitors.
WO2021108683A1 (en) 2019-11-27 2021-06-03 Revolution Medicines, Inc. Covalent ras inhibitors and uses thereof
GB202001344D0 (en) 2020-01-31 2020-03-18 Redx Pharma Plc Ras Inhibitors
TW202210633A (en) 2020-06-05 2022-03-16 法商昂席歐公司 A dbait molecule in combination with kras inhibitor for the treatment of cancer
EP4168002A1 (en) 2020-06-18 2023-04-26 Revolution Medicines, Inc. Methods for delaying, preventing, and treating acquired resistance to ras inhibitors
KR20230081726A (en) 2020-09-03 2023-06-07 레볼루션 메디슨즈, 인크. Use of SOS1 inhibitors to treat malignancies with SHP2 mutations
CN117683049A (en) 2020-09-15 2024-03-12 锐新医药公司 Indole derivatives as RAS inhibitors for the treatment of cancer
US20230107642A1 (en) 2020-12-18 2023-04-06 Erasca, Inc. Tricyclic pyridones and pyrimidones
WO2022140246A1 (en) 2020-12-21 2022-06-30 Hangzhou Jijing Pharmaceutical Technology Limited Methods and compounds for targeted autophagy
EP4267575A1 (en) 2020-12-22 2023-11-01 Nikang Therapeutics, Inc. Compounds for degrading cyclin-dependent kinase 2 via ubiquitin proteosome pathway
KR20240017811A (en) 2021-05-05 2024-02-08 레볼루션 메디슨즈, 인크. RAS inhibitors for the treatment of cancer
US20230106174A1 (en) 2021-05-05 2023-04-06 Revolution Medicines, Inc. Ras inhibitors
WO2022266206A1 (en) 2021-06-16 2022-12-22 Erasca, Inc. Kras inhibitor conjugates
TW202317100A (en) 2021-06-23 2023-05-01 瑞士商諾華公司 Pharmaceutical combinations comprising a kras g12c inhibitor and uses thereof for the treatment of cancers
WO2023284730A1 (en) 2021-07-14 2023-01-19 Nikang Therapeutics, Inc. Alkylidene derivatives as kras inhibitors
TW202327569A (en) 2021-09-01 2023-07-16 瑞士商諾華公司 Pharmaceutical combinations comprising a tead inhibitor and uses thereof for the treatment of cancers
AR127308A1 (en) 2021-10-08 2024-01-10 Revolution Medicines Inc RAS INHIBITORS
WO2023114954A1 (en) 2021-12-17 2023-06-22 Genzyme Corporation Pyrazolopyrazine compounds as shp2 inhibitors
WO2023152255A1 (en) 2022-02-10 2023-08-17 Bayer Aktiengesellschaft Fused pyrimidines as kras inhibitors
EP4227307A1 (en) 2022-02-11 2023-08-16 Genzyme Corporation Pyrazolopyrazine compounds as shp2 inhibitors
WO2023172940A1 (en) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Methods for treating immune refractory lung cancer
TW202346292A (en) 2022-03-28 2023-12-01 美商尼坎醫療公司 Sulfonamido derivatives as cyclin-dependent kinase 2 inhibitors
WO2023199180A1 (en) 2022-04-11 2023-10-19 Novartis Ag Therapeutic uses of a krasg12c inhibitor
WO2023240024A1 (en) 2022-06-08 2023-12-14 Nikang Therapeutics, Inc. Sulfamide derivatives as cyclin-dependent kinase 2 inhibitors
WO2023240263A1 (en) 2022-06-10 2023-12-14 Revolution Medicines, Inc. Macrocyclic ras inhibitors
GB202212641D0 (en) 2022-08-31 2022-10-12 Jazz Pharmaceuticals Ireland Ltd Novel compounds

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI271406B (en) * 1999-12-13 2007-01-21 Eisai Co Ltd Tricyclic condensed heterocyclic compounds, preparation method of the same and pharmaceuticals comprising the same
JP2002371078A (en) * 2001-06-12 2002-12-26 Sankyo Co Ltd Quinoline derivative and quinolone derivative
WO2003053958A1 (en) * 2001-12-20 2003-07-03 Celltech R & D Limited Quinazolinedione derivatives
US7368445B2 (en) * 2004-03-01 2008-05-06 Eli Lilly And Company Fused pyrazole derivatives as TGF-β signal transduction inhibitors for the treatment of fibrosis and neoplasms
TW200643015A (en) * 2005-03-11 2006-12-16 Akzo Nobel Nv 2-(4-oxo-4H-quinazolin-3-yl)acetamide derivatives
KR101826382B1 (en) * 2010-07-30 2018-02-06 온코세라피 사이언스 가부시키가이샤 Quinoline derivatives and melk inhibitors containing the same
AU2014244426B2 (en) * 2013-03-14 2019-02-28 Convergene Llc Methods and compositions for inhibition of bromodomain-containing proteins
EP3636639A1 (en) * 2013-10-10 2020-04-15 Araxes Pharma LLC Inhibitors of kras g12c
NZ724691A (en) * 2014-03-24 2018-02-23 Guangdong Zhongsheng Pharmaceutical Co Ltd Quinoline derivatives as smo inhibitors

Also Published As

Publication number Publication date
WO2017015562A1 (en) 2017-01-26
JP6869947B2 (en) 2021-05-12
MX2018000777A (en) 2018-03-23
CA2993013A1 (en) 2017-01-26
JP2018520195A (en) 2018-07-26

Similar Documents

Publication Publication Date Title
US10351550B2 (en) Substituted quinazoline compounds and methods of use thereof
US11639346B2 (en) Quinazoline derivatives as modulators of mutant KRAS, HRAS or NRAS
AU2016355433B9 (en) 2-substituted quinazoline compounds comprising a substituted heterocyclic group and methods of use thereof
EP3283462B1 (en) Fused-tricyclic inhibitors of kras and methods of use thereof
EP3197870B1 (en) Inhibitors of kras g12c mutant proteins
EP3325447A1 (en) Substituted quinazoline compounds and their use as inhibitors of g12c mutant kras, hras and/or nras proteins
EP3630745A2 (en) Covalent inhibitors of kras
WO2020113071A1 (en) Compounds and methods of use thereof for treatment of cancer
WO2018064510A1 (en) Inhibitors of kras g12c mutant proteins
WO2018218071A1 (en) Compounds and methods of use thereof for treatment of cancer
EP3573970A1 (en) 1-(6-(3-hydroxynaphthalen-1-yl)quinazolin-2-yl)azetidin-1-yl)prop-2-en-1-one derivatives and similar compounds as kras g12c inhibitors for the treatment of cancer
EP3573966A1 (en) Fused n-heterocyclic compounds and methods of use thereof
WO2017058728A1 (en) Inhibitors of kras g12c mutant proteins
WO2017058805A1 (en) Inhibitors of kras g12c mutant proteins
EP3356351A1 (en) Inhibitors of kras g12c mutant proteins
EP3356339A1 (en) Inhibitors of kras g12c mutant proteins
WO2017058807A1 (en) Inhibitors of kras g12c mutant proteins
WO2017058792A1 (en) Inhibitors of kras g12c mutant proteins
WO2017058915A1 (en) Inhibitors of kras g12c mutant proteins

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20180214

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1252622

Country of ref document: HK

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20190930

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20220405