WO2022266425A1 - Dérivés de 3-cyano-quinoléine et leurs utilisations - Google Patents

Dérivés de 3-cyano-quinoléine et leurs utilisations Download PDF

Info

Publication number
WO2022266425A1
WO2022266425A1 PCT/US2022/033962 US2022033962W WO2022266425A1 WO 2022266425 A1 WO2022266425 A1 WO 2022266425A1 US 2022033962 W US2022033962 W US 2022033962W WO 2022266425 A1 WO2022266425 A1 WO 2022266425A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
membered heterocycloalkyl
aryl
cycloalkyl
Prior art date
Application number
PCT/US2022/033962
Other languages
English (en)
Inventor
Iwona WRONA
Matthew C. Lucas
Stephane Ciblat
Fernando Padilla
Luca Arista
Alexander Flohr
Original Assignee
Black Diamond Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Black Diamond Therapeutics, Inc. filed Critical Black Diamond Therapeutics, Inc.
Publication of WO2022266425A1 publication Critical patent/WO2022266425A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • 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
    • C07D401/12Heterocyclic 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 linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • ErbB inhibitors are a known treatment for a number of cancers. However, not every patient is responsive satisfactorily to this treatment. Thus, there is a long-felt need in the art for new therapies that are able to address the variable responsiveness of cancer patients to known therapies.
  • the present disclosure provides compositions and methods for treating cancer in patients with these oncogenic mutations without the variable reponsivenss observed when patients having these ErbB mutants are treated using the existing standard of care.
  • the present disclosure provides a compound of Formula (I): a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein:
  • R W1 is H, halogen, -O(C 1 -C 6 alkyl) or C 1 -C 6 alkyl;
  • R W2 is H, halogen, -O(C 1 -C 6 alkyl), or C 1 -C 6 alkyl;
  • X 1 is -CH 2 -, -O-, -NH-, -N(R x1a )-*, or -NH-CH(R x1a )-*, wherein * denotes attachment to
  • R x1a is C 1 -C 6 alkyl optionally substituted with one or more OH;
  • Y is C 3 -C 8 cycloalkyl, 3- to 8-membered heterocycloalkyl, C 6 -C 10 aryl, or 5- to 9- membered heteroaryl, wherein the C 3 -C 8 cycloalkyl, 3- to 8-membered heterocycloalkyl, C 6 -C 10 aryl, or 5- to 9-membered heteroaryl is optionally substituted with one or more R Y ;
  • R Y is oxo, cyano, halogen, OH, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 alkoxyl, C 3 -C 8 cycloalkyl, 3- to 9-membered heterocycloalkyl, C 6 -C 10 aryl, 5- to 9-membered heteroaryl, - O-( C 3 -C 8 cycloalkyl), -O-(C 6 -C 10 aryl), -O-(3- to 9-membered heterocycloalkyl), or -O-(5- to 9- membered heteroaryl), wherein the C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 alkoxyl, C 3 - C 8 cycloalkyl, 3- to 9-membered heterocycloal
  • R Y1 is halogen, OH, C 1 -C 6 alkyl, C 1 -C 6 alkoxyl, C 3 -C 8 cycloalkyl, 3- to 9-membered heterocycloalkyl, C 6 -C 10 aryl, or 5- to 9-membered heteroaryl, wherein the C 6 -C 10 aryl or 3- to 9- membered heterocycloalkyl is optionally substituted with one or more halogen;
  • R z is OH, oxo, cyano, halogen, C 1 -C 6 alkyl, C 1 -C 6 alkoxyl, C 3 -C 6 cycloalkyl, or 3- to 9- membered heterocycloalkyl, or two R z together with the carbon they are attached to form a C 3 -C 6 cycloalkyl; wherein the C 1 -C 6 alkyl is optionally substituted with one or more OH or halogen;
  • the present disclosure provides a compound obtainable by, or obtained by, a method for preparing a compound as described herein (e.g., a method comprising one or more steps described in Schemes 1-2).
  • the present disclosure provides an isotopic derivative of a compound described. [006] In some aspects, the present disclosure provides a method of preparing a compound described herein.
  • the present disclosure provides an intermediate as described herein, being suitable for use in a method for preparing a compound as described herein (e.g., the intermediate is selected from the intermediates described in Examples 1-10).
  • the present disclosure provides a pharmaceutical composition comprising a compound described herein and one or more pharmaceutically acceptable carriers or excipients. [009] In some aspects, the present disclosure provides a method of inhibiting an oncogenic variant of an ErbB receptor, comprising administering the subject in need thereof a therapeutically effective amount of a compound described herein.
  • the present disclosure provides a method of preventing or treating cancer, comprising administering the subject in need thereof a therapeutically effective amount of a compound described herein.
  • the present disclosure provides a compound described herein for use in the prevention or treatment of cancer.
  • the present disclosure provides a compound described herein for use in the inhibition of an oncogenic variant of an ErbB receptor.
  • the present disclosure provides a compound described herein for use in the manufacture of a medicament for the prevention or treatment of cancer.
  • the present disclosure provides a compound described herein for use in the manufacture of a medicament for the inhibition of an oncogenic variant of an ErbB receptor.
  • the present disclosure relates to compounds, and pharmaceutically acceptable salts and stereoisomers thereof, useful in the treatment of cancers associated with ErbB oncogenic activity, including methods of preparing the compounds, compositions comprising the compounds, and methods of using the compounds (e.g., in the treatment of cancer).
  • the present disclosure provides a compound of Formula (I): a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein:
  • R W1 is H, halogen, -O(C 1 -C 6 alkyl) or C 1 -C 6 alkyl;
  • R w2 is H, halogen, -O(C 1 -C 6 alkyl), or C 1 -C 6 alkyl;
  • X 1 is -CH 2 -, -O-, -NH-, -N( R x1a )-*, or -NH-CH(R x1a )-*, wherein * denotes attachment to
  • R x1a is C 1 -C 6 alkyl optionally substituted with one or more OH;
  • Y is C 3 -C 8 cycloalkyl, 3- to 8-membered heterocycloalkyl, C 6 -C 10 aryl, or 5- to 9- membered heteroaryl, wherein the C 3 -C 8 cycloalkyl, 3- to 8-membered heterocycloalkyl, C 6 -C 10 aryl, or 5- to 9-membered heteroaryl is optionally substituted with one or more R Y ;
  • R Y is oxo, cyano, halogen, OH, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 alkoxyl, C 3 -C 8 cycloalkyl, 3- to 9-membered heterocycloalkyl, C 6 -C 10 aryl, 5- to 9-membered heteroaryl, - O-(C 3 -C 8 cycloalkyl), -O-(C 6 -C 10 aryl), -O-(3- to 9-membered heterocycloalkyl), or -O-(5- to 9- membered heteroaryl), wherein the C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 alkoxyl, C 3 - C 8 cycloalkyl, 3- to 9-membered heterocycloal
  • R Y1 is halogen, OH, C 1 -C 6 alkyl, C 1 -C 6 alkoxyl, C 3 -C 8 cycloalkyl, 3- to 9-membered heterocycloalkyl, C 6 -C 10 aryl, or 5- to 9-membered heteroaryl, wherein the C 6 -C 10 aryl or 3- to 9- membered heterocycloalkyl is optionally substituted with one or more halogen;
  • R z is OH, oxo, cyano, halogen, C 1 -C 6 alkyl, C 1 -C 6 alkoxyl, C 3 -C 6 cycloalkyl, or 3- to 9- membered heterocycloalkyl, or two R z together with the carbon they are attached to form a C 3 -C 6 cycloalkyl; wherein the C 1 -C 6 alkyl is optionally substituted with one or more OH or halogen;
  • the present disclosure provides a compound of Formula (I), a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein:
  • R W1 is H, halogen, -O(C 1 -C 6 alkyl) or C 1 -C 6 alkyl;
  • R w2 is H, halogen, -O(C 1 -C 6 alkyl), or C 1 -C 6 alkyl;
  • X 1 is -CH 2 -, -O-, -NH-, -N(R x1a )-*, or -NH-CH(R x1a )-*, wherein * denotes attachment to
  • Y is C 6 -C 10 aryl or 5- to 9-membered heteroaryl, wherein the C 6 -C 10 aryl or 5- to 9- membered heteroaryl is optionally substituted with one or more R Y ;
  • R Y is halogen, C 1 -C 6 alkyl, or C 1 -C 6 alkoxyl, wherein the C 1 -C 6 alkyl or C 1 -C 6 alkoxyl is optionally substituted with one or more R Y1 ;
  • R Y1 is C 6 -C 10 aryl or 5- to 9-membered heteroaryl, wherein the C 6 -C 10 aryl or 3- to 9- membered heterocycloalkyl is optionally substituted with one or more halogen;
  • R z is C 1 -C 6 alkyl
  • W 1 , R wl , W 2 , R w2 , X 1 , R x1a , Y, R Y , R Y1 , Z, R Z , R 1 , R 1a , and n can each be, where applicable, selected from the groups described herein, and any group described herein for any of W 1 , R wl , W 2 , R w2 , X 1 , R x1a , Y, R Y , R Y1 , Z, R z , R 1 , R 1a , and n can be combined, where applicable, with any group described herein for one or more of the remainder of W 1 , R wl , W 2 , R w2 , X 1 , R x1a , Y, R Y , R Y1 , Z, R z , R 1 , R 1a , and
  • W 1 N-.
  • R W1 is H, halogen, C 1 -C 6 alkyl, or -O-(C 1 -C 6 alkyl).
  • R W1 is H.
  • R W2 is H, halogen, C 1 -C 6 alkyl, or -O-(C 1 -C 6 alkyl).
  • R W2 is H or -O-(C 1 -C 6 alkyl).
  • R W2 is H.
  • R W2 is halogen. In some embodiments, R W2 is fluorine.
  • W 1 CR W1 - and W 2 is N.
  • W 1 is CH and W 2 is CH.
  • W 1 is N and W 2 is CH.
  • W 1 is CH and W 2 is N.
  • W 1 is N and W 2 is N.
  • X 1 is -NH-, -N(CH 3 )- or -O-.
  • X 1 is -NH-. In some embodiments, X 1 is -O-.
  • Y is C 3 -C 8 cycloalkyl, 3- to 8-membered heterocycloalkyl, C 6 -C 10 aryl, or 5- to 9-membered heteroaryl, wherein the C 3 -C 8 cycloalkyl, 3- to 8-membered heterocycloalkyl, C 6 -C 10 aryl, or 5- to 9-membered heteroaryl is optionally substituted with one or more R Y .
  • Y is C 6 -C 10 aryl, or 5- to 9-membered heteroaryl, wherein the C 6 -C 10 aryl, or 5- to 9-membered heteroaryl is substituted with one or more R Y .
  • Y is C 6 -C 10 aryl, or 5- to 9-membered heteroaryl.
  • Y is C 6 -C 10 aryl. In some embodiments, Y is phenyl.
  • Y is 5- to 9-membered heteroaryl. In some embodiments, Y is indazole.
  • Y is optionally substituted with one R Y .
  • Y is optionally substituted with two R Y .
  • Y is optionally substituted with three R Y .
  • R x1a is C 1 -C 6 alkyl optionally substituted with one or more OH.
  • R x1a is C 1 -C 6 alkyl substituted with one or more OH.
  • R Y is oxo, cyano, halogen, OH, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 alkoxyl, C 3 -C 8 cycloalkyl, 3- to 9-membered heterocycloalkyl, C 6 -C 10 aryl, 5- to 9- membered heteroaryl, -O-(C 3 -C 8 cycloalkyl), -O-(C 6 -C 10 aryl), -O-(3- to 9-membered heterocycloalkyl), or -O-(5- to 9 -membered heteroaryl), wherein the C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 alkoxyl, C 3 -C 8 cycloalkyl, 3- to
  • R Y is oxo, cyano, halogen, or OH.
  • R Y is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 alkoxyl, C 3 -C 8 cycloalkyl, 3- to 9-membered heterocycloalkyl, C 6 -C 10 aryl, 5- to 9-membered heteroaryl, wherein the C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 alkoxyl, C 3 -C 8 cycloalkyl, 3- to 9- membered heterocycloalkyl, C 6 -C 10 aryl, or 5- to 9-membered heteroaryl is optionally substituted with one or more R Y1 .
  • R Y is -O-(C 3 -C 8 cycloalkyl), -O-(C 6 -C 10 aryl), -O-(3- to 9- membered heterocycloalkyl), or -O-(5- to 9-membered heteroaryl), wherein the -O-(C 3 -C 8 cycloalkyl), -O-(C 6 -C 10 aryl), -O-(3- to 9-membered heterocycloalkyl), or -O-(5- to 9-membered heteroaryl) is optionally substituted with one or more R Y1 .
  • R Y is halogen, C 1 -C 6 alkyl, or C 1 -C 6 alkoxyl.
  • R Y is halogen. In some embodiments, R Y is fluorine. In some embodiments, R Y is chlorine. In some embodiments, R Y is bromine. In some embodiments, R Y is iodine.
  • R Y is C 1 -C 6 alkyl, hi some embodiments, R Y is Ci alkyl, hi some embodiments, R Y is C2 alkyl. In some embodiments, R Y is C3 alkyl. In some embodiments, R Y is C4 alkyl. In some embodiments, R Y is C5 alkyl. In some embodiments, R Y is C 6 alkyl.
  • R Y is C 1 -C 6 alkoxyl. In some embodiments R Y is Ci alkoxyl. In some embodiments, R Y is C2 alkoxyl. In some embodiments, R Y is C3 alkoxyl. In some embodiments, R Y is C4 alkoxyl. In some embodiments, R Y is C5 alkoxyl. In some embodiments, R Y is C 6 alkoxyl.
  • R Y1 is halogen, OH, C 1 -C 6 alkyl, C 1 -C 6 alkoxyl, C 3 -C 8 cycloalkyl, 3- to 9-membered heterocycloalkyl, C 6 -C 10 aryl, or 5- to 9-membered heteroaryl, wherein the C 6 -C 10 aryl or 3- to 9-membered heterocycloalkyl is optionally substituted with one or more halogen.
  • R Y1 is halogen or OH.
  • R Y1 is C 1 -C 6 alkyl, C 1 -C 6 alkoxyl, C 3 -C 8 cycloalkyl, 3- to 9- membered heterocycloalkyl, C 6 -C 10 aryl, or 5- to 9-membered heteroaryl, wherein the C 6 -C 10 aryl or 3- to 9-membered heterocycloalkyl is optionally substituted with one or more halogen.
  • R Y1 is C 1 -C 6 alkyl, C 1 -C 6 alkoxyl, C 3 -C 8 cycloalkyl, 3- to 9- membered heterocycloalkyl, C 6 -C 10 aryl, or 5- to 9 -membered heteroaryl.
  • R Y is substituted with one R Y1 .
  • R Y1 is C 6 -C 10 aryl, or 5- to 9-membered heteroaryl.
  • R Y1 is C 6 -C 10 aryl. In some embodiments, R Y1 is phenyl.
  • R Y1 is optionally substituted with one halogen. In some embodiments, R Y 1 is optionally substituted with fluorine. [067] In some embodiments, R Y1 is 5- to 9-membered heteroaryl. In some embodiments, R Y1 is pyridyl.
  • R Y is C 6 -C 10 aryl or 5- to 9-membered heteroaryl
  • X 1 is -O- or -NH-.
  • Z is -NH-, 3- to 9-membered heterocycloalkyl, (3- to 9-membered heterocycloalkyl)-NH-, or -O-(3- to 9-membered heterocycloalkyl), wherein the 3- to 9-membered heterocycloalkyl, (3- to 9-membered heterocycloalkyl)-NH-, oorr -O-(3- to 9-membered heterocycloalkyl) is optionally substituted with one or more R z .
  • Z is -NH-.
  • Z is 3- to 9-membered heterocycloalkyl, wherein the 3- to 9- membered heterocycloalkyl is optionally substituted with one or more R z .
  • Z is (3- to 9-membered heterocycloalkyl)-NH-, wherein the (3- to 9-membered heterocycloalkyl)-NH- is optionally substituted with one or more R z .
  • Z is -O-(3- to 9-membered heterocycloalkyl), wherein the -O-(3- to 9-membered heterocycloalkyl) is optionally substituted with one or more R z .
  • ZZ i iss 33 --mmeemmbbeerreedd heterocycloalkyl, (3 -membered heterocycloalkyl)-NH-, or -O-(3-membered heterocycloalkyl).
  • Z is 4- membered heterocycloalkyl, (4-membered heterocycloalkyl)-NH-, oorr -O-(4-membered heterocycloalkyl). In some embodiments, Z is 5-membered heterocycloalkyl, (5-membered heterocycloalkyl)-NH-, or -O-(5-membered heterocycloalkyl). In some embodiments, Z is 6- membered heterocycloalkyl, (6-membered heterocycloalkyl)-NH-, oorr -O-(6-membered heterocycloalkyl).
  • Z is 7 -membered heterocycloalkyl, (7 -membered heterocycloalkyl)-NH-, or -O-(7-membered heterocycloalkyl). In some embodiments, Z is 8- membered heterocycloalkyl, (8-membered heterocycloalkyl)-NH-, oorr -O-(8-membered heterocycloalkyl). In some embodiments, Z is 9-membered heterocycloalkyl, (9-membered heterocycloalkyl)-NH-, or -O-(9-membered heterocycloalkyl).
  • the 3- to 9-membered heterocycloalkyl of Z is 2,5- diazabicyclo[2.2.1]heptyl, piperidinyl, l,6-diazaspiro[3.3]heptyl, azetidinyl, or pyrrolidinyl.
  • Z is 2,5-diazabicyclo[2.2.1]heptyl. In some embodiments, Z is piperidinyl. In some embodiments, Z is l,6-diazaspiro[3.3]heptyl. In some embodiments, Z is azetidinyl.
  • Z is (azetidinyl)-NH-. In some embodiments, Z is -O- (pyrrolidinyl). hi some embodiments, Z is -O-(azetidinyl). In some embodiments, Z is -O- (piperidinyl).In some embodiments, Z is -O-(3- to 9-membered heterocycloalkyl) optionally substituted with one or more R z .
  • R z is OH, oxo, cyano, halogen, C 1 -C 6 alkyl, C 1 -C 6 alkoxyl, C 1 -C 6 cycloalkyl, or 3- to 9-membered heterocycloalkyl, or two R z together with the carbon they are attached to form a C 3 -C 6 cycloalkyl; wherein the C 1 -C 6 alkyl is optionally substituted with one or more OH or halogen.
  • R z is OH, oxo, cyano, or halogen.
  • R z is C 1 -C 6 alkyl. In some embodiments, R z is C 1 alkyl. In some embodiments, R z is C 2 alkyl. In some embodiments, R z is C 3 alkyl. In some embodiments, R z is C4 alkyl. In some embodiments, R z is C 5 alkyl. In some embodiments, R z is C 6 alkyl.
  • Z is 3- to 9-membered heterocycloalkyl and R z is C 1 -C 6 alkyl.
  • R 1 is -(CH 2 )n-N(Me)2.
  • R 1 is -(CH 2 )2-N(Me)2.
  • R 1 is -C ⁇ C-CH 3 .
  • n 0, 1, 2, or 3.
  • n is 2.
  • the compound is of formula (I-a), (I-b), (I-c), (I-d), or (I-e): or a pharmaceutically acceptable salt or stereoisomer thereof.
  • the compound is of formula (I-f) or (I-g):
  • m is 0, 1, 2, 3, 4, or 5; or a pharmaceutically acceptable salt or stereoisomer thereof.
  • the compound is selected from a compound described in Table I, or a pharmaceutically acceptable salt or stereoisomer thereof.
  • the compound is selected from a compound described in Table I, or a pharmaceutically acceptable salt thereof.
  • the compound is selected from a compound described in Table I. d d r Co r
  • the present disclosure provides a compound being an isotopic derivative (e.g., isotopically labeled compound) of a compound disclosed herein.
  • the compound is an isotopic derivative of a compound described in Table I, or a pharmaceutically acceptable salt thereof.
  • the compound is an isotopic derivative of a compound described in Table I.
  • the isotopic derivative can be prepared using any of a variety of art- recognized techniques.
  • the isotopic derivative can generally be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples described herein, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
  • the isotopic derivative is a deuterium labeled compound.
  • the isotopic derivative is a deuterium labeled compound of a compound of the Formulae disclosed herein.
  • the compound is a deuterium labeled compound of a compound described in Table I, or a pharmaceutically acceptable salt thereof.
  • the compound is a deuterium labeled compound of a compound described in Table I.
  • the deuterium labeled compound comprises a deuterium atom having an abundance of deuterium that is substantially greater than the natural abundance of deuterium, which is 0.015%.
  • the deuterium labeled compound has a deuterium enrichment factor for each deuterium atom of at least 3500 (52.5% deuterium incorporation at each deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).
  • the term “deuterium enrichment factor” means the ratio between the deuterium abundance and the natural abundance of a deuterium.
  • the deuterium labeled compound can be prepared using any of a variety of art-recognized techniques.
  • the deuterium labeled compound can generally be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples described herein, by substituting a deuterium labeled reagent for a non-deuterium labeled reagent.
  • a compound of the invention or a pharmaceutically acceptable salt or solvate thereof that contains the aforementioned deuterium atom(s) is within the scope of the invention.
  • substitution with deuterium i.e., 2H
  • a suitable pharmaceutically acceptable salt of a compound of the disclosure is, for example, an acid-addition salt of a compound of the disclosure which is sufficiently basic, for example, an acid-addition salt with, for example, an inorganic or organic acid, for example hydrochloric, hydrobromic, sulfuric, phosphoric, trifluoroacetic, formic, citric methane sulfonate or maleic acid.
  • an inorganic or organic acid for example hydrochloric, hydrobromic, sulfuric, phosphoric, trifluoroacetic, formic, citric methane sulfonate or maleic acid.
  • a suitable pharmaceutically acceptable salt of a compound of the disclosure which is sufficiently acidic is an alkali metal salt, for example a sodium or potassium salt, an alkaline earth metal salt, for example a calcium or magnesium salt, an ammonium salt or a salt with an organic base which affords a pharmaceutically acceptable cation, for example a salt with methylamine, dimethylamine, diethylamine, trimethylamine, piperidine, morpholine or tris-(2-hydroxyethyl)amine.
  • an alkali metal salt for example a sodium or potassium salt
  • an alkaline earth metal salt for example a calcium or magnesium salt
  • an ammonium salt or a salt with an organic base which affords a pharmaceutically acceptable cation, for example a salt with methylamine, dimethylamine, diethylamine, trimethylamine, piperidine, morpholine or tris-(2-hydroxyethyl)amine.
  • the term “isomerism” means compounds that have identical molecular formulae but differ in the sequence of bonding of their atoms or in the arrangement of their atoms in space. Isomers that differ in the arrangement of their atoms in space are termed “stereoisomers.” Stereoisomers that are not mirror images of one another are termed “diastereoisomers,” and stereoisomers that are non-superimposable mirror images of each other are termed “enantiomers” or sometimes optical isomers. A mixture containing equal amounts of individual enantiomeric forms of opposite chirality is termed a “racemic mixture.”
  • chiral center refers to a carbon atom bonded to four nonidentical substituents.
  • chiral isomer means a compound with at least one chiral center.
  • Compounds with more than one chiral center may exist either as an individual diastereomer or as a mixture of diastereomers, termed “diastereomeric mixture.”
  • a stereoisomer may be characterized by the absolute configuration (R or S) of that chiral center.
  • Absolute configuration refers to the arrangement in space of the substituents attached to the chiral center.
  • the substituents attached to the chiral center under consideration are ranked in accordance with the Sequence Rule of Cahn, Ingold and Prelog. (Cahn et al., Angew. Chem. Inter. Edit.
  • the term “geometric isomer” means the diastereomers that owe their existence to hindered rotation about double bonds or a cycloalkyl linker (e.g., 1,3-cyclobutyl). These configurations are differentiated in their names by the prefixes cis and trans, or Z and E, which indicate that the groups are on the same or opposite side of the double bond in the molecule according to the Cahn-Ingold-Prelog rules. [0113] It is to be understood that the compounds of the present disclosure may be depicted as different chiral isomers or geometric isomers.
  • atropic isomers are a type of stereoisomer in which the atoms of two isomers are arranged differently in space. Atropic isomers owe their existence to a restricted rotation caused by hindrance of rotation of large groups about a central bond. Such atropic isomers typically exist as a mixture, however as a result of recent advances in chromatography techniques, it has been possible to separate mixtures of two atropic isomers in select cases.
  • tautomer is one of two or more structural isomers that exist in equilibrium and is readily converted from one isomeric form to another. This conversion results in the formal migration of a hydrogen atom accompanied by a switch of adjacent conjugated double bonds. Tautomers exist as a mixture of a tautomeric set in solution. In solutions where tautomerization is possible, a chemical equilibrium of the tautomers will be reached. The exact ratio of the tautomers depends on several factors, including temperature, solvent and pH. The concept of tautomers that are interconvertible by tautomerizations is called tautomerism. Of the various types of tautomerism that are possible, two are commonly observed.
  • keto-enol tautomerism a simultaneous shift of electrons and a hydrogen atom occurs.
  • Ring-chain tautomerism arises as a result of the aldehyde group (-CHO) in a sugar chain molecule reacting with one of the hydroxy groups (-OH) in the same molecule to give it a cyclic (ring-shaped) form as exhibited by glucose.
  • stereoisomers that are not mirror images of one another are termed “diastereomers” and those that are non-superimposable mirror images of each other are termed “enantiomers”.
  • enantiomers When a compound has an asymmetric center, for example, it is bonded to four different groups, a pair of enantiomers is possible.
  • An enantiomer can be characterized by the absolute configuration of its asymmetric center and is described by the R- and S-sequencing rules of Cahn and Prelog, or by the manner in which the molecule rotates the plane of polarized light and designated as dextrorotatory or levorotatory (i.e., as (+) or (-)-isomers respectively).
  • a chiral compound can exist as either individual enantiomer or as a mixture thereof. A mixture containing equal proportions of the enantiomers is called a “racemic mixture”.
  • the compounds of this disclosure may possess one or more asymmetric centers; such compounds can therefore be produced as individual (R)- or (S)-stereoisomers or as mixtures thereof. Unless indicated otherwise, the description or naming of a particular compound in the specification and claims is intended to include both individual enantiomers and mixtures, racemic or otherwise, thereof.
  • the methods for the determination of stereochemistry and the separation of stereoisomers are well-known in the art (see discussion in Chapter 4 of “Advanced Organic Chemistry”, 4th edition J. March, John Wiley and Sons, New York, 2001), for example by synthesis from optically active starting materials or by resolution of a racemic form.
  • Some of the compounds of the disclosure may have geometric isomeric centers (E- and Z- isomers). It is to be understood that the present disclosure encompasses all optical, diastereoisomers and geometric isomers and mixtures thereof that possess inflammasome inhibitory activity.
  • the present disclosure also encompasses compounds of the disclosure as defined herein which comprise one or more isotopic substitutions.
  • any Formula described herein include the compounds themselves, as well as their salts, and their solvates, if applicable.
  • a salt for example, can be formed between an anion and a positively charged group (e.g., amino) on a substituted compound disclosed herein.
  • Suitable anions include chloride, bromide, iodide, sulfate, bisulfate, sulfamate, nitrate, phosphate, citrate, methanesulfonate, trifluoroacetate, glutamate, glucuronate, glutarate, malate, maleate, succinate, fumarate, tartrate, tosylate, salicylate, lactate, naphthalenesulfonate, and acetate (e.g., trifluoroacetate).
  • the term “pharmaceutically acceptable anion” refers to an anion suitable for forming a pharmaceutically acceptable salt.
  • a salt can also be formed between a cation and a negatively charged group (e.g., carboxylate) on a substituted compound disclosed herein.
  • Suitable cations include sodium ion, potassium ion, magnesium ion, calcium ion, and an ammonium cation such as tetramethylammonium ion or diethylamine ion.
  • the substituted compounds disclosed herein also include those salts containing quaternary nitrogen atoms.
  • the compounds of the present disclosure can exist in either hydrated or unhydrated (the anhydrous) form or as solvates with other solvent molecules.
  • Nonlimiting examples of hydrates include monohydrates, dihydrates, etc.
  • Nonlimiting examples of solvates include ethanol solvates, acetone solvates, etc.
  • solvate means solvent addition forms that contain either stoichiometric or non-stoichiometric amounts of solvent. Some compounds have a tendency to trap a fixed molar ratio of solvent molecules in the crystalline solid state, thus forming a solvate. If the solvent is water the solvate formed is a hydrate; and if the solvent is alcohol, the solvate formed is an alcoholate. Hydrates are formed by the combination of one or more molecules of water with one molecule of the substance in which the water retains its molecular state as H2O.
  • analog refers to a chemical compound that is structurally similar to another but differs slightly in composition (as in the replacement of one atom by an atom of a different element or in the presence of a particular functional group, or the replacement of one functional group by another functional group).
  • an analog is a compound that is similar or comparable in function and appearance, but not in structure or origin to the reference compound.
  • derivative refers to compounds that have a common core structure and are substituted with various groups as described herein.
  • bioisostere refers to a compound resulting from the exchange of an atom or of a group of atoms with another, broadly similar, atom or group of atoms.
  • the objective of a bioisosteric replacement is to create a new compound with similar biological properties to the parent compound.
  • the bioisosteric replacement may be physicochemically or topologically based.
  • Examples of carboxylic acid bioisosteres include, but are not limited to, acyl sulfonamides, tetrazoles, sulfonates and phosphonates. See, e.g., Patani and LaVoie, Chem. Rev. 96, 3147-3176, 1996.
  • solvated forms such as, for example, hydrated forms.
  • a suitable pharmaceutically acceptable solvate is, for example, a hydrate such as hemi-hydrate, a monohydrate, a di-hydrate or a tri-hydrate. It is to be understood that the disclosure encompasses all such solvated forms that possess inflammasome inhibitory activity.
  • crystalline materials may be analyzed using conventional techniques such as X-Ray Powder Diffraction analysis, Differential Scanning Calorimetry, Thermal Gravimetric Analysis, Diffuse Reflectance Infrared Fourier Transform (DRIFT) spectroscopy, Near Infrared (NIR) spectroscopy, solution and/or solid state nuclear magnetic resonance spectroscopy.
  • DRIFT Diffuse Reflectance Infrared Fourier Transform
  • NIR Near Infrared
  • solution and/or solid state nuclear magnetic resonance spectroscopy The water content of such crystalline materials may be determined by Karl Fischer analysis.
  • tautomeric forms include keto-, enol-, and enolate-forms, as in, for example, the following tautomeric pairs: keto/enol (illustrated below), imine/ enamine, amide/imino alcohol, amidine/ amidine, nitroso/oxime, thioketone/enethiol, and nitro/aci-nitro.
  • N-oxides may also form N- oxides.
  • a reference herein to a compound disclosed herein that contains an amine function also includes the N-oxide.
  • one or more than one nitrogen atom may be oxidized to form an N-oxide.
  • Particular examples of N-oxides are the N- oxides of a tertiary amine or a nitrogen atom of a nitrogen-containing heterocycle.
  • N-oxides can be formed by treatment of the corresponding amine with an oxidizing agent such as hydrogen peroxide or a peracid (e.g., a peroxycarboxylic acid), see for example Advanced Organic Chemistry, by Jerry March, 4th Edition, Wiley Interscience, pages.
  • N-oxides can be made by the procedure of L. W. Deady (Syn. Comm. 1977, 7, 509-514) in which the amine compound is reacted with meta-chloroperoxybenzoic acid (mCPBA), for example, in an inert solvent such as dichloromethane.
  • mCPBA meta-chloroperoxybenzoic acid
  • the compounds of the present disclosure may be administered in the form of a prodrug which is broken down in the human or animal body to release a compound of the disclosure.
  • a prodrug may be used to alter the physical properties and/or the pharmacokinetic properties of a compound of the disclosure.
  • a prodrug can be formed when the compound of the disclosure contains a suitable group or substituent to which a property-modifying group can be attached.
  • Examples of prodrugs include derivatives containing in vivo cleavable alkyl or acyl substituents at the sulfonylurea group in a compound of the any one of the Formulae disclosed herein.
  • the present disclosure includes those compounds of the present disclosure as defined hereinbefore when made available by organic synthesis and when made available within the human or animal body by way of cleavage of a prodrug thereof. Accordingly, the present disclosure includes those compounds of the present disclosure that are produced by organic synthetic means and also such compounds that are produced in the human or animal body by way of metabolism of a precursor compound, that is a compound of the present disclosure may be a synthetically-produced compound or a metabolically-produced compound.
  • a suitable pharmaceutically acceptable prodrug of a compound of the present disclosure is one that is based on reasonable medical judgment as being suitable for administration to the human or animal body without undesirable pharmacological activities and without undue toxicity.
  • Various forms of prodrug have been described, for example in the following documents: a) Methods in Enzymology, Vol. 42, p. 309-396, edited by K. Widder, et al. (Academic Press, 1985); b) Design of Pro-drugs, edited by H. Bundgaard, (Elsevier, 1985); c) A Textbook of Drug Design and Development, edited by Krogsgaard-Larsen and H.
  • Bundgaard Chapter 5 “Design and Application of Pro-drugs”, by H. Bundgaard p. 113-191 (1991); d) H. Bundgaard, Advanced Drug Delivery Reviews, 8, 1-38 (1992); e) H. Bundgaard, et al., Journal of Pharmaceutical Sciences, 77, 285 (1988); f) N. Kakeya, et al., Chem. Pharm. Bull., 32, 692 (1984); g) T. Higuchi and V. Stella, “Pro-Drugs as Novel Delivery Systems”, A.C.S. Symposium Series, Volume 14; and h) E. Roche (editor), “Bioreversible Carriers in Drug Design”, Pergamon Press, 1987.
  • a suitable pharmaceutically acceptable prodrug of a compound of the present disclosure that possesses a hydroxy group is, for example, an in vivo cleavable ester or ether thereof.
  • An in vivo cleavable ester or ether of a compound of the present disclosure containing a hydroxy group is, for example, a pharmaceutically acceptable ester or ether which is cleaved in the human or animal body to produce the parent hydroxy compound.
  • Suitable pharmaceutically acceptable ester forming groups for a hydroxy group include inorganic esters such as phosphate esters (including phosforamidic cyclic esters).
  • ester forming groups for a hydroxy group include C1 -C10 alkanoyl groups such as acetyl, benzoyl, phenylacetyl and substituted benzoyl and phenylacetyl groups, C1 -C10 alkoxycarbonyl groups such as ethoxycarbonyl, N,N-(C1-C6 alkyl)2carbamoyl, 2 -dialkylaminoacetyl and 2-carboxyacetyl groups.
  • C1 -C10 alkanoyl groups such as acetyl, benzoyl, phenylacetyl and substituted benzoyl and phenylacetyl groups
  • C1 -C10 alkoxycarbonyl groups such as ethoxycarbonyl, N,N-(C1-C6 alkyl)2carbamoyl, 2 -dialkylaminoacetyl and 2-carboxyacetyl groups.
  • Suitable pharmaceutically acceptable ether forming groups for a hydroxy group include A-acyloxyalkyl groups such as acetoxymethyl and pivaloyloxymethyl groups.
  • a suitable pharmaceutically acceptable prodrug of a compound of the present disclosure that possesses a carboxy group is, for example, an in vivo cleavable amide thereof, for example an amide formed with an amine such as ammonia, a C 1 -4alkylamine such as methylamine, a (C1 -C4 alkyl)2amine such as dimethylamine, N-ethyl-N -methylamine or diethylamine, a C1-C4 alkoxy-C2-C4 alkylamine such as 2 -methoxyethylamine, a phenyl-C1-C4 alkylamine such as benzylamine and amino acids such as glycine or an ester thereof.
  • an amine such as ammonia
  • a C 1 -4alkylamine such as methylamine
  • a (C1 -C4 alkyl)2amine such as dimethylamine, N-ethyl-N -methylamine or diethylamine
  • a suitable pharmaceutically acceptable prodrug of a compound of the present disclosure that possesses an amino group is, for example, an in vivo cleavable amide derivative thereof.
  • Suitable pharmaceutically acceptable amides from an amino group include, for example an amide formed with C1 -C10 alkanoyl groups such as an acetyl, benzoyl, phenylacetyl and substituted benzoyl and phenylacetyl groups.
  • ring substituents on the phenylacetyl and benzoyl groups include aminomethyl, N-alkylaminomethyl, N,N-dialkylaminomethyl, morpholinomethyl, piperazin- 1 -ylmethyl, and 4-(C1-C4 alkyl)piperazin- 1 -ylmethyl.
  • the in vivo effects of a compound of the present disclosure may be exerted in part by one or more metabolites that are formed within the human or animal body after administration of a compound of the present disclosure. As stated hereinbefore, the in vivo effects of a compound of the present disclosure may also be exerted by way of metabolism of a precursor compound (a prodrug).
  • the present disclosure provides a method of preparing a compound disclosed herein.
  • the present disclosure provides a method of preparing a compound, comprising one or more steps as described herein.
  • the present disclosure provides a compound obtainable by, or obtained by, or directly obtained by a method for preparing a compound described herein.
  • the present disclosure provides an intermediate being suitable for use in a method for preparing a compound described herein.
  • the compounds of the present disclosure can be prepared by any suitable technique known in the art. Particular processes for the preparation of these compounds are described further in the accompanying examples.
  • a suitable protecting group for an amino or alkylamino group is, for example, an acyl group, for example an alkanoyl group such as acetyl, an alkoxycarbonyl group, for example aa methoxycarbonyl, ethoxycarbonyl, or t-butoxycarbonyl group, an arylmethoxycarbonyl group, for example benzyloxycarbonyl, or an aroyl group, for example benzoyl.
  • the deprotection conditions for the above protecting groups necessarily vary with the choice of protecting group.
  • an acyl group such as an alkanoyl or alkoxycarbonyl group or an aroyl group may be removed by, for example, hydrolysis with a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide.
  • a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide.
  • an acyl group such as a tert-butoxycarbonyl group may be removed, for example, by treatment with a suitable acid as hydrochloric, sulfuric or phosphoric acid or trifluoroacetic acid and an arylmethoxycarbonyl group such as a benzyloxycarbonyl group may be removed, for example, by hydrogenation over a catalyst such as palladium on carbon, or by treatment with a Lewis acid for example boron tris(trifluoroacetate).
  • a suitable alternative protecting group for a primary amino group is, for example, a phthaloyl group which may be removed by treatment with an alkylamine, for example dimethylaminopropylamine, or with hydrazine.
  • a suitable protecting group for a hydroxy group is, for example, an acyl group, for example an alkanoyl group such as acetyl, an aroyl group, for example benzoyl, or an arylmethyl group, for example benzyl.
  • the deprotection conditions for the above protecting groups will necessarily vary with the choice of protecting group.
  • an acyl group such as an alkanoyl or an aroyl group may be removed, for example, by hydrolysis with a suitable base such as an alkali metal hydroxide, for example lithium, sodium hydroxide or ammonia.
  • an arylmethyl group such as a benzyl group may be removed, for example, by hydrogenation over a catalyst such as palladium on carbon.
  • a suitable protecting group for a carboxy group is, for example, an esterifying group, for example a methyl or an ethyl group which may be removed, for example, by hydrolysis with a base such as sodium hydroxide, or for example a tert-butyl group which may be removed, for example, by treatment with an acid, for example an organic acid such as trifluoroacetic acid, or for example a benzyl group which may be removed, for example, by hydrogenation over a catalyst such as palladium on carbon.
  • the processes may then further comprise the additional steps of: (i) removing any protecting groups present; (ii) converting the compound of the present disclosure into another compound of the present disclosure; (iii) forming a pharmaceutically acceptable salt, hydrate or solvate thereof; and/or (iv) forming a prodrug thereof.
  • a suitable solvent which is preferably inert under the respective reaction conditions.
  • suitable solvents comprise but are not limited to hydrocarbons, such as hexane, petroleum ether, benzene, toluene or xylene; chlorinated hydrocarbons, such as trichlorethylene, 1,2 -dichloroethane, tetrachloromethane, chloroform or dichloromethane; alcohols, such as methanol, ethanol, isopropanol, n-propanol, n- butanol or tert-butanol; ethers, such as diethyl ether, diisopropyl ether, tetrahydrofuran (THF), 2- methyltetrahydrofuran, cyclopentylmethyl ether (CPME), methyl tert-butyl ether (MTBE) or dioxane; glycol ethers, such as
  • the reaction temperature is suitably between about -100 °C and 300 °C, depending on the reaction step and the conditions used.
  • Reaction times are generally in the range between a fraction of a minute and several days, depending on the reactivity of the respective compounds and the respective reaction conditions. Suitable reaction times are readily determinable by methods known in the art, for example reaction monitoring. Based on the reaction temperatures given above, suitable reaction times generally lie in the range between 10 minutes and 48 hours.
  • Compounds designed, selected and/or optimized by methods described above, once produced, can be characterized using a variety of assays known to those skilled in the art to determine whether the compounds have biological activity.
  • the molecules can be characterized by conventional assays, including but not limited to those assays described below, to determine whether they have a predicted activity, binding activity and/or binding specificity.
  • high-throughput screening can be used to speed up analysis using such assays. As a result, it can be possible to rapidly screen the molecules described herein for activity, using techniques known in the art. General methodologies for performing high-throughput screening are described, for example, in Devlin (1998) High Throughput Screening, Marcel Dekker; and U.S. Patent No. 5,763,263. High-throughput assays can use one or more different assay techniques including, but not limited to, those described below.
  • in vitro or in vivo biological assays are may be suitable for detecting the effect of the compounds of the present disclosure.
  • These in vitro or in vivo biological assays can include, but are not limited to, enzymatic activity assays, electrophoretic mobility shift assays, reporter gene assays, in vitro cell viability assays, and the assays described herein.
  • the biological assay is described in the Examples herein.
  • the biological assay is an assay measuring cell proliferation.
  • the assay involves retroviral production wherein EGFR mutants may be subcloned.
  • retroviral expression vector retrovirus may be produced by transient transfection of cells (e.g., HEK 293T cells) with the retroviral EGFR mutant expression vector with the appropriate co-vectors.
  • the cells may be plated, incubated, and mixed with a medium (e.g., Optimem), followed by additional incubation steps and harvesting.
  • a medium e.g., Optimem
  • the assay involves generation of EGFR mutant stable cell lines.
  • the cells may be infected with supplemented viral supernatant and incubated.
  • the cells may be pelleted and the supernatant removed and the cells re-infected with supplemented viral supernatant, followed by incubation.
  • the cells may be maintained and then selected for retroviral infection, hi some embodiments, the resistant populations may be washed and plated to select for selective growth (e.g., IL-3 independent growth).
  • the cell proliferation assay involves resuspending cell lines (e.g., BaF3 cells) into 96 well plates and determining the effect of a compound of the present disclosure after incubation in the presence of vehicle control or a compound of the present disclosure at varying concentrations.
  • inhibition of cell growth may be determined by luminescent quantification of intracellular ATP content (e.g., using CellTiterGlo (Promega), according to the protocol provided by the manufacturer).
  • the comparison of cell number (e.g., on day 0 versus 72 hours post treatment) may be used to plot dose-response curves.
  • the number of viable cells may be determined and normalized to vehicle-treated controls.
  • the assay involves cellular protein analysis wherein the cell extracts may be prepared with detergent lysis, protease inhibitor, and phosphatase inhibitors cocktails.
  • the soluble protein concentration may be determined by micro- BSA assay.
  • the protein immunodetection may be performed by electrophoretic transfer of SDS-PAGE separated proteins to nitrocellulose, followed by incubation with an antibody, and chemiluminescent second step detection.
  • nitrocellulose membranes may be blocked and incubated with antibody.
  • the antibody may be used at a dilution (e.g., 1 : 1000 dilution or 1 :5000 dilution).
  • Exemplary compounds and compositions of the disclosure are potent inhibitors of one or more oncogenic variants of an EGFR.
  • exemplary compounds and compositions of the disclosure are potent inhibitors of one or more of a wild type HER-2 receptor or an oncogenic variant of a HER-2 receptor.
  • the oncogenic variant of a HER-2 receptor is an allosteric variant of a HER-2 receptor.
  • the present disclosure provides a pharmaceutical composition comprising a compound of the present disclosure as an active ingredient.
  • the present disclosure provides a pharmaceutical composition comprising a compound described herein and one or more pharmaceutically acceptable carriers or excipients. In some embodiments, the present disclosure provides a pharmaceutical composition comprising at least one compound selected from Table I. [0170] As used herein, the term “composition” is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • the compounds of present disclosure can be formulated for oral administration in forms such as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixirs, tinctures, suspensions, syrups and emulsions.
  • the compounds of present disclosure can also be formulated for intravenous (bolus or in-fusion), intraperitoneal, topical, subcutaneous, intramuscular or transdermal (e.g., patch) administration, all using forms well known to those of ordinary skill in the pharmaceutical arts.
  • the formulation of the present disclosure may be in the form of an aqueous solution comprising an aqueous vehicle.
  • the aqueous vehicle component may comprise water and at least one pharmaceutically acceptable excipient.
  • Suitable acceptable excipients include those selected from the group consisting of a solubility enhancing agent, chelating agent, preservative, tonicity agent, viscosity/ suspending agent, buffer, and pH modifying agent, and a mixture thereof.
  • any suitable solubility enhancing agent can be used.
  • a solubility enhancing agent include cyclodextrin, such as those selected from the group consisting of hydroxypropyl- ⁇ - cyclodextrin, methyl- ⁇ -cyclodextrin, randomly methylated- ⁇ -cyclodextrin, ethylated- ⁇ - cyclodextrin, triacetyl- ⁇ -cyclodextrin, peracetylated- ⁇ -cyclodextrin, carboxymethyl- ⁇ - cyclodextrin, hydroxyethyl- ⁇ -cyclodextrin, 2-hydroxy-3-(trimethylammonio)propyl- ⁇ - cyclodextrin, glucosyl- ⁇ -cyclodextrin, sulfated ⁇ -cyclodextrin (S- ⁇ -CD), maltosyl- ⁇ -cyclodextrin, ⁇ -cyclodextrin sulfobut
  • Any suitable chelating agent can be used.
  • a suitable chelating agent include those selected from the group consisting of ethylenediaminetetraacetic acid and metal salts thereof, disodium edetate, trisodium edetate, and tetrasodium edetate, and mixtures thereof.
  • any suitable preservative can be used.
  • a preservative include those selected from the group consisting of quaternary ammonium salts such as benzalkonium halides (preferably benzalkonium chloride), chlorhexidine gluconate, benzethonium chloride, cetyl pyridinium chloride, benzyl bromide, phenylmercury nitrate, phenylmercury acetate, phenylmercury neodecanoate, merthiolate, methylparaben, propylparaben, sorbic acid, potassium sorbate, sodium benzoate, sodium propionate, ethyl p-hydroxybenzoate, propylaminopropyl biguanide, and butyl- p-hydroxybenzoate, and sorbic acid, and mixtures thereof.
  • quaternary ammonium salts such as benzalkonium halides (preferably benzalkonium chloride), chlorhexidine
  • the aqueous vehicle may also include a tonicity agent to adjust the tonicity (osmotic pressure).
  • the tonicity agent can be selected from the group consisting of a glycol (such as propylene glycol, diethylene glycol, triethylene glycol), glycerol, dextrose, glycerin, mannitol, potassium chloride, and sodium chloride, and a mixture thereof.
  • the aqueous vehicle may also contain a viscosity/ suspending agent, Suitable viscosity/suspending agents include those selected from the group consisting of cellulose derivatives, such as methyl cellulose, ethyl cellulose, hydroxyethylcellulose, polyethylene glycols (such as polyethylene glycol 300, polyethylene glycol 400), carboxymethyl cellulose, hydroxypropylmethyl cellulose, and cross-linked acrylic acid polymers (carbomers), such as polymers of acrylic acid cross-linked with polyalkenyl ethers or divinyl glycol (Carbopols - such as Carbopol 934, Carbopol 934P, Carbopol 971, Carbopol 974 and Carbopol 974P), and a mixture thereof.
  • cellulose derivatives such as methyl cellulose, ethyl cellulose, hydroxyethylcellulose
  • polyethylene glycols such as polyethylene glycol 300, polyethylene glycol 400
  • carboxymethyl cellulose such as polyethylene
  • the formulation may contain a pH modifying agent.
  • the pH modifying agent is typically a mineral acid or metal hydroxide base, selected from the group of potassium hydroxide, sodium hydroxide, and hydrochloric acid, and mixtures thereof, and preferably sodium hydroxide and/or hydrochloric acid.
  • the aqueous vehicle may also contain a buffering agent to stabilize the pH.
  • the buffer is selected from the group consisting of a phosphate buffer (such as sodium dihydrogen phosphate and disodium hydrogen phosphate), a borate buffer (such as boric acid, or salts thereof including disodium tetraborate), a citrate buffer (such as citric acid, or salts thereof including sodium citrate), and ⁇ -aminocaproic acid, and mixtures thereof.
  • the formulation may further comprise a wetting agent.
  • wetting agents include those selected from the group consisting of polyoxypropylene-polyoxyethylene block copolymers (poloxamers), polyethoxylated ethers of castor oils, polyoxyethylenated sorbitan esters (polysorbates), polymers of oxyethylated octyl phenol (Tyloxapol), polyoxyl 40 stearate, fatty acid glycol esters, fatty acid glyceryl esters, sucrose fatty esters, and polyoxyethylene fatty esters, and mixtures thereof.
  • Oral compositions generally include an inert diluent or an edible pharmaceutically acceptable carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or com starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or com starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • a pharmaceutical composition which comprises a compound of the disclosure as defined hereinbefore, or a pharmaceutically acceptable salt, hydrate or solvate thereof, in association with a pharmaceutically acceptable diluent or carrier.
  • compositions of the disclosure may be in a form suitable for oral use (for example as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups or elixirs), for topical use (for example as creams, ointments, gels, or aqueous or oily solutions or suspensions), for administration by inhalation (for example as a finely divided powder or a liquid aerosol), for administration by insufflation (for example as a finely divided powder) or for parenteral administration (for example as a sterile aqueous or oily solution for intravenous, subcutaneous, intramuscular, intraperitoneal or intramuscular dosing or as a suppository for rectal dosing).
  • oral use for example as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups or
  • compositions of the disclosure may be obtained by conventional procedures using conventional pharmaceutical excipients, well known in the art.
  • compositions intended for oral use may contain, for example, one or more coloring, sweetening, flavoring and/or preservative agents.
  • An effective amount of a compound of the present disclosure for use in therapy is an amount sufficient to treat or prevent an inflammasome related condition referred to herein, slow its progression and/or reduce the symptoms associated with the condition.
  • the size of the dose for therapeutic or prophylactic purposes of a compound of the present disclosure will naturally vary according to the nature and severity of the conditions, the age and sex of the animal or patient and the route of administration, according to well-known principles of medicine.
  • the present disclosure provides a method of inhibiting an oncogenic variant of an ErbB receptor (e.g., an oncogenic variant of an EGFR), comprising administering to the subject in need thereof a therapeutically effective amount of a compound described herein.
  • an oncogenic variant of an ErbB receptor e.g., an oncogenic variant of an EGFR
  • the present disclosure provides a method of inhibiting an oncogenic variant of an ErbB receptor (e.g., an oncogenic variant of an EGFR), comprising administering to the subject in need thereof a compound described herein.
  • the present disclosure provides a method of inhibiting an oncogenic variant of an ErbB receptor (e.g., an oncogenic variant of an EGFR), comprising administering to the subject in need thereof a composition described herein.
  • an oncogenic variant of an ErbB receptor e.g., an oncogenic variant of an EGFR
  • the present disclosure provides a method of preventing or treating cancer, comprising administering to the subject in need thereof a therapeutically effective amount of a compound described herein.
  • the present disclosure provides a method of treating cancer, comprising administering to the subject in need thereof a therapeutically effective amount of a compound described herein.
  • the present disclosure provides a method of preventing or treating cancer, comprising administering to the subject in need thereof a compound described herein.
  • the present disclosure provides a method of treating cancer, comprising administering to the subject in need thereof a compound described herein.
  • the present disclosure provides a method of preventing or treating cancer, comprising administering to the subject in need thereof a composition described herein.
  • the present disclosure provides a method of treating cancer, comprising administering to the subject in need thereof a composition described herein.
  • the present disclosure provides a method of preventing or treating cancer, comprising: i) identifying a subject candidate as the subject in need of the treatment when that at least one oncogenic variant of an ErbB receptor described herein is present in the subject; and ii) administering to the subject in need of the treatment a therapeutically effective amount of a compound described herein.
  • the present disclosure provides a method of treating cancer, comprising: i) identifying a subject candidate as the subject in need of the treatment when that at least one oncogenic variant of an ErbB receptor described herein is present in the subject; and ii) administering to the subject in need of the treatment a therapeutically effective amount of a compound described herein.
  • the present disclosure provides a method of preventing or treating cancer, comprising: i) identifying a subject candidate as the subject in need of the treatment when that at least one oncogenic variant of an ErbB receptor described herein is present in the subject; and ii) administering to the subject in need of the treatment a compound described herein.
  • the present disclosure provides a method of treating cancer, comprising: i) identifying a subject candidate as the subject in need of the treatment when that at least one oncogenic variant of an ErbB receptor described herein is present in the subject; and ii) administering to the subject in need of the treatment a compound described herein.
  • the present disclosure provides a method of preventing or treating cancer, comprising: i) identifying a subject candidate as the subject in need of the treatment when that at least one oncogenic variant of an ErbB receptor described herein is present in the subject; and ii) administering to the subject in need of the treatment a composition described herein.
  • the present disclosure provides a method of treating cancer, comprising: i) identifying a subject candidate as the subject in need of the treatment when that at least one oncogenic variant of an ErbB receptor described herein is present in the subject; and ii) administering to the subject in need of the treatment a composition described herein.
  • the present disclosure provides a method of preventing or treating cancer, comprising: i) identifying a subject candidate as the subject in need of the treatment when that at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject; and ii) administering to the subject in need of the treatment a therapeutically effective amount of a compound described herein.
  • the present disclosure provides a method of treating cancer, comprising: i) identifying a subject candidate as the subject in need of the treatment when that at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject; and ii) administering to the subject in need of the treatment a therapeutically effective amount of a compound described herein.
  • the present disclosure provides a method of preventing or treating cancer, comprising: i) identifying a subject candidate as the subject in need of the treatment when that at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject; and ii) administering to the subject in need of the treatment a compound described herein.
  • the present disclosure provides a method of treating cancer, comprising: i) identifying a subject candidate as the subject in need of the treatment when that at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject; and ii) administering to the subject in need of the treatment a compound described herein.
  • the present disclosure provides a method of preventing or treating cancer, comprising: i) identifying a subject candidate as the subject in need of the treatment when that at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample "from the subject; and ii) administering to the subject in need of the treatment a composition described herein.
  • the present disclosure provides a method of treating cancer, comprising: i) identifying a subject candidate as the subject in need of the treatment when that at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject; and ii) administering to the subject in need of the treatment a composition described herein.
  • the present disclosure provides a method of preventing or treating cancer, comprising administering to the subject in need thereof a therapeutically effective amount of a compound described herein when that at least one oncogenic variant of an ErbB receptor described herein is identified as being present in the subject.
  • the present disclosure provides a method of treating cancer, comprising administering to the subject in need thereof a therapeutically effective amount of a compound described herein when that at least one oncogenic variant of an ErbB receptor described herein is identified as being present in the subject.
  • the present disclosure provides a method of preventing or treating cancer, comprising administering to the subject in need thereof a compound described herein when that at least one oncogenic variant of an ErbB receptor described herein is identified as being present in the subject.
  • the present disclosure provides a method of treating cancer, comprising administering to the subject in need thereof a compound described herein when that at least one oncogenic variant of an ErbB receptor described herein is identified as being present in the subject.
  • the present disclosure provides a method of preventing or treating cancer, comprising administering to the subject in need thereof a therapeutically effective amount of a compound described herein when that at least one oncogenic variant of an ErbB receptor described herein is identified as being present in a biological sample from the subject.
  • the present disclosure provides a method of treating cancer, comprising administering to the subject in need thereof a therapeutically effective amount of a compound described herein when that at least one oncogenic variant of an ErbB receptor described herein is identified as being present in a biological sample from the subject.
  • the present disclosure provides a method of preventing or treating cancer, comprising administering to the subject in need thereof a compound described herein when that at least one oncogenic variant of an ErbB receptor described herein is identified as being present in a biological sample from the subject.
  • the present disclosure provides a method of treating cancer, comprising administering to the subject in need thereof a compound described herein when that at least one oncogenic variant of an ErbB receptor described herein is identified as being present in a biological sample from the subject.
  • the present disclosure provides a method of preventing or treating cancer, comprising administering to the subject in need thereof a composition described herein when that at least one oncogenic variant of an ErbB receptor described herein is identified as being present in a biological sample from the subject.
  • the present disclosure provides a method of treating cancer, comprising administering to the subject in need thereof a composition described herein when that at least one oncogenic variant of an ErbB receptor described herein is identified as being present in a biological sample from the subject.
  • the present disclosure provides a compound described herein for use in the inhibition of an oncogenic variant of an ErbB receptor (e.g., an oncogenic variant of an EGFR).
  • an oncogenic variant of an ErbB receptor e.g., an oncogenic variant of an EGFR
  • the present disclosure provides a composition described herein for use in the inhibition of an oncogenic variant of an ErbB receptor (e.g., an oncogenic variant of an EGFR).
  • an oncogenic variant of an ErbB receptor e.g., an oncogenic variant of an EGFR
  • the present disclosure provides a compound described herein for use in the prevention or treatment of cancer.
  • the present disclosure provides a compound described herein for use in the treatment of cancer.
  • the present disclosure provides a composition described herein for use in the prevention or treatment of cancer.
  • the present disclosure provides a composition described herein for use in the treatment of cancer.
  • the present disclosure provides a compound described herein for use in the prevention or treatment of cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in the subject.
  • the present disclosure provides a compound described herein for use in the treatment of cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in the subject.
  • the present disclosure provides a composition described herein for use in the prevention or treatment of cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in the subject.
  • the present disclosure provides a composition described herein for use in the treatment of cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in the subject.
  • the present disclosure provides a compound described herein for use in the prevention or treatment of cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject.
  • the present disclosure provides a compound described herein for use in the treatment of cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject.
  • the present disclosure provides a composition described herein for use in the prevention or treatment of cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject.
  • the present disclosure provides a composition described herein for use in the treatment of cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject.
  • the present disclosure provides use of a compound described herein in the manufacture of a medicament for inhibiting an oncogenic variant of an ErbB receptor (e.g., an oncogenic variant of an EGFR).
  • an oncogenic variant of an ErbB receptor e.g., an oncogenic variant of an EGFR
  • the present disclosure provides use of a compound described herein in the manufacture of a medicament for preventing or treating cancer.
  • the present disclosure provides use of a compound described herein in the manufacture of a medicament for treating cancer.
  • the present disclosure provides use of a compound described herein in the manufacture of a medicament for preventing or treating cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in the subject.
  • the present disclosure provides use of a compound described herein in the manufacture of a medicament for treating cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in the subject.
  • the present disclosure provides use of a composition described herein in the manufacture of a medicament for preventing or treating cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in the subject.
  • the present disclosure provides use of a composition described herein in the manufacture of a medicament for treating cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in the subject.
  • the present disclosure provides use of a compound described herein in the manufacture of a medicament for preventing or treating cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject.
  • the present disclosure provides use of a compound described herein in the manufacture of a medicament for treating cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject.
  • the present disclosure provides use of a composition described herein in the manufacture of a medicament for preventing or treating cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject.
  • the present disclosure provides use of a composition described herein in the manufacture of a medicament for treating cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject.
  • the present disclosure provides use of a compound described herein for inhibiting an oncogenic variant of an ErbB receptor (e.g., an oncogenic variant of an EGFR).
  • an oncogenic variant of an ErbB receptor e.g., an oncogenic variant of an EGFR
  • the present disclosure provides use of a compound described herein for preventing or treating cancer.
  • the present disclosure provides use of a compound described herein in for treating cancer.
  • the present disclosure provides use of a compound described herein for preventing or treating cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in the subject. [0246] In some aspects, the present disclosure provides use of a compound described herein for treating cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in the subject.
  • the present disclosure provides use of a composition described herein for preventing or treating cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in the subject.
  • the present disclosure provides use of a composition described herein for treating cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in the subject.
  • the present disclosure provides use of a compound described herein for preventing or treating cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject.
  • the present disclosure provides use of a compound described herein for treating cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject.
  • the present disclosure provides use of a composition described herein for preventing or treating cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject.
  • the present disclosure provides use of a composition described herein for treating cancer in a subject, wherein at least one oncogenic variant of an ErbB receptor described herein is present in a biological sample from the subject.
  • cancer is a solid tumor.
  • the cancer is a bladder cancer, a breast cancer, a cervical cancer, a colorectal cancer, an endometrial cancer, a gastric cancer, a glioblastoma (GBM), a head and neck cancer, a lung cancer, a non-small cell lung cancer (NSCLC), or any subtype thereof.
  • a bladder cancer a breast cancer, a cervical cancer, a colorectal cancer, an endometrial cancer, a gastric cancer, a glioblastoma (GBM), a head and neck cancer, a lung cancer, a non-small cell lung cancer (NSCLC), or any subtype thereof.
  • GBM glioblastoma
  • NSCLC non-small cell lung cancer
  • the cancer is glioblastoma (GBM) or any subtype thereof.
  • the cancer is glioblastoma.
  • the cancer or a tumor or a cell thereof expresses an oncogenic variant of an epidermal growth factor receptor (EGFR).
  • EGFR epidermal growth factor receptor
  • the oncogenic variant is an oncogenic variant in an ErbB receptor.
  • the oncogenic variant in the ErbB receptor is an allosteric variant.
  • the ErbB receptor is an epidermal growth factor receptor (EGFR) or a human epidermal growth factor receptor 2 (HER2) receptor.
  • the ErbB receptor is an epidermal growth factor receptor (EGFR).
  • EGFR epidermal growth factor receptor
  • HER2 receptor HER2 receptor
  • the oncogenic variant is an oncogenic variant in an epidermal growth factor receptor (EGFR).
  • EGFR epidermal growth factor receptor
  • the oncogenic variant in the EGFR is an allosteric variant.
  • the oncogenic variant is an oncogenic variant of a HER2 receptor.
  • the oncogenic variant in the HER2 receptor is an allosteric variant.
  • the oncogenic variant in the EGFR is an EGFR variant III (EGFR- Viii) variant.
  • the oncogenic variant in the EGFR is a substitution of a valine (V) for an alanine (A) at position 289 of SEQ ID NO: 1.
  • the oncogenic variant is an oncogenic variant in an EGFR and wherein the oncogenic variant in the EGFR is an allosteric variant in the EGFR, the oncogenic variant in the EGFR is a modification of a structure of the EGFR, wherein the oncogenic variant in the EGFR is capable of forming a covalently linked dimer, wherein the covalently linked dimer is constitutively active and wherein the covalently linked dimer enhances an activity of EGFR when contacted to a Type I ErbB inhibitor.
  • the modification of the structure of the EGFR comprises a modification of one or more of a nucleic acid sequence, an amino acid sequence, a secondary structure, a tertiary structure, and a quaternary structure.
  • the modification of the structure of the EGFR occurs within a first cysteine rich (CR1) and/or second cysteine rich (CR2) region of EGFR.
  • the first cysteine rich (CR1) and/or second cysteine rich (CR2) region of EGFR comprises amino acid residues T211-R334 and/or C526-S645 of SEQ ID NO: 1, respectively.
  • the oncogenic variant in the EGFR generates a physical barrier to formation of a disulfide bond within the CR1 and/or the CR2 region. In some embodiments, the oncogenic variant in the EGFR removes a physical barrier to formation of a disulfide bond within the CR1 and/or the CR2 region. In some embodiments, the oncogenic variant in the EGFR results into one or more free or unpaired Cysteine (C) residues located at a dimer interface of the EGFR.
  • C Cysteine
  • the oncogenic variant in the EGFR results into one or more free or unpaired Cysteine (C) residues at a site selected from the group consisting of C190-C199, C194C207, C215C223, C219-C231, C232C240, C236-C248, C251C260, C264C291, C295C307, C311C326, C329-C333, C506-C515, C510-C523, C526- C535, C539-C555, C558-C571, C562C579, C582C591, C595C617, C620-C628 and C624C636 according to SEQ ID NO: 1.
  • C Cysteine
  • the modification occurs within 10 angstroms or less of an intramolecular disulfide bond at a site selected from the group consisting of Cl 90- C199, C194C207, C215C223, C219-C231, C232C240, C236-C248, C251C260, C264C291, C295C307, C311C326, C329-C333, C506-C515, C510-C523, C526-C535, C539-C555, C558- C571, C562C579, C582C591, C595C617, C620-C628 and C624C636 according to SEQ ID NO: 1.
  • the oncogenic variant is an oncogenic variant in an EGFR and wherein the oncogenic variant in the EGFR is an allosteric variant in the EGFR, wherein a nucleotide sequence encoding the EGFR having the oncogenic variant comprises a deletion or the substitution comprises one or more amino acids that encode an adenosine triphosphate (ATP) binding site.
  • ATP binding site comprises amino acids E746 to A750 of SEQ ID NO: 1.
  • the ATP binding site or the deletion or substitution thereof comprises K858 of SEQ ID NO: 1.
  • the deletion comprises K858 of SEQ ID NO: 1.
  • an arginine (R) is substituted for the lysine (K) at position 858 (K858R) of SEQ ID NO: 1. In some embodiments, an arginine (R) is substituted for the leucine (L) at position 858 (L858R) of SEQ ID NO: 1.
  • the oncogenic variant is an oncogenic variant in an EGFR and wherein the oncogenic variant in the EGFR is an allosteric variant in the EGFR, wherein a nucleotide sequence encoding the EGFR having the oncogenic variant comprises an insertion within a sequence encoding exon 20 or a portion thereof.
  • the sequence encoding eexxoonn 20 oorr aa portion thereof comprises aa sequence encoding KEILDEAYVMASVDNPHVCAR (SEQ ID NO: 7).
  • the sequence encoding exon 20 or a portion thereof comprises a sequence encoding a C -helix, a terminal end of the C-helix or a loop following the C-helix.
  • the insertion comprises the amino acid sequence of AS V, SVD, NPH, or FQEA.
  • the sequence encoding exon 20 or a portion thereof comprises one or more of: (a) an insertion of the amino acid sequence ASV between positions V769 and D770 of SEQ ID NO: 1; (b) an insertion of the amino acid sequence SVD between positions D770 andN771 of SEQ ID NO: 1; (c) an insertion of the amino acid sequence NPH between positions H773 and V774 of SEQ ID NO: 1; (d) an insertion of the amino acid sequence FQEA between positions A763 and Y764 of SEQ ID NO: 1; (e) an insertion of the amino acid sequence PH between positions H773 and V774 of SEQ ID NO: 1; (f) an insertion of the amino acid G between positions D770 and N771 of SEQ ID NO: 1 ; (g) an insertion of the amino acid H between positions H773 and V774 of SEQ ID NO: 1; (h) an insertion of the amino acid sequence HV between positions V774 and C775 of
  • the oncogenic variant is an oncogenic variant in an EGFR and wherein the oncogenic variant in the EGFR is an allosteric variant in the EGFR, the EGFR having the oncogenic variant comprises EGFR-Vii, EGFR-Vvi, EGFR-R222C, EGFR-R252C, EGFR- R252P, EGFR-R256Y, EGFR-T263P, EGFR-Y270C, EGFR-A289T, EGFR-A289V, EGFR- A289D, EGFR-H304Y, EGFR-G331R, EGFR-P596S, EGFR-P596L, EGFR-P596R, EGFR- G598V, EGFR-G598A, EGFR-G614D, EGFR-C620Y, EGFR-C614W, EGFR-C628F, EGFR
  • the oncogenic variant is an oncogenic variant in a HER-2 receptor.
  • the oncogenic variant is an oncogenic variant in a HER-2 receptor, the oncogenic variant in the HER2 receptor is an allosteric variant in the HER2 receptor.
  • the oncogenic variant is an oncogenic variant in a HER-2 receptor and wherein the oncogenic variant in the HER2 receptor is an allosteric variant in the HER2 receptor, the oncogenic mutation in the HER2 receptor comprises a substitution of a phenylalanine (F) for a serine (S) at position 310 of SEQ ID NO: 2 or 5.
  • the oncogenic variant is an oncogenic variant in a HER-2 receptor and wherein the oncogenic variant in the HER2 receptor is an allosteric variant in the HER2 receptor, the oncogenic mutation in the HER2 receptor comprises a substitution of a tyrosine (Y) for a serine (S) at position 310 of SEQ ID NO: 2 or 5.
  • the oncogenic variant is an oncogenic variant in a HER-2 receptor and wherein the oncogenic variant in the HER2 receptor is an allosteric variant in the HER2 receptor, the oncogenic mutation in the HER2 receptor comprises a substitution of a glutamine (Q) for an arginine (R) at position 678 of SEQ ID NO: 2 or 5.
  • the oncogenic variant is an oncogenic variant in a HER-2 receptor and wherein the oncogenic variant in the HER2 receptor is an allosteric variant in the HER2 receptor, the oncogenic mutation in the HER2 receptor comprises a substitution of a leucine (L) for a valine (V) at position 777 of SEQ ID NO: 2 or 5.
  • the oncogenic variant is an oncogenic variant in a HER-2 receptor and wherein the oncogenic variant in the HER2 receptor is an allosteric variant in the HER2 receptor, the oncogenic mutation in the HER2 receptor comprises a substitution of a methionine (M) for a valine (V) at position 777 of SEQ ID NO: 2 or 5.
  • M methionine
  • V valine
  • the oncogenic variant is an oncogenic variant in a HER-2 receptor and wherein the oncogenic variant in the HER2 receptor is an allosteric variant in the HER2 receptor, the oncogenic mutation in the HER2 receptor comprises a substitution of an isoleucine (I) for a valine (V) at position 842 of SEQ ID NO: 2 or 5.
  • the oncogenic variant is an oncogenic variant in a HER-2 receptor and wherein the oncogenic variant in the HER2 receptor is an allosteric variant in the HER2 receptor, the oncogenic mutation in the HER2 receptor comprises a substitution of an alanine (A) for a leucine (L) at position 755 of SEQ ID NO: 2 or 5.
  • the oncogenic variant is an oncogenic variant in a HER-2 receptor and wherein the oncogenic variant in the HER2 receptor is an allosteric variant in the HER2 receptor, the oncogenic mutation in the HER2 receptor comprises a substitution of a proline (P) for a leucine (L) at position 755 of SEQ ID NO: 2 or 5.
  • the oncogenic variant is an oncogenic variant in a HER-2 receptor and wherein the oncogenic variant in the HER2 receptor is an allosteric variant in the HER2 receptor, the oncogenic mutation in the HER2 receptor comprises a substitution of a serine (S) for a leucine (L) at position 755 of SEQ ID NO: 2 or 5.
  • the oncogenic variant is an oncogenic variant in a HER-2 receptor and wherein the oncogenic variant in the HER2 receptor is an allosteric variant in the HER2 receptor, wherein a nucleotide sequence encoding the HER2 receptor having the oncogenic variant comprises an insertion within a sequence encoding exon 20 or a portion thereof.
  • the sequence encoding exon 20 or a portion thereof comprises a sequence encoding KEILDEAYVMAGVGSPYVSR(SEQ ID NO: 8).
  • the sequence encoding exon 20 or a portion thereof comprises a sequence encoding a C -helix, a terminal end of the C- helix or a loop following the C -helix.
  • the insertion comprises the amino acid sequence of GSP or YVMA.
  • the sequence encoding exon 20 or a portion thereof comprises one or more of: (a) an insertion of the amino acid sequence YVMA between positions A775 and G776 of SEQ ID NO: 2; (b) an insertion of the amino acid sequence GSP between positions P780 and Y781 of SEQ ED NO: 2; (c) an insertion of the amino acid sequence YVMA between positions A771 and Y772 of SEQ ID NO: 2; (d) an insertion of the amino acid sequence YVMA between positions A775 and G776 of SEQ ID NO: 2; (e) an insertion of the amino acid V between positions V777 and G778 of SEQ ID NO: 2; (f) an insertion of the amino acid V between positions V777 and G778 of SEQ ID NO: 2; (g) a substitution of the amino acid sequence AVGCV for the GV between positions 776 and 777 of SEQ ID NO: 2; (h) a substitution of the amino acid sequence EC for the
  • the oncogenic variant is an oncogenic variant in a HER-2 receptor and wherein the oncogenic variant in the HER2 receptor is an allosteric variant in the HER2 receptor, the HER2 receptor having the oncogenic variant comprises HER2- ⁇ 16, HER2C311R, HER2-S310F, p95-HER2-M611 or any combination thereof.
  • the oncogenic variant is an oncogenic variant in a HER-4 receptor. In some embodiments, the oncogenic variant in the HER-4 receptor is an allosteric variant in the HER4 receptor. In some embodiments, the oncogenic variant in the HER4 receptor results into the deletion of exon 16 (HER4- ⁇ 16).
  • the subject or the cancer is insensitive or resistant to treatment with one or more of gefinitinib, erlotinib, afatinib, osimertinib, and necitunumab.
  • the subject or the cancer is insensitive or resistant to treatment with one or more of crixotinib, alectinib, and ceritinib.
  • the subject or the cancer is insensitive or resistant to treatment with one or more of dabrafenib and trametinib.
  • the subject or the cancer is insensitive or resistant to treatment with crizotinib.
  • the sequence encoding the oncogenic variant of the EGFR comprises a deletion of exon 20 or a portion thereof and wherein the cancer, tumor or cell thereof does not comprise an oncogenic variation in a sequence encoding one or more of an EGFR kinase domain (KD), BRAF, NTRK, and KRAS or wherein.
  • KD EGFR kinase domain
  • BRAF BRAF
  • NTRK NTRK
  • KRAS KRAS
  • the sequence encoding the oncogenic variant of the EGFR comprises a deletion of exon 20 or a portion thereof and wherein the cancer, tumor or cell thereof does not comprise a marker indicating responsiveness to immunotherapy.
  • the oncogenic variant e.g., allosteric variant
  • the oncogenic mutation e.g., allosteric mutation
  • FDA Food and Drug Administration
  • the subject has an adverse reaction to treatment with a therapeutic agent different from the compound of the present disclosure.
  • the subject has an adverse reaction to treatment with a Type I inhibitor.
  • the subject has an adverse reaction to treatment with one or more of gefinitinib, erlotinib, afatinib, osimertinib, necitunumab, crizotinib, alectinib, ceritinib, dabrafenib, trametinib, afatinib, sapitinib, dacomitinib, canertinib, pelitinib, WZ4002, WZ8040, WZ3146, CO-1686 and AZD9291.
  • the adverse reaction is an activation of the oncogenic variant of an EGFR and wherein the oncogenic variant comprises a mutation in an extracellular domain of the receptor. In some embodiments, the adverse reaction is an activation of the oncogenic variant of a HER-2 Receptor and wherein the oncogenic variant comprises a mutation in an extracellular domain of the receptor.
  • the compound is used in combination with a second therapeutically active agent.
  • the composition comprises a second therapeutically active agent.
  • the second therapeutically active agent comprises a second compound of the disclosure.
  • the second therapeutically active agent comprises a non-Type I inhibitor.
  • the non-Type I inhibitor comprises a Type II inhibitor.
  • the Type II inhibitor comprises a small molecule inhibitor.
  • the method comprises administering to the subject in need thereof a therapeutically effective amount of a non-Type I inhibitor. In some embodiments, the method comprises administering to the subject in need thereof a non-Type I inhibitor. In some embodiments, the non-Type I inhibitor comprises a small molecule Type II inhibitor. [0294] In some embodiments, the method comprises administering to the subject in need thereof a therapeutically effective amount of a non-Type I inhibitor. In some embodiments, the method comprises administering to the subject in need thereof a non-Type I inhibitor. In some embodiments, the non-Type I inhibitor comprises a small molecule Type II inhibitor.
  • the compound is used in combination with a therapeutically effective amount of a non-Type I inhibitor.
  • the compound is used in combination with a non-Type I inhibitor.
  • the non-Type I inhibitor comprises a small molecule Type II inhibitor.
  • the composition comprises a non-Type I inhibitor.
  • the non-Type I inhibitor comprises a small molecule Type II inhibitor.
  • the cancer comprises a solid tumor.
  • the cancer comprises a bladder cancer, a breast cancer, a cervical cancer, a colorectal cancer, an endometrial cancer, a gastric cancer, a glioblastoma (GBM), a head and neck cancer, a lung cancer, a non-small cell lung cancer (NSCLC) or any subtype thereof.
  • the cancer comprises a glioblastoma (GBM).
  • the cancer comprises a breast cancer.
  • the cancer comprises a lung cancer.
  • the therapeutically effective amount reduces a severity of a sign or symptom of the cancer.
  • the compound reduces a severity of a sign or symptom of the cancer.
  • the sign of the cancer comprises a tumor grade and wherein a reduction of the severity of the sign comprises a decrease of the tumor grade.
  • the sign of the cancer comprises a tumor metastasis and wherein a reduction of the severity of the sign comprises an elimination of the metastasis or a reduction in the rate or extent the metastasis.
  • the sign of the cancer comprises a tumor volume and wherein a reduction of the severity of the sign comprises an elimination of the tumor or a reduction in the volume.
  • the symptom of the cancer comprises pain and wherein a reduction of the severity of the sign comprises an elimination or a reduction in the pain.
  • the therapeutically effective amount induces a period of remission.
  • the compound induces a period of remission.
  • the therapeutically effective amount improves a prognosis of the subject.
  • the compound improves a prognosis of the subject.
  • the subject is a participant or a candidate for participation in in a clinical trial or protocol thereof.
  • the subject is excluded from treatment with a Type I inhibitor.
  • the Type I inhibitor comprises gefinitinib, erlotinib, afatinib, osimertinib, necitunumab, crizotinib, alectinib, ceritinib, dabrafenib, trametinib, afatinib, sapitinib, dacomitinib, canertinib, pelitinib, WZ4002, WZ8040, WZ3146, CO- 1686 or AZD9291.
  • the use comprises treating the subject with a Non-Type I inhibitor.
  • the composition comprises a Non-Type I inhibitor.
  • the Non-Type I inhibitor comprises a Type II small molecule inhibitor.
  • the Type II small molecule inhibitor comprises neratinib, AST - 1306, HKI-357, or lapatinib.
  • a compound of the present disclosure may be depicted in a neutral form, a cationic form (e.g., carrying one or more positive charges), or an anionic form (e.g., carrying one or more negative charges), all of which are intended to be included in the scope of the present disclosure.
  • a compound of the present disclosure when depicted in an anionic form, it should be understood that such depiction also refers to the various neutral forms, cationic forms, and anionic forms of the compound.
  • a compound the present disclosure is depicted in an anionic form, it should be understood that such depiction also refers to various salts (e.g., sodium salt) of the anionic form of the compound.
  • alkyl As used herein, “alkyl”, “C 1 , C 2 , C 3 , C 4 , C 5 or C 6 alkyl” or “C 1 -C 6 alkyl” is intended to include C 1 , C 2 , C 3 , C 4 , C 5 or C 6 straight chain (linear) saturated aliphatic hydrocarbon groups and C 3 , C 4 , C 5 or C 6 branched saturated aliphatic hydrocarbon groups.
  • C 1 -C 6 alkyl is intends to include C 1 , C 2 , C 3 , C 4 , C 5 or C 6 alkyl groups.
  • alkyl examples include, moieties having from one to six carbon atoms, such as, but not limited to, methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, t-butyl, n-pentyl, i-pentyl or n-hexyl.
  • a straight chain or branched alkyl has six or fewer carbon atoms (e.g., C 1 -C 6 for straight chain, C 3 -C 6 for branched chain), and in another embodiment, a straight chain or branched alkyl has four or fewer carbon atoms.
  • optionally substituted alkyl refers to unsubstituted alkyl or alkyl having designated substituents replacing one or more hydrogen atoms on one or more carbons of the hydrocarbon backbone.
  • substituents can include, for example, alkyl, alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, amino (including alkylamino, dialkylamino, arylamino, diarylamino and alkylarylamino), acylamino (including alkylcarbonylamino), acylamino (including alky
  • alkenyl includes unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double bond.
  • alkenyl includes straight chain alkenyl groups (e.g., ethenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl, nonenyl, decenyl), and branched alkenyl groups.
  • a straight chain or branched alkenyl group has six or fewer carbon atoms in its backbone (e.g., C 2 -C 6 for straight chain, C 3 -C 6 for branched chain).
  • C 2 -C 6 includes alkenyl groups containing two to six carbon atoms.
  • C 3 -C 6 includes alkenyl groups containing three to six carbon atoms.
  • optionally substituted alkenyl refers to unsubstituted alkenyl or alkenyl having designated substituents replacing one or more hydrogen atoms on one or more hydrocarbon backbone carbon atoms.
  • substituents can include, for example, alkyl, alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, amino (including alkylamino, dialkylamino, arylamino, diarylamino and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sul
  • alkynyl includes unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but which contain at least one triple bond.
  • alkynyl includes straight chain alkynyl groups (e.g., ethynyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl, octynyl, nonynyl, decynyl), and branched alkynyl groups.
  • a straight chain or branched alkynyl group has six or fewer carbon atoms in its backbone (e.g., C 2 -C 6 for straight chain, C 3 -C 6 for branched chain).
  • C 2 -C 6 includes alkynyl groups containing two to six carbon atoms.
  • C 3 -C 6 includes alkynyl groups containing three to six carbon atoms.
  • C 2 -C 6 alkenylene linker or “C 2 -C 6 alkynylene linker” is intended to include C 2 , C 3 , C 4 , C 5 or C 6 chain (linear or branched) divalent unsaturated aliphatic hydrocarbon groups.
  • C 2 -C 6 alkenyl ene linker is intended to include C 2 , C 3 , C 4 , C 5 or C 6 alkenylene linker groups.
  • optionally substituted alkynyl refers to unsubstituted alkynyl or alkynyl having designated substituents replacing one or more hydrogen atoms on one or more hydrocarbon backbone carbon atoms.
  • substituents can include, for example, alkyl, alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, amino (including alkylamino, dialkylamino, arylamino, diarylamino and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, alkyl
  • optionally substituted moieties include both the unsubstituted moieties and the moieties having one or more of the designated substituents.
  • substituted heterocycloalkyl includes those substituted with one or more alkyl groups, such as 2,2,6,6-tetramethyl-piperidinyl and 2,2,6,6-tetramethyl- 1 ,2,3,6-tetrahydropyridinyl.
  • cycloalkyl refers to a saturated or partially unsaturated hydrocarbon monocyclic or polycyclic (e.g., fused, bridged, or spiro rings) system having 3 to 30 carbon atoms (e.g., C 3 -C 12 , C 3 -C 10 , or C 3 -C 8 ).
  • cycloalkyl examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, 1,2,3,4-tetrahydronaphthalenyl, and adamantyl.
  • cycloalkyl examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, 1,2,3,4-tetrahydronaphthalenyl, and adamantyl.
  • polycyclic cycloalkyl only one of the rings in the cycloalkyl needs to be non-aromatic
  • heterocycloalkyl refers to a saturated or partially unsaturated 3-8 membered monocyclic, 6-12 membered bicyclic (fused, bridged, or spiro rings), or 11-14 membered tricyclic ring system (fused, bridged, or spiro rings) having one or more heteroatoms (such as O, N, S, P, or Se), e.g., 1 or 1-2 or 1-3 or 1-4 or 1-5 or 1-6 heteroatoms, or e.g. , 1, 2, 3, 4, 5, or 6 heteroatoms, independently selected from the group consisting of nitrogen, oxygen and sulfur, unless specified otherwise.
  • heteroatoms such as O, N, S, P, or Se
  • heterocycloalkyl groups include, but are not limited to, piperidinyl, piperazinyl, pyrrolidinyl, dioxanyl, tetrahydrofuranyl, isoindolinyl, indolinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, oxiranyl, azetidinyl, oxetanyl, thietanyl, 1 ,2,3,6-tetrahydropyridinyl, tetrahydropyranyl, dihydropyranyl, pyranyl, morpholinyl, tetrahydrothiopyranyl, 1 ,4-diazepanyl, 1,4-oxazepanyl, 2-oxa-5- azabicyclo[2.2.
  • aryl includes groups with aromaticity, including “conjugated,” or multicyclic systems with one or more aromatic rings and do not contain any heteroatom in the ring structure.
  • aryl includes both monovalent species and divalent species. Examples of aryl groups include, but are not limited to, phenyl, biphenyl, naphthyl and the like.
  • heteroaryl is intended to include a stable 5-, 6-, or 7 -membered monocyclic or 7-, 8-, 9-, 10-, 11- or 12-membered bicyclic aromatic heterocyclic ring which consists of carbon atoms and one or more heteroatoms, e.g., 1 or 1-2 or 1-3 or 1-4 or 1-5 or 1-6 heteroatoms, or e.g. , 1, 2, 3, 4, 5, or 6 heteroatoms, independently selected from the group consisting of nitrogen, oxygen and sulfur.
  • the nitrogen atom may be substituted or unsubstituted (i.e., N or NR wherein R is H or other substituents, as defined).
  • heteroaryl groups include pyrrole, furan, thiophene, thiazole, isothiazole, imidazole, triazole, tetrazole, pyrazole, oxazole, isoxazole, pyridine, pyrazine, pyridazine, pyrimidine, and the like.
  • Heteroaryl groups can also be fused or bridged with alicyclic or heterocyclic rings, which are not aromatic so as to form a multicyclic system (e.g., 4,5,6,7-tetrahydrobenzo[c]isoxazolyl).
  • aryl and heteroaryl include multicyclic aryl and heteroaryl groups, e.g., tricyclic, bicyclic, e.g., naphthalene, benzoxazole, benzodioxazole, benzothiazole, benzoimidazole, benzothiophene, quinoline, isoquinoline, naphthyridine, indole, benzofuran, purine, deazapurine, indolizine.
  • the cycloalkyl, heterocycloalkyl, aryl, or heteroaryl ring can be substituted at one or more ring positions (e.g., the ring-forming carbon or heteroatom such as N) with such substituents as described above, for example, alkyl, alkenyl, alkynyl, halogen, hydroxyl, alkoxy, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkylaminocarbonyl, aralkylaminocarbonyl, alkenylaminocarbonyl, alkylcarbonyl, arylcarbonyl, aralkylcarbonyl, alkenylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarbonyl, phosphate, phosphonato, phosphinato, amino (including alkylamino, dialkylamino
  • Aryl and heteroaryl groups can also be fused or bridged with alicyclic or heterocyclic rings, which are not aromatic so as to form a multicyclic system (e.g., tetralin, methylenedioxyphenyl such as benzo[d][l,3]dioxole-5-yl).
  • alicyclic or heterocyclic rings which are not aromatic so as to form a multicyclic system (e.g., tetralin, methylenedioxyphenyl such as benzo[d][l,3]dioxole-5-yl).
  • substituted means that any one or more hydrogen atoms on the designated atom is replaced with a selection from the indicated groups, provided that the designated atom’s normal valency is not exceeded, and that the substitution results in a stable compound.
  • Keto substituents are not present on aromatic moieties.
  • “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.
  • any variable e.g., R
  • its definition at each occurrence is independent of its definition at every other occurrence.
  • R e.g., R
  • the group may optionally be substituted with up to two R moieties and R at each occurrence is selected independently from the definition of R.
  • substituents and/or variables are permissible, but only if such combinations result in stable compounds.
  • hydroxy or “hydroxyl” includes groups with an -OH or -O".
  • halo or “halogen” refers to fluoro, chloro, bromo and iodo.
  • haloalkyl or “haloalkoxyl” refers to an alkyl or alkoxyl substituted with one or more halogen atoms.
  • optionally substituted haloalkyl refers to unsubstituted haloalkyl having designated substituents replacing one or more hydrogen atoms on one or more hydrocarbon backbone carbon atoms.
  • substituents can include, for example, alkyl, alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, amino (including alkylamino, dialkylamino, arylamino, diarylamino and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sul
  • alkoxy or “alkoxyl” includes substituted and unsubstituted alkyl, alkenyl and alkynyl groups covalently linked to an oxygen atom.
  • alkoxy groups or alkoxyl radicals include, but are not limited to, methoxy, ethoxy, isopropyloxy, propoxy, butoxy and pentoxy groups.
  • substituted alkoxy groups include halogenated alkoxy groups.
  • the alkoxy groups can be substituted with groups such as alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, amino (including alkylamino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, s
  • the expressions “one or more of A, B, or C,” “one or more A, B, or C,” “one or more of A, B, and C,” “one or more A, B, and C,” “selected from the group consisting of A, B, and C”, “selected from A, B, and C”, and the like are used interchangeably and all refer to a selection from a group consisting of A, B, and/or C, i.e., one or more As, one or more Bs, one or more Cs, or any combination thereof, unless indicated otherwise.
  • the present disclosure provides methods for the synthesis of the compounds of any of the Formulae described herein.
  • the present disclosure also provides detailed methods for the synthesis of various disclosed compounds of the present disclosure according to the following schemes as well as those shown in the Examples.
  • compositions are described as having, including, or comprising specific components, it is contemplated that compositions also consist essentially of, or consist of, the recited components. Similarly, where methods or processes are described as having, including, or comprising specific process steps, the processes also consist essentially of, or consist of, the recited processing steps. Further, it should be understood that the order of steps or order for performing certain actions is immaterial so long as the invention remains operable. Moreover, two or more steps or actions can be conducted simultaneously.
  • any description of a method of treatment includes use of the compounds to provide such treatment or prophylaxis as is described herein, as well as use of the compounds to prepare a medicament to treat or prevent such condition.
  • the treatment includes treatment of human or non-human animals including rodents and other disease models.
  • the term “subject” is interchangeable with the term “subject in need thereof’, both of which refer to a subject having a disease or having an increased risk of developing the disease.
  • a “subject” includes a mammal.
  • the mammal can be e.g., a human or appropriate non-human mammal, such as primate, mouse, rat, dog, cat, cow, horse, goat, camel, sheep or a pig.
  • the subject can also be a bird or fowl.
  • the mammal is a human.
  • a subject in need thereof can be one who has been previously diagnosed or identified as having a disease or disorder disclosed herein.
  • a subject in need thereof can also be one who has (e.g., is suffering from a disease or disorder disclosed herein.
  • a subject in need thereof can be one who has an increased risk of developing such disease or disorder relative to the population at large (i.e., a subject who is predisposed to developing such disorder relative to the population at large).
  • a subject in need thereof can have a refractory or resistant a disease or disorder disclosed herein (i.e., a disease or disorder disclosed herein that doesn't respond or hasn’t yet responded to treatment). The subject may be resistant at start of treatment or may become resistant during treatment.
  • the subject in need thereof received and failed all known effective therapies for a disease or disorder disclosed herein.
  • the subject in need thereof received at least one prior therapy.
  • the term “treating” or “treat” describes the management and care of a patient for the purpose of combating a disease, condition, or disorder and includes the administration of a compound of the present disclosure, or a pharmaceutically acceptable salt, polymorph or solvate thereof, to alleviate the symptoms or complications of a disease, condition or disorder, or to eliminate the disease, condition or disorder.
  • the term “treat” can also include treatment of a cell in vitro or an animal model. It is to be appreciated that references to “treating” or “treatment” include the alleviation of established symptoms of a condition.
  • Treating” or “treatment” of a state, disorder or condition therefore includes: (1) preventing or delaying the appearance of clinical symptoms of the state, disorder or condition developing in a human that may be afflicted with or predisposed to the state, disorder or condition but does not yet experience or display clinical or subclinical symptoms of the state, disorder or condition, (2) inhibiting the state, disorder or condition, i.e., arresting, reducing or delaying the development of the disease or a relapse thereof (in case of maintenance treatment) or at least one clinical or subclinical symptom thereof, or (3) relieving or attenuating the disease, i.e., causing regression of the state, disorder or condition or at least one of its clinical or subclinical symptoms.
  • a compound of the present disclosure can or may also be used to prevent a relevant disease, condition or disorder, or used to identify suitable candidates for such purposes.
  • preventing describes reducing or eliminating the onset of the symptoms or complications of such disease, condition or disorder.
  • compositions comprising any compound described herein in combination with at least one pharmaceutically acceptable excipient or carrier.
  • the term “pharmaceutical composition” is a formulation containing the compounds of the present disclosure in a form suitable for administration to a subject.
  • the pharmaceutical composition is in bulk or in unit dosage form.
  • the unit dosage form is any of a variety of forms, including, for example, a capsule, an IV bag, a tablet, a single pump on an aerosol inhaler or a vial.
  • the quantity of active ingredient (e.g., a formulation of the disclosed compound or salt, hydrate, solvate or isomer thereof) in a unit dose of composition is an effective amount and is varied according to the particular treatment involved.
  • routes including oral, pulmonary, rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular, intraperitoneal, inhalational, buccal, sublingual, intrapleural, intrathecal, intranasal, and the like.
  • Dosage forms for the topical or transdermal administration of a compound of this disclosure include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active compound is mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that are required.
  • the term “pharmaceutically acceptable” refers to those compounds, anions, cations, materials, compositions, carriers, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • the term “pharmaceutically acceptable excipient” means an excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes excipient that is acceptable for veterinary use as well as human pharmaceutical use.
  • a “pharmaceutically acceptable excipient” as used in the specification and claims includes both one and more than one such excipient.
  • a pharmaceutical composition of the disclosure is formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., ingestion), inhalation, transdermal (topical), and transmucosal administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • a compound or pharmaceutical composition of the disclosure can be administered to a subject in many of the well-known methods currently used for chemotherapeutic treatment.
  • a compound of the disclosure may be injected into the blood stream or body cavities or taken orally or applied through the skin with patches.
  • the dose chosen should be sufficient to constitute effective treatment but not so high as to cause unacceptable side effects.
  • the state of the disease condition (e.g., a disease or disorder disclosed herein) and the health of the patient should preferably be closely monitored during and for a reasonable period after treatment.
  • the term “therapeutically effective amount”, refers to an amount of a pharmaceutical agent to treat, ameliorate, or prevent an identified disease or condition, or to exhibit a detectable therapeutic or inhibitory effect.
  • the effect can be detected by any assay method known in the art.
  • the precise effective amount for a subject will depend upon the subject’s body weight, size, and health; the nature and extent of the condition; and the therapeutic or combination of therapeutics selected for administration.
  • the therapeutically effective amount can be estimated initially either in cell culture assays, e.g., of neoplastic cells, or in animal models, usually rats, mice, rabbits, dogs, or pigs.
  • the animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.
  • Therapeutic/prophylactic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., ED 50 (the dose therapeutically effective in 50% of the population) and LD 50 (the dose lethal to 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index, and it can be expressed as the ratio, LD 50 /ED 50 .
  • Pharmaceutical compositions that exhibit large therapeutic indices are preferred.
  • the dosage may vary within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration.
  • Dosage and administration are adjusted to provide sufficient levels of the active agent(s) or to maintain the desired effect.
  • Factors which may be taken into account include the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy.
  • compositions containing active compounds of the present disclosure may be manufactured in a manner that is generally known, e.g., by means of conventional mixing, dissolving, granulating, dragee -making, levigating, emulsifying, encapsulating, entrapping, or lyophilizing processes.
  • Pharmaceutical compositions maybe formulated in a conventional manner using one or more pharmaceutically acceptable carriers comprising excipients and/or auxiliaries that facilitate processing of the active compounds into preparations that can be used pharmaceutically.
  • the appropriate formulation is dependent upon the route of administration chosen.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor ELD (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS).
  • the composition must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, polyalcohols such as mannitol and sorbitol, and sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Oral compositions generally include an inert diluent or an edible pharmaceutically acceptable carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or com starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or com starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • the compounds are delivered in the form of an aerosol spray from pressured container or dispenser, which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
  • a suitable propellant e.g., a gas such as carbon dioxide, or a nebulizer.
  • Systemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
  • the active compounds can be prepared with pharmaceutically acceptable carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • a controlled release formulation including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art.
  • the materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc.
  • Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the disclosure are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved.
  • the dosages of the pharmaceutical compositions used in accordance with the disclosure vary depending on the agent, the age, weight, and clinical condition of the recipient patient, and the experience and judgment of the clinician or practitioner administering the therapy, among other factors affecting the selected dosage.
  • the dose should be sufficient to result in slowing, and preferably regressing, the symptoms of the disease or disorder disclosed herein and also preferably causing complete regression of the disease or disorder.
  • An effective amount of a pharmaceutical agent is that which provides an objectively identifiable improvement as noted by the clinician or other qualified observer. Improvement in survival and growth indicates regression.
  • the term “dosage effective manner” refers to amount of an active compound to produce the desired biological effect in a subject or cell.
  • the term “pharmaceutically acceptable salts” refer to derivatives of the compounds of the present disclosure wherein the parent compound is modified by making acid or base salts thereof.
  • pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines, alkali or organic salts of acidic residues such as carboxylic acids, and the like.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include, but are not limited to, those derived from inorganic and organic acids selected from 2 -acetoxybenzoic, 2-hydroxyethane sulfonic, acetic, ascorbic, benzene sulfonic, benzoic, bicarbonic, carbonic, citric, edetic, ethane disulfonic, 1,2-ethane sulfonic, fumaric, glucoheptonic, gluconic, glutamic, glycolic, glycollyarsanilic, hexylresorcinic, hydrabamic, hydrobromic, hydrochloric, hydroiodic, hydroxymaleic, hydroxynaphthoic, isethionic, lactic, lactobionic, lauryl sulfonic, maleic, malic, mandelic, methane sulfonic, napsylic, nitric, oxalic, pamoic, pantothenic, phenylacetic,
  • the pharmaceutically acceptable salt is a sodium salt, a potassium salt, a calcium salt, a magnesium salt, a diethylamine salt, a choline salt, a meglumine salt, a benzathine salt, a tromethamine salt, an ammonia salt, an arginine salt, or a lysine salt.
  • compositions include hexanoic acid, cyclopentane propionic acid, pyruvic acid, malonic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, 4- chlorobenzenesulfonic acid, 2 -naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo-[2.2.2]-oct-2-ene-l -carboxylic acid, 3 -phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, muconic acid, and the like.
  • the present disclosure also encompasses salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N -methylglucamine, and the like.
  • a metal ion e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion
  • organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N -methylglucamine, and the like.
  • the ratio of the compound to the cation or anion of the salt can be 1 :1, or any ratio other than 1:1, e.g., 3:1, 2:1, 1 :2, or 1:3.
  • references to pharmaceutically acceptable salts include solvent addition forms (solvates) or crystal forms (polymorphs) as defined herein, of the same salt.
  • the compounds, or pharmaceutically acceptable salts thereof are administered orally, nasally, transdermally, pulmonary, inhalationally, buccally, sublingually, intraperitoneally, subcutaneously, intramuscularly, intravenously, rectally, intrapleurally, intrathecally and parenterally. In some embodiments, the compound is administered orally.
  • One skilled in the art will recognize the advantages of certain routes of administration.
  • the dosage regimen utilizing the compounds is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound or salt thereof employed.
  • the compounds described herein, and the pharmaceutically acceptable salts thereof are used in pharmaceutical preparations in combination with a pharmaceutically acceptable carrier or diluent.
  • suitable pharmaceutically acceptable carriers include inert solid fillers or diluents and sterile aqueous or organic solutions.
  • the compounds will be present in such pharmaceutical compositions in amounts sufficient to provide the desired dosage amount in the range described herein.
  • compounds may be drawn with one particular configuration for simplicity. Such particular configurations are not to be construed as limiting the disclosure to one or another isomer, tautomer, regioisomer or stereoisomer, nor does it exclude mixtures of isomers, tautomers, regioisomers or stereoisomers; however, it will be understood that a given isomer, tautomer, regioisomer or stereoisomer may have a higher level of activity than another isomer, tautomer, regioisomer or stereoisomer.
  • a wild type EGFR sequence of the disclosure may comprise or consist of the amino acid sequence of:
  • a wild type HER2 Receptor sequence of the disclosure may comprise or consist of the amino acid sequence of:
  • a wild type HER2 Receptor sequence of the disclosure may comprise or consist of the amino acid sequence of:
  • a wild type HER2 Receptor sequence of the disclosure may comprise or consist of the amino acid sequence of:
  • a wild type HER2 Receptor sequence of the disclosure may comprise or consist of the amino acid sequence of:
  • a wild type HER2 Receptor sequence of the disclosure may comprise or consist of the amino acid sequence of:
  • neutral compounds of Formula (I) are synthesized and tested in the examples. It is understood that the neutral compounds of Formula (I) may be converted to the corresponding pharmaceutically acceptable salts of the compounds using routine techniques in the art (e.g., by saponification of an ester to the carboxylic acid salt, or by hydrolyzing an amide to form a corresponding carboxylic acid and then converting the carboxylic acid to a carboxylic acid salt).
  • Step 2 Synthesis of Methyl 6-chloro-3-(((dimethylamino)methylene)amino)picolinate
  • Step 4 Synthesis of 4, 6-Dichloro-1,5-naphthyridine-3 -carbonitrile
  • Step 1 Methyl 3-amino-6-chloro-pyridine-2-carboxylate [0390] To a solution of 3 -amino-6-chloro-pyridine-2 -carboxylic acid (15.0 g, 86.9 mmol) in methanol (225 mL) was added thionyl chloride (13.4 g, 113 mmol) dropwise. The mixture was stirred at 70 °C for 16 hr.
  • Step 2 Methyl 6-chloro-3-[(E)-dimethylaminomethyleneamino]pyridine-2 -carboxylate [0392] To a solution of methyl 3-amino-6-chloro-pyridine-2-carboxylate (500 mg, 2.68 mmo) in toluene (7 mL) was added N, N -dimethylformamide dimethyl acetal (958 mg, 8.04 mmol). The mixture was stirred at 100 °C for 3 hr.
  • Step 1 6-Chloro-4-[3-chloro-4-(difluoromethoxy)-2-fluoro-anilino]-l,5-naphthyridine- 3 -carbonitrile
  • Step 1 6-Chloro-4-[3-chloro-4-(cyclopropylmethoxy)-2-fluoro-anilino]-l,5- naphthyridine-3 -carbonitrile
  • Step 1 terZ-Butyl (S)-3-((3-fluoro-5-nitropyridin-2-yl)oxy)pyrrolidine-l-carboxylate [0402] To a mixture of tert-butyl (3 S)-3 -hydroxypyrrolidine- 1 -carboxylate (5 g, 26.7 mmol) in anhydrous tetrahydrofuran (100 mL) was added sodium hydride (2.14 g, 53.4 mmol, 60 wZ%). The mixture was stirred at 25 °C for 1 hour.
  • tert-Butyl (S)-3-((5-amino-3-fluoropyridin-2-yl)oxy)pyrrolidine-l -carboxylate [0404] To a mixture of tert-butyl (3S)-3-[(3-fluoro-5-nitro-2-pyridyl)oxy]pyrrolidine-l- carboxylate (4 g, 12.2 mmol) in methanol (40 mL) and water (7 mL) was added ammonium chloride (3.27 g, 61.1 mmol) and iron powder (3.41 g, 61.1 mmol). The mixture was stirred at 80 °C for 2 hours.
  • Step 3 tert-Butyl (S)-3-((5-amino-6-bromo-3-fluoropyridin-2-yl)oxy)pyrrolidine-l- carboxylate
  • Step 2. 1 -(3-Chloro-4-(difluoromethoxy)-2-fluorophenyl)-2,5 -dimethyl- 1 H -pyrrole
  • 2-chloro-4-(2,5-dimethylpyrrol-l-yl)-3-fluoro-phenol (3.00 g, 12.5 mmol)
  • (2-chloro-2,2-difluoro-acetyl)oxysodium (6.70 g, 43.9 mmol)
  • cesium carbonate (8.00 g, 24.5 mmol) in N,N- dimethylformarnide (24 mL) and water (4 mL) was stirred at 100 °C for 4 hr.
  • reaction mixture was diluted with water (100 mL) and extracted with ethyl acetate (200 mL x 3). The combined organic layers were washed with brine (200 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give l-[3-chloro- 4-(difluoromethoxy)-2-fluoro-phenyl]-2,5-dimethyl-pyrrole (3.60 g, crude) as a black oil.
  • Step 1 Synthesis of tert- Butyl (S)-3-((8-((3-chloro-2-fhiorophenyl)amino)-7-cyano-l,5- naphthyridin-2-yl)oxy)pyrrolidine- 1 -carboxylate
  • 6-chloro-4-(3-chloro-2-fluoro-anilino)-l,5-naphthyridine-3-carbonitrile 90 mg, 270 ⁇ mol
  • potassium tert-butoxide 121 mg, 1.08 mmol
  • tert-butyl (3S) -3 -hydroxypyrrolidine- 1 -carboxylate 152 mg, 810 ⁇ mol.
  • Step 3 Synthesis of (S)-6-((l-Acryloylpyrrolidin-3-yl)oxy)-4-((3-chloro-2- fhiorophenyl)amino)-l,5-naphthyridine-3-carbonitrile
  • Step 1 Synthesis of tert-Butyl (lS,4S)-5-(8-((3-chloro-2-fluorophenyl)amino)-7-cyano- l,5-naphthyridin-2-yl)-2,5-diazabicyclo[2.2.1]heptane-2-carboxylate
  • reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (10 mL x 3). The combined organic layers were washed with brine (10 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue.
  • Step 1 Synthesis of tert- Butyl (6-chloropyridin-3-yl)carbamate
  • 6-chloropyridin-3 -amine 3.00 g, 23.3 mmol
  • 1,4-dioxane 30 mL
  • di-tert-butyl dicarbonate 5.60 g, 25.6 mmol, 5.90 mL
  • the reaction mixture was stirred at 100 °C for 48 hr.
  • the reaction mixture was concentrated in vacuo to give tert-butyl (6-chloropyridin-3-yl)carbamate (5.30 g, 23.1 mmol, 99%) as a brown solid.
  • Step 8 Synthesis of tert-Butyl 5-(4-((3-chloro-2-fluorophenyl)amino)-3-cyano-l,7- naphthyridin-6-yl)-3,6-dihydropyridine- 1 (2H) -carboxylate
  • Step 9 Synthesis of tert-Butyl 3-(4-((3-chloro-2-fluorophenyl)amino)-3-cyano-l,7- naphthyridin-6-yl)piperidine- 1 -carboxylate
  • Step 10 Synthesis of 4-((3-Chloro-2-fluorophenyl)amino)-6-(piperidin-3-yl)-l,7- naphthyridine-3 -carbonitrile
  • Step 11 Synthesis of 6-(l-Acryloylpiperidin-3-yl)-4-((3-chloro-2-fluorophenyl)amino)- 1 ,7-naphthyridine-3 -carbonitrile
  • Step 1 tert- Butyl (1S,4S)-5-[8-[3-chloro-4-(difhioromethoxy)-2-fluoro-anilino]-7- cyano- 1 ,5-naphthyridin-2-yl]-2,5-diazabicyclo[2.2.1 ]heptane-2-carboxylate
  • reaction mixture was quenched by water 30 mL at 25 °C, and then diluted with ethyl acetate (30 mL) and extracted with ethyl acetate (30 mL x 3). The combined organic layers were washed with water (20 mL x 2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue.
  • Step 22 4-[3-Chloro-4-(difluoromethoxy)-2-fluoro-anilino]-6-[(lS,4S)-2,5- diazabicyclo[2.2.1 ]heptan-2-yl]- 1 ,5 -naphthyridine-3-carbonitrile
  • Step 3 4-[3-Chloro-4-(difluoromethoxy)-2-fluoro-anilino]-6-[(lS,4S)-5-prop-2-enoyl-
  • Step 1 tert- Butyl (lS,4S)-5-[8-[3-chloro-4-(cyclopropylmethoxy)-2-fluoro-anilino]-7- cyano- 1 ,5-naphthyridin-2-yl]-2,5-diazabicyclo[2.2.1 ]heptane-2-carboxylate
  • reaction mixture was quenched by addition water (20 mL) at 20 °C, and then diluted with ethyl acetate (20 mL) and extracted with ethyl acetate (20 mL x 3). The combined organic layers were washed with water (10 mL x 2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue.
  • Step 22 4-[3-Chloro-4-(cyclopropylmethoxy)-2-fluoro-anilino]-6-[(lS,4S)-2,5- diazabicyclo[2.2.1 ]heptan-2-yl]- 1 ,5-naphthyridine-3-carbonitrile
  • Step 3 4-[3-Chloro-4-(cyclopropylmethoxy)-2-fluoro-anilino]-6-[(lS,4S)-5-prop-2- enoyl-2,5-diazabicyclo[2.2.1]heptan-2-yl]-l,5-naphthyridine-3-carbonitrile
  • Step 77. 6-((lS,4S)-5-Acryloyl-2,5-diazabicyclo[2.2.1]heptan-2-yl)-4-((3-chloro-4- (difluoromethoxy)-2-fluorophenyl)amino)-7 -fluoro- 1 ,5 -naphthyridine-3 -carbonitrile
  • Step 3 4-[3-Chloro-4-(difluoromethoxy)-2-fluoro-anilino]-6-[(3S)-1-prop-2- enoylpyrrolidin-3 -yl]oxy- 1 ,5-naphthyridine-3 -carbonitrile
  • Step 1 tert-Butyl (3S)-3-[[8-[3-chloro-4-(cyclopropylmethoxy)-2-fhioro-anilino]-7- cyano- 1 ,5-naphthyridin-2-yl]oxy]pyrrolidine- 1 -carboxylate
  • reaction mixture was quenched by addition water 40 mL at 25 °C, and then diluted with ethyl acetate (20 mL) and extracted with ethyl acetate (20 mL x 3). The combined organic layers were washed with water (20 mL x 2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue.
  • Step 22 4-[3-Chloro-4-(cyclopropylmethoxy)-2-fluoro-anilino]-6-[(3S)-pyrrolidin-3- yl]oxy-l,5-naphthyridine-3-carbonitrile
  • Step 3 4-[3-Chloro-4-(cyclopropylmethoxy)-2-fluoro-anilino]-6-[(3S)-l-prop-2- enoylpyrrolidin-3-yl]oxy-l,5-naphthyridine-3-carbonitrile
  • Step 2 tert-Butyl (S)-3-((7-cyano-3-fhioro-8 -hydroxy- 1, 5-naphthyridin-2 - yl)oxy)pyrrolidine- 1 -carboxylate
  • Step 44. (S)-4-((3-Chloro-4-(difluoromethoxy)-2-fluorophenyl)amino)-7-fluoro-6- (pyrrolidin-3 -yloxy)- 1 ,5-naphthyridine-3 -carbonitrile
  • Step 55.. (S)-6-((l-Acryloylpyrrolidin-3-yl)oxy)-4-((3-chloro-4-(difluoromethoxy)-2- fluorophenyl)amino)-7 -fluoro- 1 ,5-naphthyridine-3 -carbonitrile [0503] To a solution of (S)-4-((3-chloro-4-(difluoromethoxy)-2-fluorophenyl)arnino)-7-fluoro-6- (pyrrolidin-3-yloxy)-l,5-naphthyridine-3 -carbonitrile (60 mg, 106 ⁇ mol) and sodium bicarbonate (44.1 mg, 525 ⁇ mol) in anhydrous tetrahydrofuran (1 mL) and water (1 mL) was added prop-2 - enoyl chloride (9.51 mg, 106 ⁇ mol) at 0 °C and
  • Retroviral Production EGFR mutants were subcloned into pMXs-IRES-Blasticidin (RTV-016, Cell Biolabs, San Diego, CA). Retroviral expression vector retrovirus was produced by transient transfection of HEK 293T cells with the retroviral EGFR mutant expression vector pMXs-IRES-Blasticidin (RTV-016, Cell Biolabs), pCMV-Gag-Pol vector and pCMV-VSV-G- Envelope vector. Briefly, HEK 293T/17 cells were plated in 100 mm collagen coated plate (354450, Coming Life Sciences, Tewksbury, MA) (4 ⁇ 10 5 per plate) and incubated overnight.
  • retroviral plasmids (3 ⁇ g of EGFR mutant, 1.0 ⁇ g of pCMV-Gag-Pol and 0.5 ⁇ g pCMV-VSV-G) were mixed in 500 ⁇ l of Optimem (31985, Life Technologies). The mixture was incubated at room temperature for 5 min and then added to Optimem containing transfection reagent Lipofectamine (11668, Invitrogen) and incubated for 20 minutes. Mixture was then added dropwise to HEK 293T cells. The next day the medium was replaced with fresh culture medium and retrovirus was harvested @ 24 and 48 hrs.
  • EGFR mutant stable cell lines BaF3 cells (1.5E5 cells) were infected with 1 ml of viral supernatant supplemented with 8 ⁇ g/ml polybrene by centrifuging for 30 min at 1000 rpm. Cells were placed in a 37°C incubator overnight. Cells were then spun for 5 minutes to pellet the cells. Supernatant was removed and cells re-infected a fresh 1 ml of viral supernatant supplemented with 8 ⁇ g/ml polybrene by centrifuging for 30 min at 1000 rpm. Cells were placed in 37 °C incubator overnight.
  • Cells were then maintained in RPMI containing 10% Heat Inactivated FBS, 2% L-glutamine containing 10 ng/ml IL-3. After 48 hours cells were selected for retroviral infection in 10 E ⁇ /ml Blasticidin for one week. Blasticidin resistant populations were washed twice in phosphate buffered saline before plating in media lacking IL-3 to select for IL-3 independent growth.
  • Assay for cell proliferation BaF3 cell lines were resuspended at 1.3E5 c/ml in RPMI containing 10% Heat Inactivated FBS, 2% L-glutamine and 1% Pen/Strep and dispensed in triplicate (17.5E4 c/well) into 96 well plates. To determine the effect of drug on cell proliferation, cells incubated for 3 days in the presence of vehicle control or test drug at varying concentrations. Inhibition of cell growth was determined by luminescent quantification of intracellular ATP content using CellTiterGlo (Promega), according to the protocol provided by the manufacturer. Comparison of cell number on day 0 versus 72 hours post drug treatment was used to plot dose-response curves.
  • the number of viable cells was determined and normalized to vehicle-treated controls. Inhibition of proliferation, relative to vehicle-treated controls was expressed as a fraction of 1 and graphed using PRISM® software (Graphpad Software, San Diego, CA). EC50 values were determined with the same application.
  • Cellular protein analysis Cell extracts were prepared by detergent lysis (RIP A, R0278, Sigma, St Louis, MO) containing 10 mM lodoacetamide (786-228, G-Biosciences, St, Louis, MO), protease inhibitor (P8340, Sigma, St. Louis, MO) and phosphatase inhibitors (P5726, P0044, Sigma, St. Louis, MO) cocktails.
  • the soluble protein concentration was determined by micro-BSA assay (Pierce, Rockford IL). Protein immunodetection was performed by electrophoretic transfer of SDS-PAGE separated proteins to nitrocellulose, incubation with antibody, and chemiluminescent second step detection.
  • Nitrocellulose membranes were blocked with 5% nonfat dry milk in TBS and incubated overnight with primary antibody in 5% bovine serum albumin.
  • the following primary antibodies from Cell Signaling Technology were used at 1 : 1000 dilution: phospho-EGFR[Yl 173] and total EGFR.
  • ⁇ -Actin antibody used as a control for protein loading, was purchased from Sigma Chemicals.
  • Horseradish peroxidase-conjugated secondary antibodies were obtained from Cell Signaling Technology and used at 1:5000 dilution. Horseradish peroxidase-conjugated secondary antibodies were incubated in nonfat dry milk for 1 hour.
  • SuperSignal chemiluminescent reagent (Pierce Biotechnology) was used according to the manufacturer's directions and blots were imaged using the Alpha Innotech image analyzer and
  • AlphaEaseFC software (Alpha Innotech, San Leandro CA).
  • IC50 values were calculated using PRISM® software and are shown in Table B.
  • Table B assigns each compound a potency code of A, B, C, D, or E. According to the code, A represents an IC50 value ⁇ 20 nM; B represents an IC50 value >20 nM and ⁇ 50 nM; C represents an IC50 value
  • Table B Comp. No. Baf3-HER2 BT 474 H292 EGFR A431 EGFR S310F HER2 WT WT WT

Landscapes

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

Abstract

La présente divulgation concerne des composés de Formule (I) et des sels pharmaceutiquement acceptables et des stéréoisomères de ceux-ci, utiles dans le traitement de cancers associés à l'activité oncogénique ErbB, comprenant des méthodes de préparation des composés, des compositions comprenant les composés, ainsi que des méthodes d'utilisation des composés (par exemple, dans le traitement du cancer).
PCT/US2022/033962 2021-06-17 2022-06-17 Dérivés de 3-cyano-quinoléine et leurs utilisations WO2022266425A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163211913P 2021-06-17 2021-06-17
US63/211,913 2021-06-17

Publications (1)

Publication Number Publication Date
WO2022266425A1 true WO2022266425A1 (fr) 2022-12-22

Family

ID=82547625

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/033962 WO2022266425A1 (fr) 2021-06-17 2022-06-17 Dérivés de 3-cyano-quinoléine et leurs utilisations

Country Status (1)

Country Link
WO (1) WO2022266425A1 (fr)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US5763263A (en) 1995-11-27 1998-06-09 Dehlinger; Peter J. Method and apparatus for producing position addressable combinatorial libraries
WO2013152135A1 (fr) * 2012-04-04 2013-10-10 Dawei Zhang Quinoléines substituées en tant qu'inhibiteurs de tyrosine kinases de bruton
WO2019165003A1 (fr) * 2018-02-20 2019-08-29 Dawei Zhang Quinoléines substituées utiles en tant qu'inhibiteurs de kinase
WO2019233459A1 (fr) * 2018-06-08 2019-12-12 江苏威凯尔医药科技有限公司 Inhibiteur du récepteur de facteur de croissance épidermique humain, procédé de préparation correspondant et utilisation associée
WO2020219904A1 (fr) * 2019-04-25 2020-10-29 Board Of Regents, The University Of Texas System Inhibiteurs hétérocycliques de tyrosine kinase
US20210078958A1 (en) * 2017-12-19 2021-03-18 Medshine Discovery Inc. Quinazoline derivative and use thereof

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US5763263A (en) 1995-11-27 1998-06-09 Dehlinger; Peter J. Method and apparatus for producing position addressable combinatorial libraries
WO2013152135A1 (fr) * 2012-04-04 2013-10-10 Dawei Zhang Quinoléines substituées en tant qu'inhibiteurs de tyrosine kinases de bruton
US20210078958A1 (en) * 2017-12-19 2021-03-18 Medshine Discovery Inc. Quinazoline derivative and use thereof
WO2019165003A1 (fr) * 2018-02-20 2019-08-29 Dawei Zhang Quinoléines substituées utiles en tant qu'inhibiteurs de kinase
WO2019233459A1 (fr) * 2018-06-08 2019-12-12 江苏威凯尔医药科技有限公司 Inhibiteur du récepteur de facteur de croissance épidermique humain, procédé de préparation correspondant et utilisation associée
WO2020219904A1 (fr) * 2019-04-25 2020-10-29 Board Of Regents, The University Of Texas System Inhibiteurs hétérocycliques de tyrosine kinase

Non-Patent Citations (25)

* Cited by examiner, † Cited by third party
Title
"Bioreversible Carriers in Drug Design", 1987, PERGAMON PRESS
"Methods in Enzvmology", vol. 42, 1985, ACADEMIC PRESS, pages: 309 - 396
"Remington: the Science and Practice of Pharmacy", 1995, MACK PUBLISHING CO.
AUSUBEL: "Current Protocols in Molecular Biology", 2005, JOHN WILEY AND SONS, INC
CAHN ET AL., ANGEW. CHEM. INTER. EDIT., vol. 5, 1966, pages 385
CAHN ET AL., ANGEW. CHEM., vol. 78, 1966, pages 413
CAHN ET AL., EXPERIENTIA, vol. 12, 1956, pages 81
CAHN, J. CHEM. EDUC., vol. 41, 1964, pages 116
CAHNINGOLD, J. CHEM. SOC., 1951, pages 612
COLIGAN ET AL.: "Remington's Pharmaceutical Sciences", 1975, MACK PUBLISHING CO., article "The Pharmacological Basis of Therapeutics"
GREENE, T.W.WUTS, P.G. M.: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
H. BUNDGAARD ET AL., JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 77, 1988, pages 285
H. BUNDGAARD, ADVANCED DRUG DELIVERY REVIEWS, vol. 8, 1992, pages 1 - 38
H. BUNDGAARD: "A Textbook of Drug Design and Development", 1991, article "Design and Application of Pro drugs", pages: 113 - 191
L. FIESERM. FIESER: "Neser and Fieser's Reagents for Organic Synthesis", 1994, JOHN WILEY AND SONS
L. W. DEADY, SYN. COMM., vol. 7, 1977, pages 509 - 514
N. KAKEYA ET AL., CHEM. PHARM. BULL., vol. 32, 1984, pages 692
PATANILAVOIE, CHEM. REV., vol. 96, 1996, pages 3147 - 3176
R. LAROCK: "Comprehensive Organic Transformations", 1989, VCH PUBLISHERS
SAMBROOK ET AL.: "Molecular Cloning, A laboratory Manual", 2000, COLD SPRING HARBOR PRESS
SAN JUAN ET AL: "Towards predictive inhibitor design for the EGFR autophosphorylation activity", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, ELSEVIER, AMSTERDAM, NL, vol. 43, no. 4, 1 April 2008 (2008-04-01), pages 781 - 791, XP022576481, ISSN: 0223-5234 *
SMITH, M. B.MARCH, J.: "March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure", 2001, JOHN WILEY AND SONS
T. HIGUCHIV. STELLA: "A.C.S. Symposium Series", vol. 14, article "Pro-Drugs as Novel Delivery Systems"
TSOU H-R ET AL: "Optimization of 6,7-Disubstituted-4-(arylamino)quinoline-3-carbonitriles as Orally Active, Irreverible Inhibitors of HEGFR-2 Kinase Activity", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 48, 27 January 2005 (2005-01-27), pages 1107 - 1131, XP002414228, ISSN: 0022-2623, DOI: 10.1021/JM040159C *
WUTS, TW. GREENE: "Greene's Protective Groups in Organic Synthesis", 2006, JOHN WILEY & SONS

Similar Documents

Publication Publication Date Title
JP7280928B2 (ja) 脱ユビキチン化酵素(dub)阻害剤としてのスピロ縮合ピロリジン誘導体
US11319319B1 (en) Compounds for inhibiting NLRP3 and uses thereof
EP3555070B1 (fr) Composés hétérocycliques substitués par une amine utilisés comme inhibiteurs de l'ehmt2 et leurs méthodes d'utilisation
JP2023521698A (ja) Krasの標的化分解のための化合物及び方法
CN111343989A (zh) 使用ehmt2抑制剂用于免疫疗法的方法
EA023350B1 (ru) Противомикробные соединения, способы их получения и применение
WO2018028664A1 (fr) Inhibiteur de fgfr4, son procédé de préparation et son utilisation
WO2022147302A1 (fr) Dérivés de 4-phényl-indole et utilisations associées
EP3452478A1 (fr) Antimicrobiens et leurs procédés de préparation et d'utilisation
WO2022266458A1 (fr) Dérivés de 6-hétérocycloalkyle-quinazoline et leurs utilisations
WO2021127397A1 (fr) Composés hétérocycliques azotés et leurs méthodes d'utilisation
US20170291890A1 (en) Heterocycle substituted amino-pyridine compounds and methods of use thereof
EP3452479A1 (fr) Antimicrobiens et leurs procédés de préparation et d'utilisation
WO2022087422A9 (fr) Dérivés pyrrolidine-3-carboxamide et utilisations de ces derniers
JP2023544615A (ja) アセトアミド-フェニルテトラゾール誘導体およびその使用方法
WO2022266425A1 (fr) Dérivés de 3-cyano-quinoléine et leurs utilisations
CN107428682B (zh) 酰胺类衍生物、其制备方法及其在医药上的用途
WO2022170122A1 (fr) Dérivés de quinoxaline et leurs utilisations
US20220106301A1 (en) Acetamido-phenylbenzamide derivatives and methods of using the same
IL303064A (en) NEW INDAZOLE ACETYLENE DERIVATIVES
WO2022266427A1 (fr) Dérivés de 4-(aryl-méthyl-amino)-quinazoline et utilisations associées
WO2022266426A1 (fr) Dérivés de 6- (hétérocycloalkyle-oxy)-quinazoline et leurs utilisations
US20240132522A1 (en) Heterocyclic compounds and methods of use thereof
WO2024026419A1 (fr) Dérivés de quinoxaline en tant que modulateurs de pik3 alpha
WO2023250335A1 (fr) Dérivés de 1,4-diphényl-1h-indazole et de 1-pyridin-2-yl-4-phényl-1h-indazole utilisés en tant que modulateurs pd-1/pd-l1 pour traiter le cancer

Legal Events

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

Ref document number: 22741645

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22741645

Country of ref document: EP

Kind code of ref document: A1