SMARCA INHIBITORS AND USES THEREOF

Abstract
The present invention provides compounds, pharmaceutically acceptable compositions thereof, and methods of using the same for the modulation of one or more SWI/SNF-related matrix associated actin dependent regulator of chromatin subfamily A (SMARCA) and/or polybromo-1 (PB-1) protein via ubiqitination and/or degradation by compounds. The compounds are useful in treatment of cancer.
Description
TECHNICAL FIELD OF THE INVENTION

The present invention relates to compounds and methods useful for the modulation of one or more SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily A (“SMARCA”) and/or polybromo-1 (“PB1”) protein by compounds according to the present invention. The invention also provides pharmaceutically acceptable compositions comprising compounds of the present invention and methods of using said compositions in the treatment of various disorders.


BACKGROUND OF THE INVENTION

Chromatin is a complex combination of DNA and protein that makes up chromosomes. Chromatin functions to package, strengthen, and control expression and DNA replication. The chromatin structure is controlled by a series of post-translational modifications, most commonly within the “histone tails” which extend beyond the core nucleosome structure. These epigenetic modifications including acetylation, methylation, phosphorylation, ubiquitinylation, and SUMOylation, is then interpreted by the cell to allow gene specific regulation of chromatin structure and thereby transcription. Histone modifications are dynamic, as they can be added or removed in response to specific stimuli, and these modifications direct both structural changes to chromatin and alterations in gene transcription. Distinct classes of enzymes, namely histone acetyltransferases (HATs) and histone deacetylases (HDACs), acetylate or de-acetylate specific histone lysine residues (Struhl, Genes Dev. 1989, 12(5):599).


Bromodomains, which are approximately 110 amino acids long, are found in a large number of chromatin-associated proteins and have been identified in approximately 70 human proteins, often adjacent to other protein motifs (Jeanmougin et al., Trends Biochem. Sci. 1997, 22(5):151; Tamkun et al., Cell 1992, 7(3):561). Interactions between bromodomains and modified histones may be an important mechanism underlying chromatin structural changes and gene regulation. Bromodomain-containing proteins have been implicated in disease processes including cancer, inflammation and viral replication. See, e.g., Prinjha et al, Trends Pharm. Sci. 2012, 33(3):146; Muller et al. Expert Rev. 2011, 13(29):1.


Cell-type specificity and proper tissue functionality requires the tight control of distinct transcriptional programs that are intimately influenced by their environment. Alterations to this transcriptional homeostasis are directly associated with numerous disease states, most notably cancer, immuno-inflammation, neurological disorders, and metabolic diseases. Bromodomains reside within key chromatin modifying complexes that serve to control distinctive disease-associated transcriptional pathways. An example of such a complex is the switch/sucrose nonfermenting (“SWISNF”) chromatin-remodeling complex, which has been reported to be involved in gene regulation, cell lineage specification and development, and comprises a number of bromodomain containing subunits, including SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily A members 2 and 4 (SMARCA2 and SMARCA4) and polybromo-1 (PB1).


An ongoing need exists in the art for effective treatments for disease, especially hyperplasias and cancers, e.g., lung cancer. However, non-specific effects, and the inability to target and modulate certain classes of proteins altogether, such as transcription factors, remain as obstacles to the development of effective anti-cancer agents. As such, small molecule therapeutic agents that modulate or inhibit the activity of target cancer-associated proteins such as SMARCA2, SMARCA4, and PB1 hold promise as therapeutic agents. Accordingly, there remains a need to find compounds that target these proteins and are useful as therapeutic agents.


SUMMARY OF THE INVENTION

The present application relates to novel compounds, which function to modulate SMARCA and/or PB1 proteins and methods of preparation and uses thereof. In particular, the present disclosure provides compounds which find utility as modulators of SMARCA and/or PB1 proteins, which are then inhibited by the compounds as described herein. An advantage of the compounds provided herein is that a broad range of pharmacological activities is possible, consistent with the modulation of SMARCA and/or PB1 proteins. In addition, the description provides methods of using an effective amount of the compounds as described herein for the treatment or amelioration of a disease condition, such as cancer, e.g., lung cancer.


It has now been found that compounds of this invention, and pharmaceutically acceptable compositions thereof, are effective as modulators of SMARCA and/or PB1 proteins. Such compounds have the general formula I-IV:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each variable is as defined and described herein.


Compounds of the present invention, and pharmaceutically acceptable compositions thereof, are useful for treating a variety of diseases, disorders or conditions, associated with regulation of signaling pathways implicating SMARCA and/or PB1 proteins. Such diseases, disorders, or conditions include those described herein.


Compounds provided by this invention are also useful for the study of SMARCA and/or PB1 proteins in biological and pathological phenomena; the study of intracellular signal transduction pathways occurring in bodily tissues; and the comparative evaluation of new SMARCA and/or PB1 inhibitors or other regulators of cell cycling, metastasis, angiogenesis, and immune cell evasion, in vitro or in vivo.







DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS
1. General Description of Certain Embodiments of the Invention

Compounds of the present invention, and compositions thereof, are useful as modulators of SMARCA and/or PB1 proteins. In some embodiments, a provided compound is capable of modulating and/or inhibiting one or more SMARCA2, SMARCA4, or PB1 bromodomains. In some embodiments, a provided compound is capable of modulating and/or inhibiting one or more SMARCA2, SMARCA4, or PB1 ATPase domains.


In some embodiments, a provided compound modulates and/or inhibits SMARCA2. In some embodiments, a provided compound modulates and/or inhibits SMARCA4. In some embodiments, a provided compound modulates and/or inhibits PB1. In some embodiments, a provided compound modulates and/or inhibits SMARCA2 and SMARCA4. In some embodiments, a provided compound modulates and/or inhibits SMARCA2 and PB1. In some embodiments, a provided compound modulates and/or inhibits SMARCA4 and PB1. In some embodiments, a provided compound modulates and/or inhibits SMARCA2, SMARCA4, and PB1.


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




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

  • A is a nitrogen or carbon atom;
  • B is a nitrogen or carbon atom;
  • D is a nitrogen or carbon atom;
  • E is a nitrogen or carbon atom;
  • F is a carbon atom or C(O);
  • G is a carbon atom or absent;
  • H is a nitrogen atom, a carbon atom, or absent;
  • each Rx is independently hydrogen, deuterium, R6, halogen, —CN, —NO2, —OR, —SR, —NR2, —SiR3, —S(O)2R, —S(O)2NR2, —S(O)R, —C(O)R, —C(O)OR, —C(O)NR2, —C(O)N(R)OR, —C(R)2N(R)C(O)R, —C(R)2N(R)C(O)NR2, —OC(O)R, —OC(O)NR2, —OP(O)R2, —OP(O)(OR)2, —OP(O)(OR)NR2, —OP(O)(NR2)2, —N(R)C(O)OR, —N(R)C(O)R, —N(R)C(O)NR2, —N(R)S(O)2R, —N(R)P(O)R2, —N(R)P(O)(OR)2, —N(R)P(O)(OR)NR2, —N(R)P(O)(NR2)2, or —N(R)S(O)2R; or
    • two Rx groups are optionally taken together to form an optionally substituted 5-8 membered partially unsaturated or saturated ring having 0-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur; or
  • each Rx is independently hydrogen, or an optionally substituted group selected from C1-6 aliphatic, phenyl, a 4-7 membered saturated or partially unsaturated heterocyclic having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or:
    • two R groups on the same nitrogen are optionally taken together with their intervening atoms to form an optionally substituted 4-7 membered saturated, partially unsaturated, or heteroaryl ring having 0-3 heteroatoms, in addition to the nitrogen, independently selected from nitrogen, oxygen, and sulfur;
  • each Ry is independently hydrogen, deuterium, R6, halogen, —CN, —NO2, —OR, —SR, —NR2, —SiR3, —S(O)2R, —S(O)2NR2, —S(O)R, —C(O)R, —C(O)OR, —C(O)NR2, —C(O)N(R)OR, —C(R)2N(R)C(O)R, —C(R)2N(R)C(O)NR2, —OC(O)R, —OC(O)NR2, —OP(O)R2, —OP(O)(OR)2, —OP(O)(OR)NR2, —OP(O)(NR2)2, —N(R)C(O)OR, —N(R)C(O)R, —N(R)C(O)NR2, —N(R)S(O)2R, —N(R)P(O)R2, —N(R)P(O)(OR)2, —N(R)P(O)(OR)NR2, —N(R)P(O)(NR2)2, or —N(R)S(O)2R; or
    • two Ry groups are optionally taken together to form an optionally substituted 5-8 membered partially unsaturated or aryl fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;




embedded image


  • Rz is hydrogen, —NR2, —C(O)NR2, —CF3, or N;

  • each R6 is independently an optionally substituted group selected from C1-6 aliphatic, phenyl, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur;

  • each Ring P is independently phenyl, a 4-10 membered saturated or partially unsaturated mono- or bicyclic carbocyclic or heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur;

  • each Lx is independently a covalent bond or a C1-3 bivalent straight or branched saturated or unsaturated hydrocarbon chain wherein 1-3 methylene units of the chain are independently and optionally replaced with —O—, —C(O)—, —C(S)—, —C(R)2—, —CH(R)—, —C(F)2—, —N(R)—, —S(O)2—, —CH ═CH—, or —C≡C—;

  • each custom-character is a single or double bond;

  • each x and y is independently 0, 1, 2, 3, or 4; and

  • z is 1 or 2.



2. Compounds and Definitions

Compounds of the present invention include those described generally herein, and are further illustrated by the classes, subclasses, and species disclosed herein. As used herein, the following definitions shall apply unless otherwise indicated. For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75th Ed. Additionally, general principles of organic chemistry are described in “Organic Chemistry”, Thomas Sorrell, University Science Books, Sausalito: 1999, and “March's Advanced Organic Chemistry”, 5th Ed., Ed.: Smith, M. B. and March, J., John Wiley & Sons, New York: 2001, the entire contents of which are hereby incorporated by reference.


The term “aliphatic” or “aliphatic group”, as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as “carbocycle,” “cycloaliphatic” or “cycloalkyl”), that has a single point of attachment to the rest of the molecule. Unless otherwise specified, aliphatic groups contain 1-6 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-5 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms. In some embodiments, “cycloaliphatic” (or “carbocycle” or “cycloalkyl”) refers to a monocyclic C3-C6 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule. Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl.


As used herein, the term “bridged bicyclic” refers to any bicyclic ring system, i.e. carbocyclic or heterocyclic, saturated or partially unsaturated, having at least one bridge. As defined by IUPAC, a “bridge” is an unbranched chain of atoms or an atom or a valence bond connecting two bridgeheads, where a “bridgehead” is any skeletal atom of the ring system which is bonded to three or more skeletal atoms (excluding hydrogen). In some embodiments, a bridged bicyclic group has 7-12 ring members and 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Such bridged bicyclic groups are well known in the art and include those groups set forth below where each group is attached to the rest of the molecule at any substitutable carbon or nitrogen atom. Unless otherwise specified, a bridged bicyclic group is optionally substituted with one or more substituents as set forth for aliphatic groups. Additionally or alternatively, any substitutable nitrogen of a bridged bicyclic group is optionally substituted. Exemplary bridged bicyclics include:




embedded image


The term “lower alkyl” refers to a C1-4 straight or branched alkyl group. Exemplary lower alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and tert-butyl.


The term “lower haloalkyl” refers to a C1-4 straight or branched alkyl group that is substituted with one or more halogen atoms.


The term “heteroatom” means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR+ (as in N-substituted pyrrolidinyl)).


The term “unsaturated,” as used herein, means that a moiety has one or more units of unsaturation.


As used herein, the term “bivalent C1-8 (or C1-6) saturated or unsaturated, straight or branched, hydrocarbon chain”, refers to bivalent alkylene, alkenylene, and alkynylene chains that are straight or branched as defined herein.


The term “alkylene” refers to a bivalent alkyl group. An “alkylene chain” is a polymethylene group, i.e., —(CH2)n—, wherein n is a positive integer, preferably from 1 to 6, from 1 to 4, from 1 to 3, from 1 to 2, or from 2 to 3. A substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.


The term “alkenylene” refers to a bivalent alkenyl group. A substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.


As used herein, the term “cyclopropylenyl” refers to a bivalent cyclopropyl group of the following structure:




embedded image


The term “halogen” means F, Cl, Br, or I.


The term “aryl” used alone or as part of a larger moiety as in “aralkyl,” “aralkoxy,” or “aryloxyalkyl,” refers to monocyclic or bicyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members. The term “aryl” may be used interchangeably with the term “aryl ring.” In certain embodiments of the present invention, “aryl” refers to an aromatic ring system which includes, but not limited to, phenyl, biphenyl, naphthyl, anthracyl and the like, which may bear one or more substituents. Also included within the scope of the term “aryl,” as it is used herein, is a group in which an aromatic ring is fused to one or more non-aromatic rings, such as indanyl, phthalimidyl, naphthimidyl, phenanthridinyl, or tetrahydronaphthyl, and the like.


The terms “heteroaryl” and “heteroar-,” used alone or as part of a larger moiety, e.g., “heteroaralkyl,” or “heteroaralkoxy,” refer to groups having 5 to 10 ring atoms, preferably 5, 6, or 9 ring atoms; having 6, 10, or 14 π electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms. The term “heteroatom” refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen. Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl. The terms “heteroaryl” and “heteroar-”, as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring. Nonlimiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[2,3-b]-1,4-oxazin-3(4H)-one. A heteroaryl group may be mono- or bicyclic. The term “heteroaryl” may be used interchangeably with the terms “heteroaryl ring,” “heteroaryl group,” or “heteroaromatic,” any of which terms include rings that are optionally substituted. The term “heteroaralkyl” refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted.


As used herein, the terms “heterocycle,” “heterocyclyl,” “heterocyclic radical,” and “heterocyclic ring” are used interchangeably and refer to a stable 5- to 7-membered monocyclic or 7-10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above. When used in reference to a ring atom of a heterocycle, the term “nitrogen” includes a substituted nitrogen. As an example, in a saturated or partially unsaturated ring having 0-3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl), or +NR (as in N-substituted pyrrolidinyl).


A heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted. Examples of such saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenyl pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and quinuclidinyl. The terms “heterocycle,” “heterocyclyl,” “heterocyclyl ring,” “heterocyclic group,” “heterocyclic moiety,” and “heterocyclic radical,” are used interchangeably herein, and also include groups in which a heterocyclyl ring is fused to one or more aryl, heteroaryl, or cycloaliphatic rings, such as indolinyl, 3H-indolyl, chromanyl, phenanthridinyl, or tetrahydroquinolinyl. A heterocyclyl group may be mono- or bicyclic. The term “heterocyclylalkyl” refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted.


As used herein, the term “partially unsaturated” refers to a ring moiety that includes at least one double or triple bond. The term “partially unsaturated” is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined.


As described herein, compounds of the invention may contain “optionally substituted” moieties. In general, the term “substituted,” whether preceded by the term “optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an “optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds. The term “stable,” as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.


Suitable monovalent substituents on a substitutable carbon atom of an “optionally substituted” group are independently halogen; —(CH2)0-4R; —(CH2)0-4OR; —O(CH2)0-4R, —O—(CH2)0-4C(O)OR; —(CH2)0-4CH(OR)2; —(CH2)0-4SR; —(CH2)0-4Ph, which may be substituted with R; —(CH2)0-4O(CH2)0-1Ph which may be substituted with R; —CH═CHPh, which may be substituted with R; —(CH2)0-4O(CH2)0-1-pyridyl which may be substituted with R; —NO2; —CN; —N3; —(CH2)0-4N(R)2; —(CH2)0-4N(R)C(O)R; —N(R)C(S)R; —(CH2)0-4N(R)C(O)NR2; —N(R)C(S)NR2; —(CH2)0-4N(R)C(O)OR; —N(R)N(R)C(O)R; —N(R)N(R)C(O)NR2; —N(R)N(R)C(O)OR; —(CH2)0-4C(O)R; —C(S)R; —(CH2)0-4C(O)OR; —(CH2)0-4C(O)SR; —(CH2)0-4C(O)OSiR3; —(CH2)0-4OC(O)R; —OC(O)(CH2)0-4SR—, SC(S)SR; —(CH2)0-4SC(O)R; —(CH2)0-4C(O)NR2; —C(S)NR2; —C(S)SR; —SC(S)SR, —(CH2)0-4OC(O)NR2; —C(O)N(OR)R; —C(O)C(O)R; —C(O)CH2C(O)R; —C(NOR)R; —(CH2)0-4SSR; —(CH2)0-4S(O)2R; —(CH2)0-4S(O)2OR; —(CH2)0-4OS(O)2R; —S(O)2NR2; —(CH2)0-4S(O)R; —N(R)S(O)2NR2; —N(R)S(O)2R; —N(OR)R; —C(NH)NR2; —P(O)2R; —P(O)R2; —OP(O)R2; —OP(O)(OR)2; SiR3; —(C1-4 straight or branched alkylene)O—N(R)2; or —(C1-4 straight or branched alkylene)C(O)O—N(R)2, wherein each R0 may be substituted as defined below and is independently hydrogen, C1-6 aliphatic, —CH2Ph, —O(CH2)0-1Ph, —CH2-(5-6 membered heteroaryl ring), or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R, taken together with their intervening atom(s), form a 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which may be substituted as defined below.


Suitable monovalent substituents on R (or the ring formed by taking two independent occurrences of R together with their intervening atoms), are independently halogen, —(CH2)0-2R, -(haloR), —(CH2)0-2OH, —(CH2)0-2OR, —(CH2)0-2CH(OR)2; —O(haloR), —CN, —N3, —(CH2)0-2C(O)R, —(CH2)0-2C(O)OH, —(CH2)0-2C(O)OR, —(CH2)0-2SR, —(CH2)0-2SH, —(CH2)0-2NH2, —(CH2)0-2NHR, —(CH2)0-2NR2, —NO2, —SiR3, —OSiR3, —C(O)SR, —(C1-4 straight or branched alkylene)C(O)OR, or —SSR wherein each R is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently selected from C1-4 aliphatic, —CH2Ph, —O(CH2)0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of R include ═O and ═S.


Suitable divalent substituents on a saturated carbon atom of an “optionally substituted” group include the following: ═O, ═S, ═NNR*2, ═NNHC(O)R*, ═NNHC(O)OR*, ═NNHS(O)2R*, ═NR*, ═NOR*, —O(C(R*2))2-3O—, or —S(C(R*2))2-3S—, wherein each independent occurrence of R is selected from hydrogen, C1-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an “optionally substituted” group include: —O(CR*2)2-3O—, wherein each independent occurrence of R is selected from hydrogen, C1-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.


Suitable substituents on the aliphatic group of R include halogen, —R, -(haloR), —OH, —OR, —O(haloR), —CN, —C(O)OH, —C(O)OR, —NH2, —NHR, —NR2, or —NO2, wherein each R is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C1-4 aliphatic, —CH2Ph, —O(CH2)0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.


Suitable substituents on a substitutable nitrogen of an “optionally substituted” group include —R, —NR\2, —C(O)R, —C(O)OR, —C(O)C(O)R, —C(O)CH2C(O)R, —S(O)2R, —S(O)2NR\2, —C(S)NR\2, —C(NH)NR\2, or —N(RT)S(O)2R; wherein each R is independently hydrogen, C1-6 aliphatic which may be substituted as defined below, unsubstituted —OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R, taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.


Suitable substituents on the aliphatic group of R are independently halogen, —R, -(haloR), —OH, —OR, —O(haloR), —CN, —C(O)OH, —C(O)OR, —NH2, —NHR, —NR2, or —NO2, wherein each R is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C1-4 aliphatic, —CH2Ph, —O(CH2)0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.


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


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


Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention. Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present invention


As used herein, the term “provided compound” refers to any genus, subgenus, and/or species set forth herein.


As used herein, the term “and/or” is used in this disclosure to mean either “and” or “or” unless indicated otherwise.


As used herein, the term “inhibitor” is defined as a compound that binds to and/or inhibits a SMARCA and/or PB1 protein with measurable affinity. In certain embodiments, an inhibitor has an IC50 and/or binding constant of less than about 50 μM, less than about 1 μM, less than about 500 nM, less than about 100 nM, less than about 10 nM, or less than about 1 nM.


A compound of the present invention may be tethered to a detectable moiety. It will be appreciated that such compounds are useful as imaging agents. One of ordinary skill in the art will recognize that a detectable moiety may be attached to a provided compound via a suitable substituent. As used herein, the term “suitable substituent” refers to a moiety that is capable of covalent attachment to a detectable moiety. Such moieties are well known to one of ordinary skill in the art and include groups containing, e.g., a carboxylate moiety, an amino moiety, a thiol moiety, or a hydroxyl moiety, to name but a few. It will be appreciated that such moieties may be directly attached to a provided compound or via a tethering group, such as a bivalent saturated or unsaturated hydrocarbon chain. In some embodiments, such moieties may be attached via click chemistry. In some embodiments, such moieties may be attached via a 1,3-cycloaddition of an azide with an alkyne, optionally in the presence of a copper catalyst. Methods of using click chemistry are known in the art and include those described by Rostovtsev et al., Angew. Chem. Int. Ed. 2002, 41, 2596-99 and Sun et al., Bioconjugate Chem., 2006, 17, 52-57.


As used herein, the term “detectable moiety” is used interchangeably with the term “label” and relates to any moiety capable of being detected, e.g., primary labels and secondary labels. Primary labels, such as radioisotopes (e.g., tritium, 32P, 33P, 35S, or 14C), mass-tags, and fluorescent labels are signal generating reporter groups which can be detected without further modifications. Detectable moieties also include luminescent and phosphorescent groups.


The term “secondary label” as used herein refers to moieties such as biotin and various protein antigens that require the presence of a second intermediate for production of a detectable signal. For biotin, the secondary intermediate may include streptavidin-enzyme conjugates. For antigen labels, secondary intermediates may include antibody-enzyme conjugates. Some fluorescent groups act as secondary labels because they transfer energy to another group in the process of nonradiative fluorescent resonance energy transfer (FRET), and the second group produces the detected signal.


The terms “fluorescent label”, “fluorescent dye”, and “fluorophore” as used herein refer to moieties that absorb light energy at a defined excitation wavelength and emit light energy at a different wavelength. Examples of fluorescent labels include, but are not limited to: Alexa Fluor dyes (Alexa Fluor 350, Alexa Fluor 488, Alexa Fluor 532, Alexa Fluor 546, Alexa Fluor 568, Alexa Fluor 594, Alexa Fluor 633, Alexa Fluor 660 and Alexa Fluor 680), AMCA, AMCA-S, BODIPY dyes (BODIPY FL, BODIPY R6G, BODIPY TMR, BODIPY TR, BODIPY 530/550, BODIPY 558/568, BODIPY 564/570, BODIPY 576/589, BODIPY 581/591, BODIPY 630/650, BODIPY 650/665), Carboxyrhodamine 6G, carboxy-X-rhodamine (ROX), Cascade Blue, Cascade Yellow, Coumarin 343, Cyanine dyes (Cy3, Cy5, Cy3.5, Cy5.5), Dansyl, Dapoxyl, Dialkylaminocoumarin, 4′,5′-Dichloro-2′,7′-dimethoxy-fluorescein, DM-NERF, Eosin, Erythrosin, Fluorescein, FAM, Hydroxycoumarin, IRDyes (IRD40, IRD 700, IRD 800), JOE, Lissamine rhodamine B, Marina Blue, Methoxycoumarin, Naphthofluorescein, Oregon Green 488, Oregon Green 500, Oregon Green 514, Pacific Blue, PyMPO, Pyrene, Rhodamine B, Rhodamine 6G, Rhodamine Green, Rhodamine Red, Rhodol Green, 2′,4′,5′,7′-Tetra-bromosulfone-fluorescein, Tetramethyl-rhodamine (TMR), Carboxytetramethylrhodamine (TAMRA), Texas Red, Texas Red-X.


The term “mass-tag” as used herein refers to any moiety that is capable of being uniquely detected by virtue of its mass using mass spectrometry (MS) detection techniques. Examples of mass-tags include electrophore release tags such as N-[3-[4′-[(p-Methoxytetrafluorobenzyl)oxy]phenyl]-3-methylglyceronyl]isonipecotic Acid, 4′-[2,3,5,6-Tetrafluoro-4-(pentafluorophenoxyl)]methyl acetophenone, and their derivatives. The synthesis and utility of these mass-tags is described in U.S. Pat. Nos. 4,650,750, 4,709,016, 5,360,8191, 5,516,931, 5,602,273, 5,604,104, 5,610,020, and 5,650,270. Other examples of mass-tags include, but are not limited to, nucleotides, dideoxynucleotides, oligonucleotides of varying length and base composition, oligopeptides, oligosaccharides, and other synthetic polymers of varying length and monomer composition. A large variety of organic molecules, both neutral and charged (biomolecules or synthetic compounds) of an appropriate mass range (100-2000 Daltons) may also be used as mass-tags.


The terms “measurable affinity” and “measurably inhibit,” as used herein, means a measurable change in a SMARCA and/or PB1 protein activity between a sample comprising a compound of the present invention, or composition thereof, and a SMARCA and/or PB1 protein, and an equivalent sample comprising a SMARCA and/or PB1 protein, in the absence of said compound, or composition thereof.


3. Description of Exemplary Embodiments

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




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

  • A is a nitrogen or carbon atom;
  • B is a nitrogen or carbon atom;
  • D is a nitrogen or carbon atom;
  • E is a nitrogen or carbon atom;
  • F is a carbon atom or C(O);
  • G is a carbon atom or absent;
  • H is a nitrogen atom, a carbon atom, or absent;
  • each Rx is independently hydrogen, deuterium, R6, halogen, —CN, —NO2, —OR, —SR, —NR2, —SiR3, —S(O)2R, —S(O)2NR2, —S(O)R, —C(O)R, —C(O)OR, —C(O)NR2, —C(O)N(R)OR, —C(R)2N(R)C(O)R, —C(R)2N(R)C(O)NR2, —OC(O)R, —OC(O)NR2, —OP(O)R2, —OP(O)(OR)2, —OP(O)(OR)NR2, —OP(O)(NR2)2, —N(R)C(O)OR, —N(R)C(O)R, —N(R)C(O)NR2, —N(R)S(O)2R, —N(R)P(O)R2, —N(R)P(O)(OR)2, —N(R)P(O)(OR)NR2, —N(R)P(O)(NR2)2, or —N(R)S(O)2R; or
    • two Rx groups are optionally taken together to form an optionally substituted 5-8 membered partially unsaturated or saturated ring having 0-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur; or
  • each R is independently hydrogen, or an optionally substituted group selected from C1-6 aliphatic, phenyl, a 4-7 membered saturated or partially unsaturated heterocyclic having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or:
    • two R groups on the same nitrogen are optionally taken together with their intervening atoms to form an optionally substituted 4-7 membered saturated, partially unsaturated, or heteroaryl ring having 0-3 heteroatoms, in addition to the nitrogen, independently selected from nitrogen, oxygen, and sulfur;
  • each Ry is independently hydrogen, deuterium, R6, halogen, —CN, —NO2, —OR, —SR, —NR2, —SiR3, —S(O)2R, —S(O)2NR2, —S(O)R, —C(O)R, —C(O)OR, —C(O)NR2, —C(O)N(R)OR, —C(R)2N(R)C(O)R, —C(R)2N(R)C(O)NR2, —OC(O)R, —OC(O)NR2, —OP(O)R2, —OP(O)(OR)2, —OP(O)(OR)NR2, —OP(O)(NR2)2, —N(R)C(O)OR, —N(R)C(O)R, —N(R)C(O)NR2, —N(R)S(O)2R, —N(R)P(O)R2, —N(R)P(O)(OR)2, —N(R)P(O)(OR)NR2, —N(R)P(O)(NR2)2, or —N(R)S(O)2R; or
    • two Ry groups are optionally taken together to form an optionally substituted 5-8 membered partially unsaturated or aryl fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;




embedded image


  • Rz is hydrogen, —NR2, —C(O)NR2, —CF3, or N;

  • each R6 is independently an optionally substituted group selected from C1-6 aliphatic, phenyl, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur;

  • each Ring P is independently phenyl, a 4-10 membered saturated or partially unsaturated mono- or bicyclic carbocyclic or heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur;

  • each Lx is independently a covalent bond or a C1-3 bivalent straight or branched saturated or unsaturated hydrocarbon chain wherein 1-3 methylene units of the chain are independently and optionally replaced with —O—, —C(O)—, —C(S)—, —C(R)2—, —CH(R)—, —C(F)2—, —N(R)—, —S(O)2—, —CH ═CH—, or —C≡C—;

  • each custom-character is a single or double bond;

  • each x and y is independently 0, 1, 2, 3, or 4; and

  • z is 1 or 2.



As provided herein, a compound




embedded image


represents, for example




embedded image


As defined above and described herein, in some embodiments, A is a nitrogen or carbon atom.


In some embodiments, A is a nitrogen atom. In some embodiments, A is a carbon atom.


In some embodiments, A is selected from those depicted in Table 1, below.


As defined above and described herein, in some embodiments, B is a nitrogen or carbon atom.


In some embodiments, B is a nitrogen atom. In some embodiments, B is a carbon atom.


In some embodiments, B is selected from those depicted in Table 1, below.


As defined above and described herein, in some embodiments, D is a nitrogen or carbon atom.


In some embodiments, D is a nitrogen atom. In some embodiments, D is a carbon atom.


In some embodiments, D is selected from those depicted in Table 1, below.


As defined above and described herein, in some embodiments, E is a nitrogen or carbon atom.


In some embodiments, E is a nitrogen atom. In some embodiments, E is a carbon atom.


In some embodiments, E is selected from those depicted in Table 1, below.


As defined above and described herein, in some embodiments, F is a carbon atom or C(O).


In some embodiments, F is a carbon atom. In some embodiments, F is C(O).


In some embodiments, F is selected from those depicted in Table 1, below.


As defined above and described herein, in some embodiments, G is a carbon atom or absent.


In some embodiments, G is a carbon atom. In some embodiments, G is absent.


In some embodiments, G is selected from those depicted in Table 1, below.


As defined above and described herein, in some embodiments, H is a nitrogen atom, a carbon atom, or absent


In some embodiments, H is a carbon atom. In some embodiments, H is a nitrogen atom. In some embodiments, H is absent.


In some embodiments, H is selected from those depicted in Table 1, below.


As defined above and described herein, in some embodiments, each Rx is independently hydrogen, deuterium, R6, halogen, —CN, —NO2, —OR, —SR, —NR2, —SiR3, —S(O)2R, —S(O)2NR2, —S(O)R, —C(O)R, —C(O)OR, —C(O)NR2, —C(O)N(R)OR, —C(R)2N(R)C(O)R, —C(R)2N(R)C(O)NR2, —OC(O)R, —OC(O)NR2, —OP(O)R2, —OP(O)(OR)2, —OP(O)(OR)NR2, —OP(O)(NR2)2, —N(R)C(O)OR, —N(R)C(O)R, —N(R)C(O)NR2, —N(R)S(O)2R, —NP(O)R2, —N(R)P(O)(OR)2, —N(R)P(O)(OR)NR2, —N(R)P(O)(NR2)2, or —N(R)S(O)2R; or two Rx groups are optionally taken together to form a 5-8 membered partially unsaturated or saturated ring having 0-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.


In some embodiments, Rx is hydrogen. In some embodiments, Rx is deuterium. In some embodiments, Rx is R6. In some embodiments, Rx is halogen. In some embodiments, Rx is —CN. In some embodiments, Rx is —NO2. In some embodiments, Rx is —OR. In some embodiments, Rx is —SR. In some embodiments, Rx is —NR2. In some embodiments, Rx is —SiR3. In some embodiments, Rx is —S(O)2R. In some embodiments, Rx is —S(O)2NR2. In some embodiments, Rx is —S(O)R. In some embodiments, Rx is —C(O)R. In some embodiments, Rx is —C(O)OR. In some embodiments, Rx is —C(O)NR2. In some embodiments, Rx is —C(O)N(R)OR. In some embodiments, Rx is —C(R)2N(R)C(O)R. In some embodiments, Rx is —C(R)2N(R)C(O)NR2. In some embodiments, Rx is —OC(O)R. In some embodiments, Rx is —OC(O)NR2. In some embodiments, Rx is —OP(O)R2. In some embodiments, Rx is —OP(O)(OR)2. In some embodiments, Rx is —OP(O)(OR)NR2. In some embodiments, Rx is —OP(O)(NR2)2. In some embodiments, Rx is —N(R)C(O)OR. In some embodiments, Rx is —N(R)C(O)R. In some embodiments, Rx is —N(R)C(O)NR2. In some embodiments, Rx is —N(R)S(O)2R. In some embodiments, Rx is —NP(O)R2. In some embodiments, Rx is —N(R)P(O)(OR)2. In some embodiments, Rx is —N(R)P(O)(OR)NR2. In some embodiments, Rx is —N(R)P(O)(NR2)2. In some embodiments, Rx is —N(R)S(O)2R. In some embodiments, two Rx groups are optionally taken together to form an optionally substituted 5-8 membered partially unsaturated or aryl fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.


In some embodiments, Rx is




embedded image


In some embodiments, Rx is




embedded image


In some embodiments, Rx is




embedded image


In some embodiments, Rx is —CO2tBu. In some embodiments, Rx is




embedded image


In some embodiments, Rx is —OH. In some embodiments, Rx is




embedded image


In some embodiments, Rx is




embedded image


In some embodiments, Rx is Cl. In some embodiments, Rx is —CH3. In some embodiments, Rx is




embedded image


In some embodiments, Rx is —CH3. In some embodiments, Rx is




embedded image


In some embodiments, each Rx is selected from those depicted in Table 1, below.


As defined above and described herein, in some embodiments, each R is independently hydrogen, or an optionally substituted group selected from C1-6 aliphatic, phenyl, a 4-7 membered saturated or partially unsaturated heterocyclic having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or two R groups on the same nitrogen are optionally taken together with their intervening atoms to form a 4-7 membered saturated, partially unsaturated, or heteroaryl ring having 0-3 heteroatoms, in addition to the nitrogen, independently selected from nitrogen, oxygen, and sulfur.


In some embodiments, R is independently hydrogen. In some embodiments, R is an optionally substituted group selected from C1-6 aliphatic. In some embodiments, R is an optionally substituted phenyl. In some embodiments, R is an optionally substituted 4-7 membered saturated or partially unsaturated heterocyclic having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an optionally substituted 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, two R groups on the same nitrogen are optionally taken together with their intervening atoms to form an optionally substituted 4-7 membered saturated, partially unsaturated, or heteroaryl ring having 0-3 heteroatoms, in addition to the nitrogen, independently selected from nitrogen, oxygen, and sulfur.


In some embodiments, R is selected from those depicted in Table 1, below.


As defined above and described herein, in some embodiments, each Ry is independently hydrogen, deuterium, R6, halogen, —CN, —NO2, —OR, —SR, —NR2, —SiR3, —S(O)2R, —S(O)2NR2, —S(O)R, —C(O)R, —C(O)OR, —C(O)NR2, —C(O)N(R)OR, —C(R)2N(R)C(O)R, —C(R)2N(R)C(O)NR2, —OC(O)R, —OC(O)NR2, —OP(O)R2, —OP(O)(OR)2, —OP(O)(OR)NR2, —OP(O)(NR2)2, —N(R)C(O)OR, —N(R)C(O)R, —N(R)C(O)NR2, —N(R)S(O)2R, —NP(O)R2, —N(R)P(O)(OR)2, —N(R)P(O)(OR)NR2, —N(R)P(O)(NR2)2, or —N(R)S(O)2R; or two Ry groups are optionally taken together to form a 5-8 membered partially unsaturated or fused aryl ring having 0-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.


In some embodiments, Ry is hydrogen. In some embodiments, Ry is deuterium. In some embodiments, Ry is R6. In some embodiments, Ry is halogen. In some embodiments, Ry is —CN. In some embodiments, Ry is —NO2. In some embodiments, Ry is —OR. In some embodiments, Ry is —SR. In some embodiments, Ry is —NR2. In some embodiments, Ry is —SiR3. In some embodiments, Ry is —S(O)2R. In some embodiments, Ry is —S(O)2NR2. In some embodiments, Ry is —S(O)R. In some embodiments, Ry is —C(O)R. In some embodiments, Ry is —C(O)OR. In some embodiments, Ry is —C(O)NR2. In some embodiments, Ry is —C(O)N(R)OR. In some embodiments, Ry is —C(R)2N(R)C(O)R. In some embodiments, Ry is —C(R)2N(R)C(O)NR2. In some embodiments, Ry is —OC(O)R. In some embodiments, Ry is —OC(O)NR2. In some embodiments, Ry is —OP(O)R2. In some embodiments, Ry is —OP(O)(OR)2. In some embodiments, Ry is —OP(O)(OR)NR2. In some embodiments, Ry is —OP(O)(NR2)2. In some embodiments, Ry is —N(R)C(O)OR. In some embodiments, Ry is —N(R)C(O)R. In some embodiments, Ry is —N(R)C(O)NR2. In some embodiments, Ry is —N(R)S(O)2R. In some embodiments, Ry is —NP(O)R2. In some embodiments, Ry is —N(R)P(O)(OR)2. In some embodiments, Ry is —N(R)P(O)(OR)NR2. In some embodiments, Ry is —N(R)P(O)(NR2)2. In some embodiments, Ry is —N(R)S(O)2R. In some embodiments, two Ry groups are optionally taken together to form an optionally substituted 5-8 membered partially unsaturated or aryl fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.


In some embodiments, Ry is




embedded image


In some embodiments, Ry is




embedded image


In some embodiments, Ry is —NH2.


In some embodiments, each Ry is selected from those depicted in Table 1, below.


As defined above and described herein, in some embodiments, Rz is hydrogen, —NR2, —C(O)NR2, —CF3, or




embedded image


In some embodiments, Rz is hydrogen. In some embodiments, Rz is —NR2. In some embodiments, Rz is —C(O)NR2. In some embodiments, Rz is —CF3. In some embodiments, Rz is




embedded image


In some embodiments, Rz is —NH2. In some embodiments, Rz is —C(O)NH2.


In some embodiments, each Rz is selected from those depicted in Table 1, below.


As defined above and described herein, in some embodiments, each R6 is independently an optionally substituted group selected from C1-6 aliphatic, phenyl, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.


In some embodiments, each R6 is independently an optionally substituted C1-6 aliphatic. In some embodiments, each R6 is independently an optionally substituted phenyl. In some embodiments, each R6 is independently an optionally substituted 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, each R6 is independently an optionally substituted 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.


In some embodiments, each R6 is selected from those depicted in Table 1, below.


As generally defined above and described herein, in some embodiments, each Ring P is phenyl, a 4-10 membered saturated or partially unsaturated mono- or bicyclic carbocyclic or heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, Ring A is optionally substituted with 1-2 oxo groups.


In some embodiments, Ring P is phenyl. In some embodiments, Ring P is a 4-10 membered saturated or partially unsaturated mono- or bicyclic carbocyclic or heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, Ring P is a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, Ring P is optionally substituted with 1-2 oxo groups.


In some embodiments, Ring P is




embedded image


In some embodiments, Ring P is




embedded image


In some embodiments, Ring P is




embedded image


In some embodiments, Ring P is




embedded image


In some embodiments, Ring P is




embedded image


In some embodiments, Ring P is




embedded image


In some embodiments, Ring P is




embedded image


In some embodiments, Ring P is




embedded image


In some embodiments, Ring P is




embedded image


In some embodiments, Ring P is




embedded image


In some embodiments, Ring P is




embedded image


In some embodiments, each Ring P is selected from those depicted in Table 1, below.


As generally defined above and described herein, each IP is a bivalent moiety selected from a covalent bond or a C1.3 bivalent straight or branched saturated or unsaturated hydrocarbon chain wherein 1-3 methylene units of the chain are independently and optionally replaced with —O—, —C(O)—, —C(S)—, —C(R)2—, —CH(R)—, —C(F)2—, —N(R)—, —S(O)2—, —CH═CH—, or —C≡C—.


In some embodiments, Lx a covalent bond. In some embodiments, Lx is a C1-3 bivalent straight or branched saturated or unsaturated hydrocarbon chain wherein 1-3 methylene units of the chain are independently and optionally replaced with —O—, —C(O)—, —C(S)—, —C(R)2—, —CH(R)—, —C(F)2—, —N(R)—, —S(O)2—, —CH═CH—, or —C≡C—.


In some embodiments, Lx is —CH2—. In some embodiments, Lx is —NRC(O)—. In some embodiments, Lx is —C(O)NR—. In some embodiments, Lx is —C≡C—. In some embodiments, Lx is




embedded image


In some embodiments, each Lx is selected from those depicted in Table 1, below.


As defined above and described herein, in some embodiments, each custom-character is a single or double bond.


In some embodiments, custom-character is a single. In some embodiments, custom-character is a double bond.


In some embodiments, custom-character is selected from those depicted in Table 1, below.


As defined above and described herein, each x is independently 0, 1, 2, 3, or 4.


In some embodiments, x is 0. In some embodiments, x is 1. In some embodiments, x is 2. In some embodiments, x is 3. In some embodiments, x is 4.


In some embodiments, each x is selected from those depicted in Table 1, below.


As defined above and described herein, y is independently 0, 1, 2, 3, or 4.


In some embodiments, y is 0. In some embodiments, y is 1. In some embodiments, y is 2. In some embodiments, y is 3. In some embodiments, y is 4.


In some embodiments, y is selected from those depicted in Table 1, below.


As defined above and described herein, z is 1 or 2.


In some embodiments, z is 1. In some embodiments, z is 2.


In some embodiments, z is selected from those depicted in Table 1, below.


In some embodiments, the present invention provides the compound of formula I wherein A and E are carbon atoms, and B and D are nitrogen atoms, thereby forming a compound of formula I-a:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of Rx, Ry, Ring P, L, x, y, and z is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present invention provides the compound of formula I wherein A and E are carbon atoms, and B and D are nitrogen atoms, and z is 1, thereby forming a compound of formula I-b:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of Rx, Ry, Ring P, Lx, x, and y is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present invention provides the compound of formula I wherein A and E are carbon atoms, and B and D are nitrogen atoms, Ring P is phenyl, Lx is a covalent bond, and z is 2, thereby forming a compound of formula I-c:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of Rx, Ry, Ring P, Lx, x, and y is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present invention provides the compound of formula I wherein A and E are carbon atoms, and B and D are nitrogen atoms, Ring P is phenyl, Lx is a covalent bond, and z is 1, thereby forming a compound of formula I-d:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of Rx, Ry, x, and y is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present invention provides the compound of formula I wherein A and E are carbon atoms, and B and D are nitrogen atoms, Ring P is phenyl and piperidine, Lx is a covalent bond, and z is 2, thereby forming a compound of formula I-e:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of Rx, Ry, x, and y is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present invention provides the compound of formula II wherein A and B are carbon atoms, D is a nitrogen atom, and Rz is —NH2, thereby forming a compound of formula II-a:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of Rx, Ry, Ring P, Lx, x, y, and z is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present invention provides the compound of formula II wherein A and B are carbon atoms, D is a nitrogen atom, Ry is hydrogen, Rz is —NH2, y is 1, and z is 2, thereby forming a compound of formula II-b:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of Rx, Ring P, Lx, and x is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present invention provides the compound of formula III wherein A, B, D, G, and H are carbon atoms, and F is C(O), thereby forming a compound of formula III-a:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of Rx, Ry, Ring P, Lx, x, y, and z is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present invention provides the compound of formula III wherein A, B, D, G, and H are carbon atoms, and F is C(O), and z is 1, thereby forming a compound of formula III-b:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of Rx, Ry, Ring P, Lx, x, and y is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present invention provides the compound of formula III wherein A, B, and D are carbon atoms, F is C(O), G and H are absent, and z is 1, thereby forming a compound of formula III-c:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of Rx, Ry, Ring P, L, x, and y is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present invention provides the compound of formula IV wherein Ry is hydrogen, y is 1, and z is 2, thereby forming a compound of formula IV-a:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of Rx, Ring P, LP, and x is as defined above and described in embodiments herein, both singly and in combination.


Exemplary compounds of the invention are set forth in Table 1, below.









TABLE 1







Exemplary Compounds








I-#
Structure





I-1


embedded image







I-2


embedded image







I-3


embedded image







I-4


embedded image







I-5


embedded image







I-6


embedded image







I-7


embedded image







I-8


embedded image







I-9


embedded image







I-10


embedded image







I-11


embedded image







I-12


embedded image







I-13


embedded image







I-14


embedded image







I-15


embedded image







I-16


embedded image







I-17


embedded image







I-18


embedded image







I-19


embedded image







I-20


embedded image







I-21


embedded image







I-22


embedded image







I-23


embedded image







I-24


embedded image







I-25


embedded image







I-26


embedded image







I-27


embedded image







I-28


embedded image







I-29


embedded image







I-30


embedded image







I-31


embedded image







I-32


embedded image







I-33


embedded image







I-34


embedded image







I-35


embedded image







I-36


embedded image







I-37


embedded image







I-38


embedded image







I-39


embedded image







I-40


embedded image







I-41


embedded image







I-42


embedded image







I-43


embedded image







I-44


embedded image











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


4. General Methods of Providing the Present Compounds

The compounds of this invention may be prepared or isolated in general by synthetic and/or semi-synthetic methods known to those skilled in the art for analogous compounds and by methods described in detail in the Examples, herein.


In the Schemes below, where a particular protecting group, leaving group, or transformation condition is depicted, one of ordinary skill in the art will appreciate that other protecting groups, leaving groups, and transformation conditions are also suitable and are contemplated. Such groups and transformations are described in detail in March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, M. B. Smith and J. March, 5th Edition, John Wiley & Sons, 2001, Comprehensive Organic Transformations, R. C. Larock, 2nd Edition, John Wiley & Sons, 1999, and Protecting Groups in Organic Synthesis, T. W. Greene and P. G. M. Wuts, 3rd edition, John Wiley & Sons, 1999, the entirety of each of which is hereby incorporated herein by reference.


As used herein, the phrase “oxygen protecting group” includes, for example, carbonyl protecting groups, hydroxyl protecting groups, etc. Hydroxyl protecting groups are well known in the art and include those described in detail in Protecting Groups in Organic Synthesis, T. W. Greene and P. G. M. Wuts, 3rd edition, John Wiley & Sons, 1999, the entirety of each of which is herein incorporated by reference. Examples of suitable hydroxyl protecting groups include, but are not limited to, esters, allyl ethers, ethers, silyl ethers, alkyl ethers, arylalkyl ethers, and alkoxyalkyl ethers. Examples of such esters include formates, acetates, carbonates, and sulfonates. Specific examples include formate, benzoyl formate, chloroacetate, trifluoroacetate, methoxyacetate, triphenylmethoxyacetate, p-chlorophenoxyacetate, 3-phenylpropionate, 4-oxopentanoate, 4,4-(ethylenedithio)pentanoate, pivaloate (trimethylacetyl), crotonate, 4-methoxy-crotonate, benzoate, p-benylbenzoate, 2,4,6-trimethylbenzoate, carbonates such as methyl, 9-fluorenylmethyl, ethyl, 2,2,2-trichloroethyl, 2-(trimethylsilyl)ethyl, 2-(phenylsulfonyl)ethyl, vinyl, allyl, and p-nitrobenzyl. Examples of such silyl ethers include trimethylsilyl, triethylsilyl, t-butyldimethylsilyl, t-butyldiphenylsilyl, triisopropylsilyl, and other trialkylsilyl ethers. Alkyl ethers include methyl, benzyl, p-methoxybenzyl, 3,4-dimethoxybenzyl, trityl, t-butyl, allyl, and allyloxycarbonyl ethers or derivatives. Alkoxyalkyl ethers include acetals such as methoxymethyl, methylthiomethyl, (2-methoxyethoxy)methyl, benzyloxymethyl, beta-(trimethylsilyl)ethoxymethyl, and tetrahydropyranyl ethers. Examples of arylalkyl ethers include benzyl, p-methoxybenzyl (MPM), 3,4-dimethoxybenzyl, O-nitrobenzyl, p-nitrobenzyl, p-halobenzyl, 2,6-dichlorobenzyl, p-cyanobenzyl, and 2- and 4-picolyl.


Amino protecting groups are well known in the art and include those described in detail in Protecting Groups in Organic Synthesis, T. W. Greene and P. G. M. Wuts, 3rd edition, John Wiley & Sons, 1999, the entirety of each of which is herein incorporated by reference. Suitable amino protecting groups include, but are not limited to, aralkylamines, carbamates, cyclic imides, allyl amines, amides, and the like. Examples of such groups include t-butyloxycarbonyl (BOC), ethyloxycarbonyl, methyloxycarbonyl, trichloroethyloxycarbonyl, allyloxycarbonyl (Alloc), benzyloxocarbonyl (CBZ), allyl, phthalimide, benzyl (Bn), fluorenylmethylcarbonyl (Fmoc), formyl, acetyl, chloroacetyl, dichloroacetyl, trichloroacetyl, phenylacetyl, trifluoroacetyl, benzoyl, and the like.


5. Uses, Formulation and Administration

Pharmaceutically Acceptable Compositions


According to another embodiment, the invention provides a composition comprising a compound of this invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle. The amount of compound in compositions of this invention is such that is effective to measurably modulate and/or inhibit a one or more SMARCA and/or PB1 protein, or a mutant thereof, in a biological sample or in a patient. In certain embodiments, the amount of compound in compositions of this invention is such that is effective to measurably modulate and/or inhibit one or more SMARCA and/or PB1 protein, or a mutant thereof, in a biological sample or in a patient. In certain embodiments, a composition of this invention is formulated for administration to a patient in need of such composition. In some embodiments, a composition of this invention is formulated for oral administration to a patient.


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


The term “pharmaceutically acceptable carrier, adjuvant, or vehicle” refers to a non-toxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.


A “pharmaceutically acceptable derivative” means any non-toxic salt, ester, salt of an ester or other derivative of a compound of this invention that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof.


As used herein, the term “inhibitorily active metabolite or residue thereof” means that a metabolite or residue thereof is also an inhibitor of one or more SMARCA and/or PB1 protein, or a mutant thereof.


Compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term “parenteral” as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.


For this purpose, any bland fixed oil may be employed including synthetic mono- or di-glycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used surfactants, such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.


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


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


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


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


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


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


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


Most preferably, pharmaceutically acceptable compositions of this invention are formulated for oral administration. Such formulations may be administered with or without food. In some embodiments, pharmaceutically acceptable compositions of this invention are administered without food. In other embodiments, pharmaceutically acceptable compositions of this invention are administered with food.


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


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


Uses of Compounds and Pharmaceutically Acceptable Compositions


Compounds and compositions described herein are generally useful for the modulation and/or inhibition of a SMARCA and/or PB1 protein activity.


Examples of SMARCA proteins that are degraded and/or inhibited by the compounds and compositions described herein and against which the methods described herein are useful include those of the SWI/SNF-related matrix-associated actin-dependent regulators of chromatin subfamily A (“SMARCA”) family of proteins, the members of which include SMARCA1, SMARCA2, SMARCA4, or SMARCA5, or a mutant thereof. See e.g., Shain and Pollack “The Spectrum of SWI/SNF Mutations, Ubiquitous in Human Cancers. PLoS One 2013, 8:e55119; Kadoch and Crabtree “Mammalian SWI/SNF Chromatin Remodeling Complexes and Cancer: Mechanistic Insights Gained from Human Genomics” Sci. Adv. 2015, 1:e1500447; Wilson and Roberts, SWI/SNF Nucleosome Remodellers and Cancer” Nat. Rev. Cancer 2011, 11:481; and Son and Crabtree “The Role of BAF (mSWI/SNF) Complexes in Mammalian Neural Development” Am. J. Med. Genet., Part C 2014, 166:333, the entirety of each of which is herein incorporated by reference.


The activity of a compound utilized in this invention as a modulator and/or inhibitor of one or more SMARCA2, SMARCA4, or PB1, or a mutant thereof, may be assayed in vitro, in vivo or in a cell line. In vitro assays include assays that determine inhibition of either the activity and/or the subsequent functional consequences of activated SMARCA or PB1 protein, or a mutant thereof. Alternate in vitro assays quantitate the ability of the inhibitor to bind to a SMARCA or PB1 protein. Inhibitor binding may be measured by radiolabeling the inhibitor prior to binding, isolating the inhibitor/SMARCA or PB1 complex and determining the amount of radiolabel bound. Alternatively, inhibitor binding may be determined by running a competition experiment where new inhibitors are incubated with a SMARCA of PB1 protein bound to known radioligands. Representative in vitro and in vivo assays useful in assaying a SMARCA or PB1 inhibitor include those described and disclosed in, e.g., Tanaka et al. “Design and Characterization of Bivalent BET Inhibitors” Nat. Chem. Biol. 2016, 12(12):1089; Schiaffino-Ortega et al. “SWI/SNF as targets in cancer therapy” J. Hematol. Oncol. 2014, 7:81; Filippakopoulos et al. “Histone Recognition and Large-Scale Structural Analysis of the Human Bromodomain Family” Cell 2012, 149:214. Detailed conditions for assaying a compound utilized in this invention as a modulator and/or inhibitor of a SMARCA or PB1 protein, or a mutant thereof, are set forth in the Examples below.


SMARCA2 and SMARCA4, also known as transcription activators Brahma homologue (BRM) and Brahma-related gene 1 (BRG1) respectively, are mutually exclusive helicase/ATPase proteins of the large ATP-dependent SWI/SNF chromatin-remodeling complexes involved in transcriptional regulation of gene expression. SMARCA2, SMARCA4, and PB1 are bromodomain and/or ATPase domain containing subunits of the larger SWI/SNF chromatin-remodeling complexes. In some embodiments, a provided compound binds to one or more SMARCA2, SMARCA4, or PB1 bromodomains. In some embodiments, a provided compound binds to one or more SMARCA2, SMARCA4, or PB1 ATPase domains.


Representative SMARCA2, SMARCA4, and/or PB1 inhibitors include those described and disclosed in e.g., Gerstenberger et al. J. Med. Chem. 2016, 59(10):4800; Theodoulou et al. Curr. Opin. Chem. Bio. 2016, 33:58; Vangamudi et al. Cancer Res. 2015, 75(18):3865; the entirety of each of which is herein incorporated by reference.


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


Provided compounds are modulators and/or inhibitors of one of more SMARCA and/or PB1 protein and are therefore useful for treating one or more disorders associated with activity of one or more SMARCA and/or PB1 protein. Thus, in certain embodiments, the present invention provides a method for treating a SMARCA2-mediated, SMARCA4-mediated, or PB1-mediated disorder comprising the step of administering to a patient in need thereof a compound of the present invention, or pharmaceutically acceptable composition thereof.


As used herein, the terms “SMARCA2-mediated”, “SMARCA4-mediated”, or “PB1-mediated” disorders, diseases, and/or conditions as used herein means any disease or other deleterious condition in which one or more SMARCA2, SMARCA4, or PB1, or a mutant thereof, are known to play a role. Accordingly, another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which one or more SMARCA2, SMARCA4, or PB1, or a mutant thereof, are known to play a role.


In some embodiments, the present invention provides a method for treating one or more disorders, diseases, and/or conditions wherein the disorder, disease, or condition is a cancer, a neurodegenerative disorder, a viral disease, an autoimmune disease, an inflammatory disorder, a hereditary disorder, a hormone-related disease, a metabolic disorder, conditions associated with organ transplantation, immunodeficiency disorders, a destructive bone disorder, a proliferative disorder, an infectious disease, a condition associated with cell death, thrombin-induced platelet aggregation, liver disease, pathologic immune conditions involving T cell activation, a cardiovascular disorder, or a CNS disorder.


Diseases and conditions treatable according to the methods of this invention include, but are not limited to, cancer (see, e.g., Schiaffino-Ortega et al. J. Hematol. Oncol. 2014, 7:81; Medina et al. Gene Chromosome Canc. 2014, 41:170), diabetes, cardiovascular disease (see, e.g., Bevilacqua et al., Cardiovasc. Pathol. 2013, 23(2):85), viral disease, autoimmune diseases such as lupus, and rheumatoid arthritis, autoinflammatory syndromes, atherosclerosis (see, e.g., Ortiz-Mao et al., J. Proteom Genom Res. 2017, 2(1):1), psoriasis, allergic disorders, inflammatory bowel disease, inflammation, acute and chronic gout and gouty arthritis, neurological disorders (see, e.g., Pandey et al., J. Hum. Genet. 2004, 49:596), metabolic syndrome, immunodeficiency disorders such as AIDS and HIV (see, e.g., Boehm et al., Viruses 2013, 5:1571), genetic disorders (see, e.g., Kosho et al., Am. J. Med. Genet. 2014, 166(3):262; Tang et al., Am. J. Med. Genet. 2015, 173(1):195), destructive bone disorders, osteoarthritis (see, e.g., Tian, J. Orthop. Surg. Res. 2018, 13:49), proliferative disorders (see, e.g., Cruickshank et al., PLoS One 2015, 10(11):e0142806), Waldenstrom's Macroglobulinemia. infectious diseases, conditions associated with cell death, pathologic immune conditions involving T cell activation, and CNS disorders (see, e.g., Koga et al., Human Mol. Gen. 2009, 18(13):2483) in a patient. In one embodiment, a human patient is treated with a compound of the current invention and a pharmaceutically acceptable carrier, adjuvant, or vehicle, wherein said compound is present in an amount to measurably modulate and/or inhibit one or more SMARCA2, SMARCA4, or PB1, or a mutant thereof


Compounds of the current invention are useful in the treatment of a proliferative disease selected from a benign or malignant tumor, solid tumor, carcinoma of the brain, kidney, liver, adrenal gland, bladder, breast, stomach, gastric tumors, ovaries, colon, rectum, prostate, pancreas, lung, vagina, cervix, testis, genitourinary tract, esophagus, larynx, skin, bone or thyroid, sarcoma, glioblastomas, neuroblastomas, multiple myeloma, gastrointestinal cancer, especially colon carcinoma or colorectal adenoma, a tumor of the neck and head, an epidermal hyperproliferation, psoriasis, prostate hyperplasia, a neoplasia, a neoplasia of epithelial character, adenoma, adenocarcinoma, keratoacanthoma, epidermoid carcinoma, large cell carcinoma, non-small-cell lung carcinoma, lymphomas, Hodgkins and Non-Hodgkins, a mammary carcinoma, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, an IL-1 driven disorder, an MyD88 driven disorder, Smoldering of indolent multiple myeloma, or hematological malignancies (including leukemia, diffuse large B-cell lymphoma (DLBCL), ABC DLBCL, chronic lymphocytic leukemia (CLL), chronic lymphocytic lymphoma, primary effusion lymphoma, Burkitt lymphoma/leukemia, acute lymphocytic leukemia, B-cell prolymphocytic leukemia, lymphoplasmacytic lymphoma, Waldenstrom's macroglobulinemia (WM), splenic marginal zone lymphoma, multiple myeloma, plasmacytoma, intravascular large B-cell lymphoma).


In some embodiments, the cancer is lung cancer, breast cancer, pancreatic cancer, colorectal cancer, and/or melanoma. In some embodiments, the cancer is lung cancer. In some embodiments, the lung cancer is NSCLC. In some embodiments, the cancer is breast cancer. In some embodiments, the cancer is melanoma.


SMARCA2 has recently been reported as a synthetic lethal target in SMARCA4-deficient cancers (e.g., cancers comprising SMARCA4 loss of function mutations and/or cancers having reduced or absent expression, e.g., due to epigenetic alterations). SMARCA2 depletion has been shown to selectively inhibit the growth of SMARCA4-mutant cancer cells (Hoffman et al., PNAS 2014, 111(8):3128; Oike et al., Cancer Res. 2013, 73(17):5508). In some embodiments, the cancer is a SMARCA4-deficient cancer (e.g., a cancer harboring a loss of function mutation and/or having reduced or absent SMARCA4 expression).


It has also been shown that certain cancers are dependent on SMARCA4 for disease progression and are vulnerable to SMARCA4 inhibition, including certain acute leukemias and small cell lung cancers (Hohmann et al., Trends in Genetics, 2014, 30(8):356). In some embodiments, the cancer is leukemia (e.g., acute leukemia, e.g., acute myeloid leukemia), breast cancer, small cell lung cancer, or malignant rhabdoid tumor (MRT) (e.g., a SNF5-deficient malignant rhabdoid tumor).


Compounds according to the invention are useful in the treatment of inflammatory or obstructive airways diseases, resulting, for example, in reduction of tissue damage, airways inflammation, bronchial hyperreactivity, remodeling or disease progression. Inflammatory or obstructive airways diseases to which the present invention is applicable include asthma of whatever type or genesis including both intrinsic (non-allergic) asthma and extrinsic (allergic) asthma, mild asthma, moderate asthma, severe asthma, bronchitic asthma, exercise-induced asthma, occupational asthma and asthma induced following bacterial infection. Treatment of asthma is also to be understood as embracing treatment of subjects, e.g. of less than 4 or 5 years of age, exhibiting wheezing symptoms and diagnosed or diagnosable as “wheezy infants”, an established patient category of major medical concern and now often identified as incipient or early-phase asthmatics.


Compounds according to the invention are useful in the treatment of heteroimmune diseases. Examples of such heteroimmune diseases include, but are not limited to, graft versus host disease, transplantation, transfusion, anaphylaxis, allergies (e.g., allergies to plant pollens, latex, drugs, foods, insect poisons, animal hair, animal dander, dust mites, or cockroach calyx), type I hypersensitivity, allergic conjunctivitis, allergic rhinitis, and atopic dermatitis.


Prophylactic efficacy in the treatment of asthma will be evidenced by reduced frequency or severity of symptomatic attack, e.g. of acute asthmatic or bronchoconstrictor attack, improvement in lung function or improved airways hyperreactivity. It may further be evidenced by reduced requirement for other, symptomatic therapy, such as therapy for or intended to restrict or abort symptomatic attack when it occurs, for example antiinflammatory or bronchodilatory. Prophylactic benefit in asthma may in particular be apparent in subjects prone to “morning dipping”. “Morning dipping” is a recognized asthmatic syndrome, common to a substantial percentage of asthmatics and characterized by asthma attack, e.g. between the hours of about 4 to 6 am, i.e. at a time normally substantially distant form any previously administered symptomatic asthma therapy.


Compounds of the current invention can be used for other inflammatory or obstructive airways diseases and conditions to which the present invention is applicable and include acute lung injury (ALI), adult/acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary, airways or lung disease (COPD, COAD or COLD), including chronic bronchitis or dyspnea associated therewith, emphysema, as well as exacerbation of airways hyperreactivity consequent to other drug therapy, in particular other inhaled drug therapy. The invention is also applicable to the treatment of bronchitis of whatever type or genesis including, but not limited to, acute, arachidic, catarrhal, croupus, chronic or phthinoid bronchitis. Further inflammatory or obstructive airways diseases to which the present invention is applicable include pneumoconiosis (an inflammatory, commonly occupational, disease of the lungs, frequently accompanied by airways obstruction, whether chronic or acute, and occasioned by repeated inhalation of dusts) of whatever type or genesis, including, for example, aluminosis, anthracosis, asbestosis, chalicosis, ptilosis, siderosis, silicosis, tabacosis and byssinosis.


With regard to their anti-inflammatory activity, in particular in relation to inhibition of eosinophil activation, compounds of the invention are also useful in the treatment of eosinophil related disorders, e.g. eosinophilia, in particular eosinophil related disorders of the airways (e.g. involving morbid eosinophilic infiltration of pulmonary tissues) including hypereosinophilia as it effects the airways and/or lungs as well as, for example, eosinophil-related disorders of the airways consequential or concomitant to Loffler's syndrome, eosinophilic pneumonia, parasitic (in particular metazoan) infestation (including tropical eosinophilia), bronchopulmonary aspergillosis, polyarteritis nodosa (including Churg-Strauss syndrome), eosinophilic granuloma and eosinophil-related disorders affecting the airways occasioned by drug-reaction.


Compounds of the invention are also useful in the treatment of inflammatory or allergic conditions of the skin, for example psoriasis, contact dermatitis, atopic dermatitis, alopecia areata, erythema multiforma, dermatitis herpetiformis, scleroderma, vitiligo, hypersensitivity angiitis, urticaria, bullous pemphigoid, lupus erythematosus, systemic lupus erythematosus, pemphigus vulgaris, pemphigus foliaceus, paraneoplastic pemphigus, epidermolysis bullosa acquisita, acne vulgaris, and other inflammatory or allergic conditions of the skin.


Compounds of the invention may also be used for the treatment of other diseases or conditions, such as diseases or conditions having an inflammatory component, for example, treatment of diseases and conditions of the eye such as ocular allergy, conjunctivitis, keratoconjunctivitis sicca, and vernal conjunctivitis, diseases affecting the nose including allergic rhinitis, and inflammatory disease in which autoimmune reactions are implicated or having an autoimmune component or etiology, including autoimmune hematological disorders (e.g. hemolytic anemia, aplastic anemia, pure red cell anemia and idiopathic thrombocytopenia), systemic lupus erythematosus, rheumatoid arthritis, polychondritis, scleroderma, Wegener granulamatosis, dermatomyositis, chronic active hepatitis, myasthenia gravis, Steven-Johnson syndrome, idiopathic sprue, autoimmune inflammatory bowel disease (e.g. ulcerative colitis and Crohn's disease), irritable bowel syndrome, celiac disease, periodontitis, hyaline membrane disease, kidney disease, glomerular disease, alcoholic liver disease, multiple sclerosis, endocrine opthalmopathy, Grave's disease, sarcoidosis, alveolitis, chronic hypersensitivity pneumonitis, multiple sclerosis, primary biliary cirrhosis, uveitis (anterior and posterior), Sjogren's syndrome, keratoconjunctivitis sicca and vernal keratoconjunctivitis, interstitial lung fibrosis, psoriatic arthritis, systemic juvenile idiopathic arthritis, cryopyrin-associated periodic syndrome, nephritis, vasculitis, diverticulitis, interstitial cystitis, glomerulonephritis (with and without nephrotic syndrome, e.g. including idiopathic nephrotic syndrome or minal change nephropathy), chronic granulomatous disease, endometriosis, leptospiriosis renal disease, glaucoma, retinal disease, ageing, headache, pain, complex regional pain syndrome, cardiac hypertrophy, musclewasting, catabolic disorders, obesity, fetal growth retardation, hyperchlolesterolemia, heart disease, chronic heart failure, mesothelioma, anhidrotic ecodermal dysplasia, Behcet's disease, incontinentia pigmenti, Paget's disease, pancreatitis, hereditary periodic fever syndrome, asthma (allergic and non-allergic, mild, moderate, severe, bronchitic, and exercise-induced), acute lung injury, acute respiratory distress syndrome, eosinophilia, hypersensitivities, anaphylaxis, nasal sinusitis, ocular allergy, silica induced diseases, COPD (reduction of damage, airways inflammation, bronchial hyperreactivity, remodeling or disease progression), pulmonary disease, cystic fibrosis, acid-induced lung injury, pulmonary hypertension, polyneuropathy, cataracts, muscle inflammation in conjunction with systemic sclerosis, inclusion body myositis, myasthenia gravis, thyroiditis, Addison's disease, lichen planus, Type 1 diabetes, or Type 2 diabetes, appendicitis, atopic dermatitis, asthma, allergy, blepharitis, bronchiolitis, bronchitis, bursitis, cervicitis, cholangitis, cholecystitis, chronic graft rejection, colitis, conjunctivitis, Crohn's disease, cystitis, dacryoadenitis, dermatitis, dermatomyositis, encephalitis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, Henoch-Schonlein purpura, hepatitis, hidradenitis suppurativa, immunoglobulin A nephropathy, interstitial lung disease, laryngitis, mastitis, meningitis, myelitis myocarditis, myositis, nephritis, oophoritis, orchitis, osteitis, otitis, pancreatitis, parotitis, pericarditis, peritonitis, pharyngitis, pleuritis, phlebitis, pneumonitis, pneumonia, polymyositis, proctitis, prostatitis, pyelonephritis, rhinitis, salpingitis, sinusitis, stomatitis, synovitis, tendonitis, tonsillitis, ulcerative colitis, uveitis, vaginitis, vasculitis, or vulvitis.


In some embodiments the inflammatory disease which can be treated according to the methods of this invention is a disease of the skin. In some embodiments, the inflammatory disease of the skin is selected from contact dermatitis, atopic dermatitis, alopecia areata, erythema multiforma, dermatitis herpetiformis, scleroderma, vitiligo, hypersensitivity angiitis, urticaria, bullous pemphigoid, pemphigus vulgaris, pemphigus foliaceus, paraneoplastic pemphigus, epidermolysis bullosa acquisita, and other inflammatory or allergic conditions of the skin.


In some embodiments the inflammatory disease which can be treated according to the methods of this invention is selected from acute and chronic gout, chronic gouty arthritis, psoriasis, psoriatic arthritis, rheumatoid arthritis, Juvenile rheumatoid arthritis, Systemic juvenile idiopathic arthritis (SJIA), Cryopyrin Associated Periodic Syndrome (CAPS), and osteoarthritis.


In some embodiments the inflammatory disease which can be treated according to the methods of this invention is a TH17 mediated disease. In some embodiments the TH17 mediated disease is selected from Systemic lupus erythematosus, Multiple sclerosis, and inflammatory bowel disease (including Crohn's disease or ulcerative colitis).


In some embodiments the inflammatory disease which can be treated according to the methods of this invention is selected from Sjogren's syndrome, allergic disorders, osteoarthritis, conditions of the eye such as ocular allergy, conjunctivitis, keratoconjunctivitis sicca and vernal conjunctivitis, and diseases affecting the nose such as allergic rhinitis.


Cardiovascular diseases which can be treated according to the methods of this invention include, but are not limited to, restenosis, cardiomegaly, atherosclerosis, myocardial infarction, ischemic stroke, congestive heart failure, angina pectoris, reocclusion after angioplasty, restenosis after angioplasty, reocclusion after aortocoronary bypass, restenosis after aortocoronary bypass, stroke, transitory ischemia, a peripheral arterial occlusive disorder, pulmonary embolism, and deep venous thrombosis.


In some embodiments, the neurodegenerative disease which can be treated according to the methods of this invention include, but are not limited to, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, cerebral ischemia, and neurodegenerative disease caused by traumatic injury, glutamate neurotoxicity, hypoxia, epilepsy, treatment of diabetes, metabolic syndrome, obesity, organ transplantation and graft versus host disease.


In some embodiments the invention provides a method of treating, preventing or lessening the severity of Alzheimer's disease comprising administering to a patient in need thereof a provided compound or a pharmaceutically acceptable salt or composition thereof.


In some embodiments the invention provides a method of treating a disease or condition commonly occurring in connection with transplantation. In some embodiments, the disease or condition commonly occurring in connection with transplantation is selected from organ transplantation, organ transplant rejection, and graft versus host disease.


In some embodiments the invention provides a method of treating a metabolic disease. In some embodiments the metabolic disease is selected from Type 1 diabetes, Type 2 diabetes, metabolic syndrome, and obesity.


In some embodiments the invention provides a method of treating a viral disease. In some embodiments, the viral infection is HIV infection.


Furthermore, the invention provides the use of a compound according to the definitions herein, or a pharmaceutically acceptable salt, or a hydrate or solvate thereof for the preparation of a medicament for the treatment of a proliferative disease, an inflammatory disease, an obstructive respiratory disease, a cardiovascular disease, a metabolic disease, a neurological disease, a neurodegenerative disease, a viral disease, or a disorder commonly occurring in connection with transplantation.


Combination Therapies


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


In certain embodiments, a provided combination, or composition thereof, is administered in combination with another therapeutic agent.


In some embodiments, the present invention provides a method of treating a disclosed disease or condition comprising administering to a patient in need thereof an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof and co-administering simultaneously or sequentially an effective amount of one or more additional therapeutic agents, such as those described herein. In some embodiments, the method includes co-administering one additional therapeutic agent. In some embodiments, the method includes co-administering two additional therapeutic agents. In some embodiments, the combination of the disclosed compound and the additional therapeutic agent or agents acts synergistically.


Examples of agents the combinations of this invention may also be combined with include, without limitation: treatments for Alzheimer's Disease such as Aricept® and Excelon®; treatments for HIV such as ritonavir; treatments for Parkinson's Disease such as L-DOPA/carbidopa, entacapone, ropinrole, pramipexole, bromocriptine, pergolide, trihexephendyl, and amantadine; agents for treating Multiple Sclerosis (MS) such as beta interferon (e.g., Avonex® and Rebif®), Copaxone*, and mitoxantrone; treatments for asthma such as albuterol and Singulair®; agents for treating schizophrenia such as zyprexa, risperdal, seroquel, and haloperidol; anti-inflammatory agents such as corticosteroids, TNF blockers, IL-1 RA, azathioprine, cyclophosphamide, and sulfasalazine; immunomodulatory and immunosuppressive agents such as cyclosporin, tacrolimus, rapamycin, mycophenolate mofetil, interferons, corticosteroids, cyclophophamide, azathioprine, and sulfasalazine; neurotrophic factors such as acetylcholinesterase inhibitors, MAO inhibitors, interferons, anti-convulsants, ion channel blockers, riluzole, and anti-Parkinsonian agents; agents for treating cardiovascular disease such as beta-blockers, ACE inhibitors, diuretics, nitrates, calcium channel blockers, and statins; agents for treating liver disease such as corticosteroids, cholestyramine, interferons, and anti-viral agents; agents for treating blood disorders such as corticosteroids, anti-leukemic agents, and growth factors; agents that prolong or improve pharmacokinetics such as cytochrome P450 inhibitors (i.e., inhibitors of metabolic breakdown) and CYP3A4 inhibitors (e.g., ketokenozole and ritonavir), and agents for treating immunodeficiency disorders such as gamma globulin.


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


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


As used herein, the term “combination,” “combined,” and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this invention. For example, a combination of the present invention may be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form.


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


One or more other therapeutic agent may be administered separately from a compound or composition of the invention, as part of a multiple dosage regimen. Alternatively, one or more other therapeutic agents may be part of a single dosage form, mixed together with a compound of this invention in a single composition. If administered as a multiple dosage regime, one or more other therapeutic agent and a compound or composition of the invention may be administered simultaneously, sequentially or within a period of time from one another, for example within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 18, 20, 21, 22, 23, or 24 hours from one another. In some embodiments, one or more other therapeutic agent and a compound or composition of the invention are administered as a multiple dosage regimen within greater than 24 hours apart.


In one embodiment, the present invention provides a composition comprising a provided compound and one or more additional therapeutic agents. The therapeutic agent may be administered together with a provided compound, or may be administered prior to or following administration of a provided compound. Suitable therapeutic agents are described in further detail below. In certain embodiments, a provided compound may be administered up to 5 minutes, 10 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5, hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, or 18 hours before the therapeutic agent. In other embodiments, a provided compound may be administered up to 5 minutes, 10 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5, hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, or 18 hours following the therapeutic agent.


In another embodiment, the present invention provides a method of treating an inflammatory disease, disorder or condition by administering to a patient in need thereof a provided compound and one or more additional therapeutic agents. Such additional therapeutic agents may be small molecules or recombinant biologic agents and include, for example, acetaminophen, non-steroidal anti-inflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, etodolac (Lodine®) and celecoxib, colchicine (Colcrys®), corticosteroids such as prednisone, prednisolone, methylprednisolone, hydrocortisone, and the like, probenecid, allopurinol, febuxostat (Uloric®), sulfasalazine (Azulfidine®), antimalarials such as hydroxychloroquine (Plaquenil®) and chloroquine (Aralen®), methotrexate (Rheumatrex®), gold salts such as gold thioglucose (Solganal®), gold thiomalate (Myochrysine®) and auranofin (Ridaura®), D-penicillamine (Depen® or Cuprimine®), azathioprine (Imuran®), cyclophosphamide (Cytoxan®), chlorambucil (Leukeran®), cyclosporine (Sandimmune®), leflunomide (Arava®) and “anti-TNF” agents such as etanercept (Enbrel®), infliximab (Remicade®), golimumab (Simponi®), certolizumab pegol (Cimzia®) and adalimumab (Humira®), “anti-IL-1” agents such as anakinra (Kineret®) and rilonacept (Arcalyst®), canakinumab (Ilaris®), anti-Jak inhibitors such as tofacitinib, antibodies such as rituximab (Rituxan®), “anti-T-cell” agents such as abatacept (Orencia®), “anti-IL-6” agents such as tocilizumab (Actemra®), diclofenac, cortisone, hyaluronic acid (Synvisc® or Hyalgan®), monoclonal antibodies such as tanezumab, anticoagulants such as heparin (Calcinparine® or Liquaemin®) and warfarin (Coumadin®), antidiarrheals such as diphenoxylate (Lomotil®) and loperamide (Imodium®), bile acid binding agents such as cholestyramine, alosetron (Lotronex®), lubiprostone (Amitiza®), laxatives such as Milk of Magnesia, polyethylene glycol (MiraLax®), Dulcolax®, Correctol® and Senokot®, anticholinergics or antispasmodics such as dicyclomine (Bentyl®), Singulair®, beta-2 agonists such as albuterol (Ventolin® HFA, Proventil® HFA), levalbuterol (Xopenex®), metaproterenol (Alupent®), pirbuterol acetate (Maxair®), terbutaline sulfate (Brethaire®), salmeterol xinafoate (Serevent®) and formoterol (Foradil®), anticholinergic agents such as ipratropium bromide (Atrovent®) and tiotropium (Spiriva®), inhaled corticosteroids such as beclomethasone dipropionate (Beclovent®, Qvar®, and Vanceril®), triamcinolone acetonide (Azmacort®), mometasone (Asthmanex®), budesonide (Pulmocort®), and flunisolide (Aerobid®), Afviar®, Symbicort®, Dulera®, cromolyn sodium (Intal®), methylxanthines such as theophylline (Theo-Dur®, Theolair®, Slo-bid®, Uniphyl®, Theo-24®) and aminophylline, IgE antibodies such as omalizumab (Xolair®), nucleoside reverse transcriptase inhibitors such as zidovudine (Retrovir®), abacavir (Ziagen®), abacavir/lamivudine (Epzicom®), abacavir/lamivudine/zidovudine (Trizivir®), didanosine (Videx®), emtricitabine (Emtriva®), lamivudine (Epivir®), lamivudine/zidovudine (Combivir®), stavudine (Zerit®), and zalcitabine (Hivid®), non-nucleoside reverse transcriptase inhibitors such as delavirdine (Rescriptor®), efavirenz (Sustiva®), nevairapine (Viramune®) and etravirine (Intelence®), nucleotide reverse transcriptase inhibitors such as tenofovir (Viread®), protease inhibitors such as amprenavir (Agenerase®), atazanavir (Reyataz®), darunavir (Prezista®), fosamprenavir (Lexiva®), indinavir (Crixivan®), lopinavir and ritonavir (Kaletra®), nelfinavir (Viracept®), ritonavir (Norvir®), saquinavir (Fortovase® or Invirase®), and tipranavir (Aptivus®), entry inhibitors such as enfuvirtide (Fuzeon®) and maraviroc (Selzentry®), integrase inhibitors such as raltegravir (Isentress®), doxorubicin (Hydrodaunorubicin®), vincristine (Oncovin®), bortezomib (Velcade®), and dexamethasone (Decadron®) in combination with lenalidomide (Revlimid®), or any combination(s) thereof.


In another embodiment, the present invention provides a method of treating gout comprising administering to a patient in need thereof a provided compound and one or more additional therapeutic agents selected from non-steroidal anti-inflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, etodolac (Lodine®) and celecoxib, colchicine (Colcrys®), corticosteroids such as prednisone, prednisolone, methylprednisolone, hydrocortisone, and the like, probenecid, allopurinol and febuxostat (Uloric®).


In another embodiment, the present invention provides a method of treating rheumatoid arthritis comprising administering to a patient in need thereof a provided compound and one or more additional therapeutic agents selected from non-steroidal anti-inflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, etodolac (Lodine®) and celecoxib, corticosteroids such as prednisone, prednisolone, methylprednisolone, hydrocortisone, and the like, sulfasalazine (Azulfidine®), antimalarials such as hydroxychloroquine (Plaquenil®) and chloroquine (Aralen®), methotrexate (Rheumatrex®), gold salts such as gold thioglucose (Solganal®), gold thiomalate (Myochrysine®) and auranofin (Ridaura®), D-penicillamine (Depen® or Cuprimine®), azathioprine (Imuran®), cyclophosphamide (Cytoxan®), chlorambucil (Leukeran®), cyclosporine (Sandimmune®), leflunomide (Arava®) and “anti-TNF” agents such as etanercept (Enbrel®), infliximab (Remicade®), golimumab (Simponi®), certolizumab pegol (Cimzia®) and adalimumab (Humira®), “anti-IL-i” agents such as anakinra (Kineret®) and rilonacept (Arcalyst®), antibodies such as rituximab (Rituxan®), “anti-T-cell” agents such as abatacept (Orencia®) and “anti-IL-6” agents such as tocilizumab (Actemra®).


In some embodiments, the present invention provides a method of treating osteoarthritis comprising administering to a patient in need thereof a provided compound and one or more additional therapeutic agents selected from acetaminophen, non-steroidal anti-inflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, etodolac (Lodine®) and celecoxib, diclofenac, cortisone, hyaluronic acid (Synvisc® or Hyalgan®) and monoclonal antibodies such as tanezumab.


In some embodiments, the present invention provides a method of treating lupus comprising administering to a patient in need thereof a provided compound and one or more additional therapeutic agents selected from acetaminophen, non-steroidal anti-inflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, etodolac (Lodine®) and celecoxib, corticosteroids such as prednisone, prednisolone, methylprednisolone, hydrocortisone, and the like, antimalarials such as hydroxychloroquine (Plaquenil®) and chloroquine (Aralen®), cyclophosphamide (Cytoxan®), methotrexate (Rheumatrex®), azathioprine (Imuran®) and anticoagulants such as heparin (Calcinparine® or Liquaemin®) and warfarin (Coumadin®).


In some embodiments, the present invention provides a method of treating inflammatory bowel disease comprising administering to a patient in need thereof a provided compound and one or more additional therapeutic agents selected from mesalamine (Asacol®) sulfasalazine (Azulfidine®), antidiarrheals such as diphenoxylate (Lomotil®) and loperamide (Imodium®), bile acid binding agents such as cholestyramine, alosetron (Lotronex®), lubiprostone (Amitiza®), laxatives such as Milk of Magnesia, polyethylene glycol (MiraLax®), Dulcolax®, Correctol® and Senokot® and anticholinergics or antispasmodics such as dicyclomine (Bentyl®), anti-TNF therapies, steroids, and antibiotics such as Flagyl or ciprofloxacin.


In some embodiments, the present invention provides a method of treating asthma comprising administering to a patient in need thereof a provided compound and one or more additional therapeutic agents selected from Singulair®, beta-2 agonists such as albuterol (Ventolin® HFA, Proventil® HFA), levalbuterol (Xopenex®), metaproterenol (Alupent®), pirbuterol acetate (Maxair®), terbutaline sulfate (Brethaire®), salmeterol xinafoate (Serevent®) and formoterol (Foradil®), anticholinergic agents such as ipratropium bromide (Atrovent®) and tiotropium (Spiriva®), inhaled corticosteroids such as prednisone, prednisolone, beclomethasone dipropionate (Beclovent®, Qvar®, and Vanceril®), triamcinolone acetonide (Azmacort®), mometasone (Asthmanex®), budesonide (Pulmocort®), flunisolide (Aerobid®), Afviar®, Symbicort®, and Dulera®, cromolyn sodium (Intal®), methylxanthines such as theophylline (Theo-Dur®, Theolair®, Slo-bid®, Uniphyl®, Theo-24®) and aminophylline, and IgE antibodies such as omalizumab (Xolair®).


In some embodiments, the present invention provides a method of treating COPD comprising administering to a patient in need thereof a provided compound and one or more additional therapeutic agents selected from beta-2 agonists such as albuterol (Ventolin® HFA, Proventil® HFA), levalbuterol (Xopenex®), metaproterenol (Alupent®), pirbuterol acetate (Maxair®), terbutaline sulfate (Brethaire®), salmeterol xinafoate (Serevent®) and formoterol (Foradil®), anticholinergic agents such as ipratropium bromide (Atrovent®) and tiotropium (Spiriva®), methylxanthines such as theophylline (Theo-Dur®, Theolair®, Slo-bid®, Uniphyl®, Theo-24®) and aminophylline, inhaled corticosteroids such as prednisone, prednisolone, beclomethasone dipropionate (Beclovent®, Qvar®, and Vanceril®), triamcinolone acetonide (Azmacort®), mometasone (Asthmanex®), budesonide (Pulmocort®), flunisolide (Aerobid®), Afviar®, Symbicort®, and Dulera®,


In some embodiments, the present invention provides a method of treating HIV comprising administering to a patient in need thereof a provided compound and one or more additional therapeutic agents selected from nucleoside reverse transcriptase inhibitors such as zidovudine (Retrovir®), abacavir (Ziagen®), abacavir/lamivudine (Epzicom®), abacavir/lamivudine/zidovudine (Trizivir®), didanosine (Videx®), emtricitabine (Emtriva®), lamivudine (Epivir®), lamivudine/zidovudine (Combivir®), stavudine (Zerit®), and zalcitabine (Hivid®), non-nucleoside reverse transcriptase inhibitors such as delavirdine (Rescriptor®), efavirenz (Sustiva®), nevairapine (Viramune®) and etravirine (Intelence®), nucleotide reverse transcriptase inhibitors such as tenofovir (Viread®), protease inhibitors such as amprenavir (Agenerase®), atazanavir (Reyataz®), darunavir (Prezista®), fosamprenavir (Lexiva®), indinavir (Crixivan®), lopinavir and ritonavir (Kaletra®), nelfinavir (Viracept®), ritonavir (Norvir®), saquinavir (Fortovase® or Invirase®), and tipranavir (Aptivus®), entry inhibitors such as enfuvirtide (Fuzeon®) and maraviroc (Selzentry®), integrase inhibitors such as raltegravir (Isentress®), and combinations thereof.


In another embodiment, the present invention provides a method of treating a hematological malignancy comprising administering to a patient in need thereof a provided compound and one or more additional therapeutic agents selected from rituximab (Rituxan®), cyclophosphamide (Cytoxan®), doxorubicin (Hydrodaunorubicin®), vincristine (Oncovin®), prednisone, a hedgehog signaling inhibitor, a BTK inhibitor, a JAK/pan-JAK inhibitor, a TYK2 inhibitor, a PI3K inhibitor, a SYK inhibitor, and combinations thereof.


In another embodiment, the present invention provides a method of treating a solid tumor comprising administering to a patient in need thereof a provided compound and one or more additional therapeutic agents selected from rituximab (Rituxan®), cyclophosphamide (Cytoxan®), doxorubicin (Hydrodaunorubicin®), vincristine (Oncovin®), prednisone, a hedgehog signaling inhibitor, a BTK inhibitor, a JAK/pan-JAK inhibitor, a TYK2 inhibitor, a PI3K inhibitor, a SYK inhibitor, and combinations thereof.


In another embodiment, the present invention provides a method of treating a hematological malignancy comprising administering to a patient in need thereof a provided compound and a Hedgehog (Hh) signaling pathway inhibitor. In some embodiments, the hematological malignancy is DLBCL (Ramirez et al “Defining causative factors contributing in the activation of hedgehog signaling in diffuse large B-cell lymphoma” Leuk. Res. (2012), published online July 17, and incorporated herein by reference in its entirety).


In another embodiment, the present invention provides a method of treating diffuse large B-cell lymphoma (DLBCL) comprising administering to a patient in need thereof a provided compound and one or more additional therapeutic agents selected from rituximab (Rituxan®), cyclophosphamide (Cytoxan®), doxorubicin (Hydrodaunorubicin®), vincristine (Oncovin®), prednisone, a hedgehog signaling inhibitor, and combinations thereof.


In another embodiment, the present invention provides a method of treating multiple myeloma comprising administering to a patient in need thereof a provided compound and one or more additional therapeutic agents selected from bortezomib (Velcade®), and dexamethasone (Decadron®), a hedgehog signaling inhibitor, a BTK inhibitor, a JAK/pan-JAK inhibitor, a TYK2 inhibitor, a PI3K inhibitor, a SYK inhibitor in combination with lenalidomide (Revlimid®).


In another embodiment, the present invention provides a method of treating Waldenstrom's macroglobulinemia comprising administering to a patient in need thereof a provided compound and one or more additional therapeutic agents selected from chlorambucil (Leukeran®), cyclophosphamide (Cytoxan®, Neosar®), fludarabine (Fludara®), cladribine (Leustatin®), rituximab (Rituxan®), a hedgehog signaling inhibitor, a BTK inhibitor, a JAK/pan-JAK inhibitor, a TYK2 inhibitor, a P13K inhibitor, and a SYK inhibitor.


In some embodiments, one or more other therapeutic agent is an antagonist of the hedgehog pathway. Approved hedgehog pathway inhibitors which may be used in the present invention include sonidegib (Odomzo®, Sun Pharmaceuticals); and vismodegib (Erivedge®, Genentech), both for treatment of basal cell carcinoma.


In some embodiments, one or more other therapeutic agent is a Poly ADP ribose polymerase (PARP) inhibitor. In some embodiments, a PARP inhibitor is selected from olaparib (Lynparza®, AstraZeneca); rucaparib (Rubraca®, Clovis Oncology); niraparib (Zejula®, Tesaro); talazoparib (MDV3800/BMN 673/LT00673, Medivation/Pfizer/Biomarin); veliparib (ABT-888, AbbVie); and BGB-290 (BeiGene, Inc.).


In some embodiments, one or more other therapeutic agent is a histone deacetylase (HDAC) inhibitor. In some embodiments, an HDAC inhibitor is selected from vorinostat (Zolinza®, Merck); romidepsin (Istodax®, Celgene); panobinostat (Farydak®, Novartis); belinostat (Beleodaq®, Spectrum Pharmaceuticals); entinostat (SNDX-275, Syndax Pharmaceuticals) (NCT00866333); and chidamide (Epidaza®, HBI-8000, Chipscreen Biosciences, China).


In some embodiments, one or more other therapeutic agent is a CDK inhibitor, such as a CDK4/CDK6 inhibitor. In some embodiments, a CDK 4/6 inhibitor is selected from palbociclib (Ibrance®, Pfizer); ribociclib (Kisqali®, Novartis); abemaciclib (Ly2835219, Eli Lilly); and trilaciclib (G1T28, G1 Therapeutics).


In some embodiments, one or more other therapeutic agent is a folic acid inhibitor. Approved folic acid inhibitors useful in the present invention include pemetrexed (Alimta®, Eli Lilly).


In some embodiments, one or more other therapeutic agent is a CC chemokine receptor 4 (CCR4) inhibitor. CCR4 inhibitors being studied that may be useful in the present invention include mogamulizumab (Poteligeo®, Kyowa Hakko Kirin, Japan).


In some embodiments, one or more other therapeutic agent is an isocitrate dehydrogenase (IDH) inhibitor. IDH inhibitors being studied which may be used in the present invention include AG120 (Celgene; NCT02677922); AG221 (Celgene, NCT02677922; NCT02577406); BAY1436032 (Bayer, NCT02746081); IDH305 (Novartis, NCT02987010).


In some embodiments, one or more other therapeutic agent is an arginase inhibitor. Arginase inhibitors being studied which may be used in the present invention include AEB1102 (pegylated recombinant arginase, Aeglea Biotherapeutics), which is being studied in Phase 1 clinical trials for acute myeloid leukemia and myelodysplastic syndrome (NCT02732184) and solid tumors (NCT02561234); and CB-1158 (Calithera Biosciences).


In some embodiments, one or more other therapeutic agent is a glutaminase inhibitor. Glutaminase inhibitors being studied which may be used in the present invention include CB-839 (Calithera Biosciences).


In some embodiments, one or more other therapeutic agent is an antibody that binds to tumor antigens, that is, proteins expressed on the cell surface of tumor cells. Approved antibodies that bind to tumor antigens which may be used in the present invention include rituximab (Rituxan®, Genentech/BiogenIdec); ofatumumab (anti-CD20, Arzerra®, GlaxoSmithKline); obinutuzumab (anti-CD20, Gazyva®, Genentech), ibritumomab (anti-CD20 and Yttrium-90, Zevalin®, Spectrum Pharmaceuticals); daratumumab (anti-CD38, Darzalex®, Janssen Biotech), dinutuximab (anti-glycolipid GD2, Unituxin®, United Therapeutics); trastuzumab (anti-HER2, Herceptin®, Genentech); ado-trastuzumab emtansine (anti-HER2, fused to emtansine, Kadcyla®, Genentech); and pertuzumab (anti-HER2, Perjeta®, Genentech); and brentuximab vedotin (anti-CD30-drug conjugate, Adcetris®, Seattle Genetics).


In some embodiments, one or more other therapeutic agent is a topoisomerase inhibitor. Approved topoisomerase inhibitors useful in the present invention include irinotecan (Onivyde®, Merrimack Pharmaceuticals); topotecan (Hycamtin®, GlaxoSmithKline). Topoisomerase inhibitors being studied which may be used in the present invention include pixantrone (Pixuvri®, CTI Biopharma).


In some embodiments, one or more other therapeutic agent is an inhibitor of anti-apoptotic proteins, such as BCL-2. Approved anti-apoptotics which may be used in the present invention include venetoclax (Venclexta®, AbbVie/Genentech); and blinatumomab (Blincyto®, Amgen). Other therapeutic agents targeting apoptotic proteins which have undergone clinical testing and may be used in the present invention include navitoclax (ABT-263, Abbott), a BCL-2 inhibitor (NCT02079740).


In some embodiments, one or more other therapeutic agent is an androgen receptor inhibitor. Approved androgen receptor inhibitors useful in the present invention include enzalutamide (Xtandi®, Astellas/Medivation); approved inhibitors of androgen synthesis include abiraterone (Zytiga®, Centocor/Ortho); approved antagonist of gonadotropin-releasing hormone (GnRH) receptor (degaralix, Firmagon®, Ferring Pharmaceuticals).


In some embodiments, one or more other therapeutic agent is a selective estrogen receptor modulator (SERM), which interferes with the synthesis or activity of estrogens. Approved SERMs useful in the present invention include raloxifene (Evista®, Eli Lilly).


In some embodiments, one or more other therapeutic agent is an inhibitor of bone resorption. An approved therapeutic which inhibits bone resorption is Denosumab (Xgeva®, Amgen), an antibody that binds to RANKL, prevents binding to its receptor RANK, found on the surface of osteoclasts, their precursors, and osteoclast-like giant cells, which mediates bone pathology in solid tumors with osseous metastases. Other approved therapeutics that inhibit bone resorption include bisphosphonates, such as zoledronic acid (Zometa®, Novartis).


In some embodiments, one or more other therapeutic agent is an inhibitor of interaction between the two primary p53 suppressor proteins, MDMX and MDM2. Inhibitors of p53 suppression proteins being studied which may be used in the present invention include ALRN-6924 (Aileron), a stapled peptide that equipotently binds to and disrupts the interaction of MDMX and MDM2 with p53. ALRN-6924 is currently being evaluated in clinical trials for the treatment of AML, advanced myelodysplastic syndrome (MDS) and peripheral T-cell lymphoma (PTCL) (NCT02909972; NCT02264613).


In some embodiments, one or more other therapeutic agent is an inhibitor of transforming growth factor-beta (TGF-beta or TGFβ). Inhibitors of TGF-beta proteins being studied which may be used in the present invention include NIS793 (Novartis), an anti-TGF-beta antibody being tested in the clinic for treatment of various cancers, including breast, lung, hepatocellular, colorectal, pancreatic, prostate and renal cancer (NCT 02947165). In some embodiments, the inhibitor of TGF-beta proteins is fresolimumab (GC1008; Sanofi-Genzyme), which is being studied for melanoma (NCT00923169); renal cell carcinoma (NCT00356460); and non-small cell lung cancer (NCT02581787). Additionally, in some embodiments, the additional therapeutic agent is a TGF-beta trap, such as described in Connolly et al. (2012) Int'l J. Biological Sciences 8:964-978. One therapeutic compound currently in clinical trials for treatment of solid tumors is M7824 (Merck KgaA—formerly MSB0011459X), which is a bispecific, anti-PD-L1/TGFβ trap compound (NCT02699515); and (NCT02517398). M7824 is comprised of a fully human IgG1 antibody against PD-L1 fused to the extracellular domain of human TGF-beta receptor II, which functions as a TGFβ “trap.”


In some embodiments, one or more other therapeutic agent is selected from glembatumumab vedotin-monomethyl auristatin E (MMAE) (Celldex), an anti-glycoprotein NMB (gpNMB) antibody (CR011) linked to the cytotoxic MMAE. gpNMB is a protein overexpressed by multiple tumor types associated with cancer cells' ability to metastasize.


In some embodiments, one or more other therapeutic agent is an antiproliferative compound. Such antiproliferative compounds include, but are not limited to aromatase inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase II inhibitors; microtubule active compounds; alkylating compounds; histone deacetylase inhibitors; compounds which induce cell differentiation processes; cyclooxygenase inhibitors; MMP inhibitors; mTOR inhibitors; antineoplastic antimetabolites; platin compounds; compounds targeting/decreasing a protein or lipid kinase activity and further anti-angiogenic compounds; compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase; gonadorelin agonists; anti-androgens; methionine aminopeptidase inhibitors; matrix metalloproteinase inhibitors; bisphosphonates; biological response modifiers; antiproliferative antibodies; heparanase inhibitors; inhibitors of Ras oncogenic isoforms; telomerase inhibitors; proteasome inhibitors; compounds used in the treatment of hematologic malignancies; compounds which target, decrease or inhibit the activity of Flt-3; Hsp90 inhibitors such as 17-AAG (17-allylaminogeldanamycin, NSC330507), 17-DMAG (17-dimethylaminoethylamino-17-demethoxy-geldanamycin, NSC707545), IPI-504, CNF1010, CNF2024, CNF1010 from Conforma Therapeutics; temozolomide (Temodal®); kinesin spindle protein inhibitors, such as SB715992 or SB743921 from GlaxoSmithKline, or pentamidine/chlorpromazine from CombinatoRx; MEK inhibitors such as ARRY142886 from Array BioPharma, AZd6244 from AstraZeneca, PD181461 from Pfizer and leucovorin.


In some embodiments, the present invention provides a method of treating Alzheimer's disease comprising administering to a patient in need thereof a provided compound and one or more additional therapeutic agents selected from donepezil (Aricept®), rivastigmine (Excelon®), galantamine (Razadyne®), tacrine (Cognex®), and memantine (Namenda®).


In some embodiments, one or more other therapeutic agent is a taxane compound, which causes disruption of microtubules, which are essential for cell division. In some embodiments, a taxane compound is selected from paclitaxel (Taxol®, Bristol-Myers Squibb), docetaxel (Taxotere®, Sanofi-Aventis; Docefrez®, Sun Pharmaceutical), albumin-bound paclitaxel (Abraxane®; Abraxis/Celgene), cabazitaxel (Jevtana®, Sanofi-Aventis), and SID530 (SK Chemicals, Co.) (NCT00931008).


In some embodiments, one or more other therapeutic agent is a nucleoside inhibitor, or a therapeutic agent that interferes with normal DNA synthesis, protein synthesis, cell replication, or will otherwise inhibit rapidly proliferating cells.


In some embodiments, a nucleoside inhibitor is selected from trabectedin (guanidine alkylating agent, Yondelis®, Janssen Oncology), mechlorethamine (alkylating agent, Valchlor®, Aktelion Pharmaceuticals); vincristine (Oncovin®, Eli Lilly; Vincasar®, Teva Pharmaceuticals; Marqibo®, Talon Therapeutics); temozolomide (prodrug to alkylating agent 5-(3-methyltriazen-1-yl)-imidazole-4-carboxamide (MTIC) Temodar®, Merck); cytarabine injection (ara-C, antimetabolic cytidine analog, Pfizer); lomustine (alkylating agent, CeeNU®, Bristol-Myers Squibb; Gleostine®, NextSource Biotechnology); azacitidine (pyrimidine nucleoside analog of cytidine, Vidaza®, Celgene); omacetaxine mepesuccinate (cephalotaxine ester) (protein synthesis inhibitor, Synribo®; Teva Pharmaceuticals); asparaginase Erwinia chrysanthemi (enzyme for depletion of asparagine, Elspar®, Lundbeck; Erwinaze®, EUSA Pharma); eribulin mesylate (microtubule inhibitor, tubulin-based antimitotic, Halaven®, Eisai); cabazitaxel (microtubule inhibitor, tubulin-based antimitotic, Jevtana®, Sanofi-Aventis); capacetrine (thymidylate synthase inhibitor, Xeloda®, Genentech); bendamustine (bifunctional mechlorethamine derivative, believed to form interstrand DNA cross-links, Treanda®, Cephalon/Teva); ixabepilone (semi-ynthetic analog of epothilone B, microtubule inhibitor, tubulin-based antimitotic, Ixempra®, Bristol-Myers Squibb); nelarabine (prodrug of deoxyguanosine analog, nucleoside metabolic inhibitor, Arranon®, Novartis); clorafabine (prodrug of ribonucleotide reductase inhibitor, competitive inhibitor of deoxycytidine, Clolar®, Sanofi-Aventis); and trifluridine and tipiracil (thymidine-based nucleoside analog and thymidine phosphorylase inhibitor, Lonsurf®, Taiho Oncology).


In some embodiments, one or more other therapeutic agent is a kinase inhibitor or VEGF-R antagonist. Approved VEGF inhibitors and kinase inhibitors useful in the present invention include: bevacizumab (Avastin®, Genentech/Roche) an anti-VEGF monoclonal antibody; ramucirumab (Cyramza®, Eli Lilly), an anti-VEGFR-2 antibody and ziv-aflibercept, also known as VEGF Trap (Zaltrap®; Regeneron/Sanofi). VEGFR inhibitors, such as regorafenib (Stivarga®, Bayer); vandetanib (Caprelsa®, AstraZeneca); axitinib (Inlyta®, Pfizer); and lenvatinib (Lenvima®, Eisai); Raf inhibitors, such as sorafenib (Nexavar®, Bayer AG and Onyx); dabrafenib (Tafinlar®, Novartis); and vemurafenib (Zelboraf®, Genentech/Roche); MEK inhibitors, such as cobimetanib (Cotellic®, Exelexis/Genentech/Roche); trametinib (Mekinist®, Novartis); Bcr-Abl tyrosine kinase inhibitors, such as imatinib (Gleevec®, Novartis); nilotinib (Tasigna®, Novartis); dasatinib (Sprycel®, BristolMyersSquibb); bosutinib (Bosulif®, Pfizer); and ponatinib (Inclusig®, Ariad Pharmaceuticals); Her2 and EGFR inhibitors, such as gefitinib (Iressa®, AstraZeneca); erlotinib (Tarceeva®, Genentech/Roche/Astellas); lapatinib (Tykerb®, Novartis); afatinib (Gilotrif®, Boehringer Ingelheim); osimertinib (targeting activated EGFR, Tagrisso®, AstraZeneca); and brigatinib (Alunbrig®, Ariad Pharmaceuticals); c-Met and VEGFR2 inhibitors, such as cabozanitib (Cometriq®, Exelexis); and multikinase inhibitors, such as sunitinib (Sutent®, Pfizer); pazopanib (Votrient®, Novartis); ALK inhibitors, such as crizotinib (Xalkori®, Pfizer); ceritinib (Zykadia®, Novartis); and alectinib (Alecenza®, Genentech/Roche); Bruton's tyrosine kinase inhibitors, such as ibrutinib (Imbruvica®, Pharmacyclics/Janssen); and Flt3 receptor inhibitors, such as midostaurin (Rydapt®, Novartis).


Other kinase inhibitors and VEGF-R antagonists that are in development and may be used in the present invention include tivozanib (Aveo Pharmaecuticals); vatalanib (Bayer/Novartis); lucitanib (Clovis Oncology); dovitinib (TK1258, Novartis); Chiauanib (Chipscreen Biosciences); CEP-11981 (Cephalon); linifanib (Abbott Laboratories); neratinib (HKI-272, Puma Biotechnology); radotinib (Supect®, IY5511, Il-Yang Pharmaceuticals, S. Korea); ruxolitinib (Jakafi®, Incyte Corporation); PTC299 (PTC Therapeutics); CP-547,632 (Pfizer); foretinib (Exelexis, GlaxoSmithKline); quizartinib (Daiichi Sankyo) and motesanib (Amgen/Takeda).


In another embodiment, the present invention provides a method of treating organ transplant rejection or graft vs. host disease comprising administering to a patient in need thereof a provided compound and one or more additional therapeutic agents selected from a steroid, cyclosporin, FK506, rapamycin, a hedgehog signaling inhibitor, a BTK inhibitor, a JAK/pan-JAK inhibitor, a TYK2 inhibitor, a PI3K inhibitor, and a SYK inhibitor.


In another embodiment, the present invention provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a provided compound and a BTK inhibitor, wherein the disease is selected from inflammatory bowel disease, arthritis, systemic lupus erythematosus (SLE), vasculitis, idiopathic thrombocytopenic purpura (ITP), rheumatoid arthritis, psoriatic arthritis, osteoarthritis, Still's disease, juvenile arthritis, diabetes, myasthenia gravis, Hashimoto's thyroiditis, Ord's thyroiditis, Graves' disease, autoimmune thyroiditis, Sjogren's syndrome, multiple sclerosis, systemic sclerosis, Lyme neuroborreliosis, Guillain-Barre syndrome, acute disseminated encephalomyelitis, Addison's disease, opsoclonus-myoclonus syndrome, ankylosing spondylosis, antiphospholipid antibody syndrome, aplastic anemia, autoimmune hepatitis, autoimmune gastritis, pernicious anemia, celiac disease, Goodpasture's syndrome, idiopathic thrombocytopenic purpura, optic neuritis, scleroderma, primary biliary cirrhosis, Reiter's syndrome, Takayasu's arteritis, temporal arteritis, warm autoimmune hemolytic anemia, Wegener's granulomatosis, psoriasis, alopecia universalis, Behcet's disease, chronic fatigue, dysautonomia, membranous glomerulonephropathy, endometriosis, interstitial cystitis, pemphigus vulgaris, bullous pemphigoid, neuromyotonia, scleroderma, vulvodynia, a hyperproliferative disease, rejection of transplanted organs or tissues, Acquired Immunodeficiency Syndrome (AIDS, also known as HIV), type 1 diabetes, graft versus host disease, transplantation, transfusion, anaphylaxis, allergies (e.g., allergies to plant pollens, latex, drugs, foods, insect poisons, animal hair, animal dander, dust mites, or cockroach calyx), type I hypersensitivity, allergic conjunctivitis, allergic rhinitis, and atopic dermatitis, asthma, appendicitis, atopic dermatitis, asthma, allergy, blepharitis, bronchiolitis, bronchitis, bursitis, cervicitis, cholangitis, cholecystitis, chronic graft rejection, colitis, conjunctivitis, Crohn's disease, cystitis, dacryoadenitis, dermatitis, dermatomyositis, encephalitis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, Henoch-Schonlein purpura, hepatitis, hidradenitis suppurativa, immunoglobulin A nephropathy, interstitial lung disease, laryngitis, mastitis, meningitis, myelitis myocarditis, myositis, nephritis, oophoritis, orchitis, osteitis, otitis, pancreatitis, parotitis, pericarditis, peritonitis, pharyngitis, pleuritis, phlebitis, pneumonitis, pneumonia, polymyositis, proctitis, prostatitis, pyelonephritis, rhinitis, salpingitis, sinusitis, stomatitis, synovitis, tendonitis, tonsillitis, ulcerative colitis, uveitis, vaginitis, vasculitis, or vulvitis, B-cell proliferative disorder, e.g., diffuse large B cell lymphoma, follicular lymphoma, chronic lymphocytic lymphoma, chronic lymphocytic leukemia, acute lymphocytic leukemia, B-cell prolymphocytic leukemia, lymphoplasmacytic lymphoma/Waldenstrom macroglobulinemia, splenic marginal zone lymphoma, multiple myeloma (also known as plasma cell myeloma), non-Hodgkin's lymphoma, Hodgkin's lymphoma, plasmacytoma, extranodal marginal zone B cell lymphoma, nodal marginal zone B cell lymphoma, mantle cell lymphoma, mediastinal (thymic) large B cell lymphoma, intravascular large B cell lymphoma, primary effusion lymphoma, Burkitt lymphoma/leukemia, or lymphomatoid granulomatosis, breast cancer, prostate cancer, or cancer of the mast cells (e.g., mastocytoma, mast cell leukemia, mast cell sarcoma, systemic mastocytosis), bone cancer, colorectal cancer, pancreatic cancer, diseases of the bone and joints including, without limitation, rheumatoid arthritis, seronegative spondyloarthropathies (including ankylosing spondylitis, psoriatic arthritis and Reiter's disease), Behcet's disease, Sjogren's syndrome, systemic sclerosis, osteoporosis, bone cancer, bone metastasis, a thromboembolic disorder, (e.g., myocardial infarct, angina pectoris, reocclusion after angioplasty, restenosis after angioplasty, reocclusion after aortocoronary bypass, restenosis after aortocoronary bypass, stroke, transitory ischemia, a peripheral arterial occlusive disorder, pulmonary embolism, deep venous thrombosis), inflammatory pelvic disease, urethritis, skin sunburn, sinusitis, pneumonitis, encephalitis, meningitis, myocarditis, nephritis, osteomyelitis, myositis, hepatitis, gastritis, enteritis, dermatitis, gingivitis, appendicitis, pancreatitis, cholocystitus, agammaglobulinemia, psoriasis, allergy, Crohn's disease, irritable bowel syndrome, ulcerative colitis, Sjogren's disease, tissue graft rejection, hyperacute rejection of transplanted organs, asthma, allergic rhinitis, chronic obstructive pulmonary disease (COPD), autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome), autoimmune alopecia, pernicious anemia, glomerulonephritis, dermatomyositis, multiple sclerosis, scleroderma, vasculitis, autoimmune hemolytic and thrombocytopenic states, Goodpasture's syndrome, atherosclerosis, Addison's disease, Parkinson's disease, Alzheimer's disease, diabetes, septic shock, systemic lupus erythematosus (SLE), rheumatoid arthritis, psoriatic arthritis, juvenile arthritis, osteoarthritis, chronic idiopathic thrombocytopenic purpura, Waldenstrom macroglobulinemia, myasthenia gravis, Hashimoto's thyroiditis, atopic dermatitis, degenerative joint disease, vitiligo, autoimmune hypopituitarism, Guillain-Barre syndrome, Behcet's disease, scleraderma, mycosis fungoides, acute inflammatory responses (such as acute respiratory distress syndrome and ischemia/reperfusion injury), and Graves' disease.


In another embodiment, the present invention provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a provided compound and a PI3K inhibitor, wherein the disease is selected from a cancer, a neurodegenative disorder, an angiogenic disorder, a viral disease, an autoimmune disease, an inflammatory disorder, a hormone-related disease, conditions associated with organ transplantation, immunodeficiency disorders, a destructive bone disorder, a proliferative disorder, an infectious disease, a condition associated with cell death, thrombin-induced platelet aggregation, chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), liver disease, pathologic immune conditions involving T cell activation, a cardiovascular disorder, and a CNS disorder.


In another embodiment, the present invention provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a provided compound and a PI3K inhibitor, wherein the disease is selected from benign or malignant tumor, carcinoma or solid tumor of the brain, kidney (e.g., renal cell carcinoma (RCC)), liver, adrenal gland, bladder, breast, stomach, gastric tumors, ovaries, colon, rectum, prostate, pancreas, lung, vagina, endometrium, cervix, testis, genitourinary tract, esophagus, larynx, skin, bone or thyroid, sarcoma, glioblastomas, neuroblastomas, multiple myeloma or gastrointestinal cancer, especially colon carcinoma or colorectal adenoma or a tumor of the neck and head, an epidermal hyperproliferation, psoriasis, prostate hyperplasia, a neoplasia, a neoplasia of epithelial character, adenoma, adenocarcinoma, keratoacanthoma, epidermoid carcinoma, large cell carcinoma, non-mall-cell lung carcinoma, lymphomas, (including, for example, non-Hodgkin's Lymphoma (NHL) and Hodgkin's lymphoma (also termed Hodgkin's or Hodgkin's disease)), a mammary carcinoma, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, or a leukemia, diseases include Cowden syndrome, Lhermitte-Dudos disease and Bannayan-Zonana syndrome, or diseases in which the PI3K/PKB pathway is aberrantly activated, asthma of whatever type or genesis including both intrinsic (non-allergic) asthma and extrinsic (allergic) asthma, mild asthma, moderate asthma, severe asthma, bronchitic asthma, exercise-induced asthma, occupational asthma and asthma induced following bacterial infection, acute lung injury (ALI), adult/acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary, airways or lung disease (COPD, COAD or COLD), including chronic bronchitis or dyspnea associated therewith, emphysema, as well as exacerbation of airways hyperreactivity consequent to other drug therapy, in particular other inhaled drug therapy, bronchitis of whatever type or genesis including, but not limited to, acute, arachidic, catarrhal, croupus, chronic or phthinoid bronchitis, pneumoconiosis (an inflammatory, commonly occupational, disease of the lungs, frequently accompanied by airways obstruction, whether chronic or acute, and occasioned by repeated inhalation of dusts) of whatever type or genesis, including, for example, aluminosis, anthracosis, asbestosis, chalicosis, ptilosis, siderosis, silicosis, tabacosis and byssinosis, Loffler's syndrome, eosinophilic, pneumonia, parasitic (in particular metazoan) infestation (including tropical eosinophilia), bronchopulmonary aspergillosis, polyarteritis nodosa (including Churg-Strauss syndrome), eosinophilic granuloma and eosinophil-related disorders affecting the airways occasioned by drug-reaction, psoriasis, contact dermatitis, atopic dermatitis, alopecia areata, erythema multiforma, dermatitis herpetiformis, scleroderma, vitiligo, hypersensitivity angiitis, urticaria, bullous pemphigoid, lupus erythematosus, pemphisus, epidermolysis bullosa acquisita, conjunctivitis, keratoconjunctivitis sicca, and vernal conjunctivitis, diseases affecting the nose including allergic rhinitis, and inflammatory disease in which autoimmune reactions are implicated or having an autoimmune component or etiology, including autoimmune hematological disorders (e.g. hemolytic anemia, aplastic anemia, pure red cell anemia and idiopathic thrombocytopenia), systemic lupus erythematosus, rheumatoid arthritis, polychondritis, sclerodoma, Wegener granulamatosis, dermatomyositis, chronic active hepatitis, myasthenia gravis, Steven-Johnson syndrome, idiopathic sprue, autoimmune inflammatory bowel disease (e.g. ulcerative colitis and Crohn's disease), endocrine opthalmopathy, Grave's disease, sarcoidosis, alveolitis, chronic hypersensitivity pneumonitis, multiple sclerosis, primary biliary cirrhosis, uveitis (anterior and posterior), keratoconjunctivitis sicca and vernal keratoconjunctivitis, interstitial lung fibrosis, psoriatic arthritis and glomerulonephritis (with and without nephrotic syndrome, e.g. including idiopathic nephrotic syndrome or minal change nephropathy, restenosis, cardiomegaly, atherosclerosis, myocardial infarction, ischemic stroke and congestive heart failure, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, and cerebral ischemia, and neurodegenerative disease caused by traumatic injury, glutamate neurotoxicity and hypoxia.


In some embodiments, one or more other therapeutic agent is a phosphatidylinositol 3 kinase (PI3K) inhibitor. In some embodiments, a PI3K inhibitor is selected from idelalisib (Zydelig®, Gilead), alpelisib (BYL719, Novartis), taselisib (GDC-0032, Genentech/Roche); pictilisib (GDC-0941, Genentech/Roche); copanlisib (BAY806946, Bayer); duvelisib (formerly IPI-145, Infinity Pharmaceuticals); PQR309 (Piqur Therapeutics, Switzerland); and TGR1202 (formerly RP5230, TG Therapeutics).


The compounds and compositions, according to the method of the present invention, may be administered using any amount and any route of administration effective for treating or lessening the severity of a cancer, an autoimmune disorder, a proliferative disorder, an inflammatory disorder, a neurodegenerative or neurological disorder, schizophrenia, a bone-elated disorder, liver disease, or a cardiac disorder. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like. Compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage. The expression “dosage unit form” as used herein refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.


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


Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.


Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.


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


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


Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.


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


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


The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.


Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this invention. Additionally, the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.


According to one embodiment, the invention relates to a method of inhibiting protein kinase activity or degrading a protein kinase in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.


According to another embodiment, the invention relates to a method of inhibiting or degrading SMARCA2, SMARCA4, or PB1, or a mutant thereof, activity in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.


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


Inhibition and/or degradation of a SMARCA or PB1 protein, or a protein selected from SMARCA2, SMARCA4, or PB1, or a mutant thereof, activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ-transplantation, biological specimen storage, and biological assays.


Another embodiment of the present invention relates to a method of degrading a protein kinase and/or inhibiting protein kinase activity in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound.


According to another embodiment, the invention relates to a method of degrading and/or inhibiting one or more SMARCA2, SMARCA4, or PB1, or a mutant thereof, activity in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound. In other embodiments, the present invention provides a method for treating a disorder mediated by one or more SMARCA2, SMARCA4, or PB1, or a mutant thereof, in a patient in need thereof, comprising the step of administering to said patient a compound according to the present invention or pharmaceutically acceptable composition thereof. Such disorders are described in detail herein.


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


A compound of the current invention may also be used to advantage in combination with other antiproliferative compounds. Such antiproliferative compounds include, but are not limited to aromatase inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase II inhibitors; microtubule active compounds; alkylating compounds; histone deacetylase inhibitors; compounds which induce cell differentiation processes; cyclooxygenase inhibitors; MMP inhibitors; mTOR inhibitors; antineoplastic antimetabolites; platin compounds; compounds targeting/decreasing a protein or lipid kinase activity and further anti-angiogenic compounds; compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase; gonadorelin agonists; anti-androgens; methionine aminopeptidase inhibitors; matrix metalloproteinase inhibitors; bisphosphonates; biological response modifiers; antiproliferative antibodies; heparanase inhibitors; inhibitors of Ras oncogenic isoforms; telomerase inhibitors; proteasome inhibitors; compounds used in the treatment of hematologic malignancies; compounds which target, decrease or inhibit the activity of Flt-3; Hsp90 inhibitors such as 17-AAG (17-allylaminogeldanamycin, NSC330507), 17-DMAG (17-dimethylaminoethylamino-17-demethoxy-geldanamycin, NSC707545), IPI-504, CNF1010, CNF2024, CNF1010 from Conforma Therapeutics; temozolomide (Temodal®); kinesin spindle protein inhibitors, such as SB715992 or SB743921 from GlaxoSmithKline, or pentamidine/chlorpromazine from CombinatoRx; MEK inhibitors such as ARRY142886 from Array BioPharma, AZD6244 from AstraZeneca, PD181461 from Pfizer and leucovorin.


The term “aromatase inhibitor” as used herein relates to a compound which inhibits estrogen production, for instance, the conversion of the substrates androstenedione and testosterone to estrone and estradiol, respectively. The term includes, but is not limited to steroids, especially atamestane, exemestane and formestane and, in particular, non-steroids, especially aminoglutethimide, roglethimide, pyridoglutethimide, trilostane, testolactone, ketokonazole, vorozole, fadrozole, anastrozole and letrozole. Exemestane is marketed under the trade name Aromasin™. Formestane is marketed under the trade name Lentaron™. Fadrozole is marketed under the trade name Afema™. Anastrozole is marketed under the trade name Arimidex™ Letrozole is marketed under the trade names Femara™ or Femar™. Aminoglutethimide is marketed under the trade name Orimeten™. A combination of the invention comprising a chemotherapeutic agent which is an aromatase inhibitor is particularly useful for the treatment of hormone receptor positive tumors, such as breast tumors.


In some embodiments, one or more other therapeutic agent is an mTOR inhibitor, which inhibits cell proliferation, angiogenesis and glucose uptake. In some embodiments, an mTOR inhibitor is everolimus (Afinitor®, Novartis); temsirolimus (Torisel®, Pfizer); and sirolimus (Rapamune®, Pfizer).


In some embodiments, one or more other therapeutic agent is an aromatase inhibitor. In some embodiments, an aromatase inhibitor is selected from exemestane (Aromasin®, Pfizer); anastazole (Arimidex®, AstraZeneca) and letrozole (Femara®, Novartis).


The term “antiestrogen” as used herein relates to a compound which antagonizes the effect of estrogens at the estrogen receptor level. The term includes, but is not limited to tamoxifen, fulvestrant, raloxifene and raloxifene hydrochloride. Tamoxifen is marketed under the trade name Nolvadex™. Raloxifene hydrochloride is marketed under the trade name Evista™. Fulvestrant can be administered under the trade name Faslodex™. A combination of the invention comprising a chemotherapeutic agent which is an antiestrogen is particularly useful for the treatment of estrogen receptor positive tumors, such as breast tumors.


The term “anti-androgen” as used herein relates to any substance which is capable of inhibiting the biological effects of androgenic hormones and includes, but is not limited to, bicalutamide (Casodex™). The term “gonadorelin agonist” as used herein includes, but is not limited to abarelix, goserelin and goserelin acetate. Goserelin can be administered under the trade name Zoladex™.


The term “topoisomerase I inhibitor” as used herein includes, but is not limited to topotecan, gimatecan, irinotecan, camptothecian and its analogues, 9-nitrocamptothecin and the macromolecular camptothecin conjugate PNU-166148. Irinotecan can be administered, e.g. in the form as it is marketed, e.g. under the trademark Camptosar™. Topotecan is marketed under the trade name Hycamptin™.


The term “topoisomerase II inhibitor” as used herein includes, but is not limited to the anthracyclines such as doxorubicin (including liposomal formulation, such as Caelyx™) daunorubicin, epirubicin, idarubicin and nemorubicin, the anthraquinones mitoxantrone and losoxantrone, and the podophillotoxines etoposide and teniposide. Etoposide is marketed under the trade name Etopophos™. Teniposide is marketed under the trade name VM 26-Bristol Doxorubicin is marketed under the trade name Acriblastin™ or Adriamycin™. Epirubicin is marketed under the trade name Farmorubicin™. Idarubicin is marketed. under the trade name Zavedos™. Mitoxantrone is marketed under the trade name Novantron.


The term “microtubule active agent” relates to microtubule stabilizing, microtubule destabilizing compounds and microtublin polymerization inhibitors including, but not limited to taxanes, such as paclitaxel and docetaxel; vinca alkaloids, such as vinblastine or vinblastine sulfate, vincristine or vincristine sulfate, and vinorelbine; discodermolides; cochicine and epothilones and derivatives thereof. Paclitaxel is marketed under the trade name Taxol™ Docetaxel is marketed under the trade name Taxotere™. Vinblastine sulfate is marketed under the trade name Vinblastin R.P™. Vincristine sulfate is marketed under the trade name Farmistin™.


The term “alkylating agent” as used herein includes, but is not limited to, cyclophosphamide, ifosfamide, melphalan or nitrosourea (BCNU or Gliadel). Cyclophosphamide is marketed under the trade name Cyclostin™. Ifosfamide is marketed under the trade name Holoxan™.


The term “histone deacetylase inhibitors” or “HDAC inhibitors” relates to compounds which inhibit the histone deacetylase and which possess antiproliferative activity. This includes, but is not limited to, suberoylanilide hydroxamic acid (SAHA).


The term “antineoplastic antimetabolite” includes, but is not limited to, 5-fluorouracil or 5-FU, capecitabine, gemcitabine, DNA demethylating compounds, such as 5-azacytidine and decitabine, methotrexate and edatrexate, and folic acid antagonists such as pemetrexed. Capecitabine is marketed under the trade name Xeloda™. Gemcitabine is marketed under the trade name Gemzar™.


The term “platin compound” as used herein includes, but is not limited to, carboplatin, cis-platin, cisplatinum and oxaliplatin. Carboplatin can be administered, e.g., in the form as it is marketed, e.g. under the trademark Carboplat™. Oxaliplatin can be administered, e.g., in the form as it is marketed, e.g. under the trademark Eloxatin™.


The term “Bcl-2 inhibitor” as used herein includes, but is not limited to compounds having inhibitory activity against B-cell lymphoma 2 protein (Bcl-2), including but not limited to ABT-199, ABT-731, ABT-737, apogossypol, Ascenta's pan-Bcl-2 inhibitors, curcumin (and analogs thereof), dual Bcl-2/Bcl-xL inhibitors (Infinity Pharmaceuticals/Novartis Pharmaceuticals), Genasense (G3139), HA14-1 (and analogs thereof; see WO2008118802), navitoclax (and analogs thereof, see U.S. Pat. No. 7,390,799), NH-1 (Shenayng Pharmaceutical University), obatoclax (and analogs thereof, see WO2004106328), S-001 (Gloria Pharmaceuticals), TW series compounds (Univ. of Michigan), and venetoclax. In some embodiments the Bcl-2 inhibitor is a small molecule therapeutic. In some embodiments the Bcl-2 inhibitor is a peptidomimetic.


The term “compounds targeting/decreasing a protein or lipid kinase activity; or a protein or lipid phosphatase activity; or further anti-angiogenic compounds” as used herein includes, but is not limited to, protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, such as a) compounds targeting, decreasing or inhibiting the activity of the platelet-derived growth factor-receptors (PDGFR), such as compounds which target, decrease or inhibit the activity of PDGFR, especially compounds which inhibit the PDGF receptor, such as an N-phenyl-2-pyrimidine-amine derivative, such as imatinib, SU101, SU6668 and GFB-111; b) compounds targeting, decreasing or inhibiting the activity of the fibroblast growth factor-eceptors (FGFR); c) compounds targeting, decreasing or inhibiting the activity of the insulin-like growth factor receptor I (IGF-IR), such as compounds which target, decrease or inhibit the activity of IGF-IR, especially compounds which inhibit the kinase activity of IGF-I receptor, or antibodies that target the extracellular domain of IGF-I receptor or its growth factors; d) compounds targeting, decreasing or inhibiting the activity of the Trk receptor tyrosine kinase family, or ephrin B4 inhibitors; e) compounds targeting, decreasing or inhibiting the activity of the AxI receptor tyrosine kinase family; f) compounds targeting, decreasing or inhibiting the activity of the Ret receptor tyrosine kinase; g) compounds targeting, decreasing or inhibiting the activity of the Kit/SCFR receptor tyrosine kinase, such as imatinib; h) compounds targeting, decreasing or inhibiting the activity of the C-kit receptor tyrosine kinases, which are part of the PDGFR family, such as compounds which target, decrease or inhibit the activity of the c-Kit receptor tyrosine kinase family, especially compounds which inhibit the c-Kit receptor, such as imatinib; i) compounds targeting, decreasing or inhibiting the activity of members of the c-Abl family, their gene-fusion products (e.g. BCR-Abl kinase) and mutants, such as compounds which target decrease or inhibit the activity of c-Abl family members and their gene fusion products, such as an N-phenyl-2-pyrimidine-amine derivative, such as imatinib or nilotinib (AMN107); PD180970; AG957; NSC 680410; PD173955 from ParkeDavis; or dasatinib (BMS-354825); j) compounds targeting, decreasing or inhibiting the activity of members of the protein kinase C (PKC) and Raf family of serine/threonine kinases, members of the MEK, SRC, JAK/pan-JAK, FAK, PDK1, PKB/Akt, Ras/MAPK, PI3K, SYK, TYK2, BTK and TEC family, and/or members of the cyclin-ependent kinase family (CDK) including staurosporine derivatives, such as midostaurin; examples of further compounds include UCN-01, safingol, BAY 43-9006, Bryostatin 1, Perifosine; llmofosine; RO 318220 and RO 320432; GO 6976; lsis 3521; LY333531/LY379196; isochinoline compounds; FTIs; PD184352 or QAN697 (a P13K inhibitor) or AT7519 (CDK inhibitor); k) compounds targeting, decreasing or inhibiting the activity of protein-tyrosine kinase inhibitors, such as compounds which target, decrease or inhibit the activity of protein-tyrosine kinase inhibitors include imatinib mesylate (Gleevec™) or tyrphostin such as Tyrphostin A23/RG-50810; AG 99; Tyrphostin AG 213; Tyrphostin AG 1748; Tyrphostin AG 490; Tyrphostin B44; Tyrphostin B44 (+) enantiomer; Tyrphostin AG 555; AG 494; Tyrphostin AG 556, AG957 and adaphostin (4-{[(2,5-dihydroxyphenyl)methyl]amino}-benzoic acid adamantyl ester; NSC 680410, adaphostin); 1) compounds targeting, decreasing or inhibiting the activity of the epidermal growth factor family of receptor tyrosine kinases (EGFRi ErbB2, ErbB3, ErbB4 as homo- or heterodimers) and their mutants, such as compounds which target, decrease or inhibit the activity of the epidermal growth factor receptor family are especially compounds, proteins or antibodies which inhibit members of the EGF receptor tyrosine kinase family, such as EGF receptor, ErbB2, ErbB3 and ErbB4 or bind to EGF or EGF related ligands, CP 358774, ZD 1839, ZM 105180; trastuzumab (Herceptin™), cetuximab (Erbitux™), Iressa, Tarceva, OSI-774, C1-1033, EKB-569, GW-2016, E1.1, E2.4, E2.5, E6.2, E6.4, E2.11, E6.3 or E7.6.3, and 7H-pyrrolo-[2,3-d]pyrimidine derivatives; m) compounds targeting, decreasing or inhibiting the activity of the c-Met receptor, such as compounds which target, decrease or inhibit the activity of c-Met, especially compounds which inhibit the kinase activity of c-Met receptor, or antibodies that target the extracellular domain of c-Met or bind to HGF, n) compounds targeting, decreasing or inhibiting the kinase activity of one or more JAK family members (JAK1/JAK2/JAK3/TYK2 and/or pan-JAK), including but not limited to PRT-062070, SB-1578, baricitinib, pacritinib, momelotinib, VX-509, AZD-1480, TG-101348, tofacitinib, and ruxolitinib; o) compounds targeting, decreasing or inhibiting the kinase activity of PI3 kinase (P13K) including but not limited to ATU-027, SF-1126, DS-7423, PBI-05204, GSK-2126458, ZSTK-474, buparlisib, pictrelisib, PF-4691502, BYL-719, dactolisib, XL-147, XL-765, and idelalisib; and; and q) compounds targeting, decreasing or inhibiting the signaling effects of hedgehog protein (Hh) or smoothened receptor (SMO) pathways, including but not limited to cyclopamine, vismodegib, itraconazole, erismodegib, and IPI-926 (saridegib).


Compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase are e.g. inhibitors of phosphatase 1, phosphatase 2A, or CDC25, such as okadaic acid or a derivative thereof.


In some embodiments, one or more other therapeutic agent is a growth factor antagonist, such as an antagonist of platelet-derived growth factor (PDGF), or epidermal growth factor (EGF) or its receptor (EGFR). Approved PDGF antagonists which may be used in the present invention include olaratumab (Lartruvo®; Eli Lilly). Approved EGFR antagonists which may be used in the present invention include cetuximab (Erbitux®, Eli Lilly); necitumumab (Portrazza®, Eli Lilly), panitumumab (Vectibix®, Amgen); and osimertinib (targeting activated EGFR, Tagrisso®, AstraZeneca).


The term “PI3K inhibitor” as used herein includes, but is not limited to compounds having inhibitory activity against one or more enzymes in the phosphatidylinositol-3-kinase family, including, but not limited to PI3Kα, PI3Kγ, PI3Kδ, PI3Kβ, PI3K-C2α, PI3K-C2β, PI3K-C2γ, Vps34, p110-α, p110-β, p110-γ, p110-δ, p85-α, p85-β, p55-γ, p150, p101, and p87. Examples of PI3K inhibitors useful in this invention include but are not limited to ATU-027, SF-1126, DS-7423, PBI-05204, GSK-2126458, ZSTK-474, buparlisib, pictrelisib, PF-4691502, BYL-719, dactolisib, XL-147, XL-765, and idelalisib.


The term “BTK inhibitor” as used herein includes, but is not limited to compounds having inhibitory activity against Bruton's Tyrosine Kinase (BTK), including, but not limited to AVL-292 and ibrutinib.


The term “SYK inhibitor” as used herein includes, but is not limited to compounds having inhibitory activity against spleen tyrosine kinase (SYK), including but not limited to PRT-062070, R-343, R-333, Excellair, PRT-062607, and fostamatinib


Further examples of BTK inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this invention can be found in WO2008039218 and WO2011090760, the entirety of which are incorporated herein by reference.


Further examples of SYK inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this invention can be found in WO2003063794, WO2005007623, and WO2006078846, the entirety of which are incorporated herein by reference.


Further examples of PI3K inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this invention can be found in WO2004019973, WO2004089925, WO2007016176, U.S. Pat. No. 8,138,347, WO2002088112, WO2007084786, WO2007129161, WO2006122806, WO2005113554, and WO2007044729 the entirety of which are incorporated herein by reference.


Further examples of JAK inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this invention can be found in WO2009114512, WO2008109943, WO2007053452, WO2000142246, and WO2007070514, the entirety of which are incorporated herein by reference.


Further anti-angiogenic compounds include compounds having another mechanism for their activity, e.g. unrelated to protein or lipid kinase inhibition e.g. thalidomide (Thalomid™) and TNP-470.


Examples of proteasome inhibitors useful for use in combination with compounds of the invention include, but are not limited to bortezomib, disulfiram, epigallocatechin-3-gallate (EGCG), salinosporamide A, carfilzomib, ONX-0912, CEP-18770, and MLN9708.


Compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase are e.g. inhibitors of phosphatase 1, phosphatase 2A, or CDC25, such as okadaic acid or a derivative thereof.


Compounds which induce cell differentiation processes include, but are not limited to, retinoic acid, α- γ- or δ-tocopherol or α- γ- or δ-tocotrienol.


The term cyclooxygenase inhibitor as used herein includes, but is not limited to, Cox-2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acid and derivatives, such as celecoxib (Celebrex™), rofecoxib (Vioxx™), etoricoxib, valdecoxib or a 5-alkyl-2-arylaminophenylacetic acid, such as 5-methyl-2-(2′-chloro-6′-fluoroanilino)phenyl acetic acid, lumiracoxib.


The term “bisphosphonates” as used herein includes, but is not limited to, etridonic, clodronic, tiludronic, pamidronic, alendronic, ibandronic, risedronic and zoledronic acid. Etridonic acid is marketed under the trade name Didronel™. Clodronic acid is marketed under the trade name Bonefos™. Tiludronic acid is marketed under the trade name Skelid™. Pamidronic acid is marketed under the trade name Aredia™. Alendronic acid is marketed under the trade name Fosamax™. Ibandronic acid is marketed under the trade name Bondranat™. Risedronic acid is marketed under the trade name Actonel™. Zoledronic acid is marketed under the trade name Zometa™. The term “mTOR inhibitors” relates to compounds which inhibit the mammalian target of rapamycin (mTOR) and which possess antiproliferative activity such as sirolimus (Rapamune®), everolimus (Certican™), CCI-779 and ABT578.


The term “heparanase inhibitor” as used herein refers to compounds which target, decrease or inhibit heparin sulfate degradation. The term includes, but is not limited to, PI-88. The term “biological response modifier” as used herein refers to a lymphokine or interferons.


The term “inhibitor of Ras oncogenic isoforms”, such as H-Ras, K-Ras, or N-Ras, as used herein refers to compounds which target, decrease or inhibit the oncogenic activity of Ras; for example, a “farnesyl transferase inhibitor” such as L-744832, DK8G557 or R115777 (Zarnestra™). The term “telomerase inhibitor” as used herein refers to compounds which target, decrease or inhibit the activity of telomerase. Compounds which target, decrease or inhibit the activity of telomerase are especially compounds which inhibit the telomerase receptor, such as telomestatin.


The term “methionine aminopeptidase inhibitor” as used herein refers to compounds which target, decrease or inhibit the activity of methionine aminopeptidase. Compounds which target, decrease or inhibit the activity of methionine aminopeptidase include, but are not limited to, bengamide or a derivative thereof.


The term “proteasome inhibitor” as used herein refers to compounds which target, decrease or inhibit the activity of the proteasome. Compounds which target, decrease or inhibit the activity of the proteasome include, but are not limited to, Bortezomib (Velcade™); carfilzomib (Kyprolis®, Amgen); and ixazomib (Ninlaro®, Takeda), and MLN 341.


The term “matrix metalloproteinase inhibitor” or (“MMP” inhibitor) as used herein includes, but is not limited to, collagen peptidomimetic and nonpeptidomimetic inhibitors, tetracycline derivatives, e.g. hydroxamate peptidomimetic inhibitor batimastat and its orally bioavailable analogue marimastat (BB-2516), prinomastat (AG3340), metastat (NSC 683551) BMS-279251, BAY 12-9566, TAA211, MMI270B or AAJ996.


The term “compounds used in the treatment of hematologic malignancies” as used herein includes, but is not limited to, FMS-like tyrosine kinase inhibitors, which are compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon, 1-O-D-arabinofuransylcytosine (ara-c) and bisulfan; and ALK inhibitors, which are compounds which target, decrease or inhibit anaplastic lymphoma kinase.


Compounds which target, decrease or inhibit the activity of FMS-like tyrosine kinase receptors (Flt-3R) are especially compounds, proteins or antibodies which inhibit members of the Flt-3R receptor kinase family, such as PKC412, midostaurin, a staurosporine derivative, SU11248 and MLN518.


The term “HSP90 inhibitors” as used herein includes, but is not limited to, compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90; degrading, targeting, decreasing or inhibiting the HSP90 client proteins via the ubiquitin proteosome pathway. Compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90 are especially compounds, proteins or antibodies which inhibit the ATPase activity of HSP90, such as 17-allylamino, 17-demethoxygeldanamycin (17AAG), a geldanamycin derivative; other geldanamycin related compounds; radicicol and HDAC inhibitors.


The term “antiproliferative antibodies” as used herein includes, but is not limited to, trastuzumab (Herceptin™), Trastuzumab-DM1, erbitux, bevacizumab (Avastin™), rituximab (Rituxan®), PR064553 (anti-CD40) and 2C4 Antibody. By antibodies is meant intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least 2 intact antibodies, and antibodies fragments so long as they exhibit the desired biological activity.


For the treatment of acute myeloid leukemia (AML), compounds of the current invention can be used in combination with standard leukemia therapies, especially in combination with therapies used for the treatment of AML. In particular, compounds of the current invention can be administered in combination with, for example, farnesyl transferase inhibitors and/or other drugs useful for the treatment of AML, such as Daunorubicin, Adriamycin, Ara-C, VP-16, Teniposide, Mitoxantrone, Idarubicin, Carboplatinum and PKC412.


Other anti-leukemic compounds include, for example, Ara-C, a pyrimidine analog, which is the 2′-alpha-hydroxy ribose (arabinoside) derivative of deoxycytidine. Also included is the purine analog of hypoxanthine, 6-mercaptopurine (6-MP) and fludarabine phosphate. Compounds which target, decrease or inhibit activity of histone deacetylase (HDAC) inhibitors such as sodium butyrate and suberoylanilide hydroxamic acid (SAHA) inhibit the activity of the enzymes known as histone deacetylases. Specific HDAC inhibitors include MS275, SAHA, FK228 (formerly FR901228), Trichostatin A and compounds disclosed in U.S. Pat. No. 6,552,065 including, but not limited to, N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)-ethyl]-amino]methyl]phenyl]—2E-2-propenamide, or a pharmaceutically acceptable salt thereof and N-hydroxy-3-[4-[(2-hydroxyethyl){2-(1H-indol-3-yl)ethyl]-amino]methyl]phenyl]-2E-2-propenamide, or a pharmaceutically acceptable salt thereof, especially the lactate salt. Somatostatin receptor antagonists as used herein refer to compounds which target, treat or inhibit the somatostatin receptor such as octreotide, and SOM230. Tumor cell damaging approaches refer to approaches such as ionizing radiation. The term “ionizing radiation” referred to above and hereinafter means ionizing radiation that occurs as either electromagnetic rays (such as X-rays and gamma rays) or particles (such as alpha and beta particles). Ionizing radiation is provided in, but not limited to, radiation therapy and is known in the art. See Hellman, Principles of Radiation Therapy, Cancer, in Principles and Practice of Oncology, Devita et al., Eds., 4th Edition, Vol. 1, pp. 248-275 (1993).


Also included are EDG binders and ribonucleotide reductase inhibitors. The term “EDG binders” as used herein refers to a class of immunosuppressants that modulates lymphocyte recirculation, such as FTY720. The term “ribonucleotide reductase inhibitors” refers to pyrimidine or purine nucleoside analogs including, but not limited to, fludarabine and/or cytosine arabinoside (ara-C), 6-thioguanine, 5-fluorouracil, cladribine, 6-mercaptopurine (especially in combination with ara-C against ALL) and/or pentostatin. Ribonucleotide reductase inhibitors are especially hydroxyurea or 2-hydroxy-1H-isoindole-1,3-dione derivatives.


Also included are in particular those compounds, proteins or monoclonal antibodies of VEGF such as 1-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine or a pharmaceutically acceptable salt thereof, 1-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine succinate; Angiostatin™; Endostatin™; anthranilic acid amides; ZD4190; ZD6474; SU5416; SU6668; bevacizumab; or anti-VEGF antibodies or anti-VEGF receptor antibodies, such as rhuMAb and RHUFab, VEGF aptamer such as Macugon; FLT-4 inhibitors, FLT-3 inhibitors, VEGFR-2 IgGI antibody, Angiozyme (RPI 4610) and Bevacizumab (Avastin™).


Photodynamic therapy as used herein refers to therapy which uses certain chemicals known as photosensitizing compounds to treat or prevent cancers. Examples of photodynamic therapy include treatment with compounds, such as Visudyne™ and porfimer sodium.


Angiostatic steroids as used herein refers to compounds which block or inhibit angiogenesis, such as, e.g., anecortave, triamcinolone, hydrocortisone, 11-α-epihydrocotisol, cortexolone, 17α-hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone and dexamethasone.


Implants containing corticosteroids refers to compounds, such as fluocinolone and dexamethasone.


Other chemotherapeutic compounds include, but are not limited to, plant alkaloids, hormonal compounds and antagonists; biological response modifiers, preferably lymphokines or interferons; antisense oligonucleotides or oligonucleotide derivatives; shRNA or siRNA; or miscellaneous compounds or compounds with other or unknown mechanism of action.


The compounds of the invention are also useful as co-therapeutic compounds for use in combination with other drug substances such as anti-inflammatory, bronchodilatory or antihistamine drug substances, particularly in the treatment of obstructive or inflammatory airways diseases such as those mentioned hereinbefore, for example as potentiators of therapeutic activity of such drugs or as a means of reducing required dosaging or potential side effects of such drugs. A compound of the invention may be mixed with the other drug substance in a fixed pharmaceutical composition or it may be administered separately, before, simultaneously with or after the other drug substance. Accordingly the invention includes a combination of a compound of the invention as hereinbefore described with an anti-inflammatory, bronchodilatory, antihistamine or anti-tussive drug substance, said compound of the invention and said drug substance being in the same or different pharmaceutical composition.


Suitable anti-inflammatory drugs include steroids, in particular glucocorticosteroids such as budesonide, beclamethasone dipropionate, fluticasone propionate, ciclesonide or mometasone furoate; non-steroidal glucocorticoid receptor agonists; LTB4 antagonists such LY293111, CGS025019C, CP-195543, SC-53228, BIIL 284, ONO 4057, SB 209247; LTD4 antagonists such as montelukast and zafirlukast; PDE4 inhibitors such cilomilast (Ariflo® GlaxoSmithKline), Roflumilast (Byk Gulden), V-11294A (Napp), BAY19-8004 (Bayer), SCH-351591 (Schering-Plough), Arofylline (Almirall Prodesfarma), PD189659/PD168787 (Parke-Davis), AWD-12-281 (Asta Medica), CDC-801 (Celgene), SeICID™ CC-10004 (Celgene), VM554/UM565 (Vernalis), T-440 (Tanabe), KW-4490 (Kyowa Hakko Kogyo); A2a agonists; A2b antagonists; and beta-2 adrenoceptor agonists such as albuterol (salbutamol), metaproterenol, terbutaline, salmeterol fenoterol, procaterol, and especially, formoterol and pharmaceutically acceptable salts thereof. Suitable bronchodilatory drugs include anticholinergic or antimuscarinic compounds, in particular ipratropium bromide, oxitropium bromide, tiotropium salts and CHF 4226 (Chiesi), and glycopyrrolate.


Suitable antihistamine drug substances include cetirizine hydrochloride, acetaminophen, clemastine fumarate, promethazine, loratidine, desloratidine, diphenhydramine and fexofenadine hydrochloride, activastine, astemizole, azelastine, ebastine, epinastine, mizolastine and tefenadine.


Other useful combinations of compounds of the invention with anti-inflammatory drugs are those with antagonists of chemokine receptors, e.g. CCR-1, CCR-2, CCR-3, CCR-4, CCR-5, CCR-6, CCR-7, CCR-8, CCR-9 and CCR10, CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, particularly CCR-5 antagonists such as Schering-Plough antagonists SC-351125, SCH-55700 and SCH-D, and Takeda antagonists such as N-[[4-[[[6,7-dihydro-2-(4-methylphenyl)-5H-benzo-cyclohepten-8-yl]carbonyl]amino]phenyl]-methyl]tetrahydro-N,N-dimethyl-2H-pyran-4-aminium chloride (TAK-770).


The structure of the active compounds identified by code numbers, generic or trade names may be taken from the actual edition of the standard compendium “The Merck Index” or from databases, e.g. Patents International (e.g. IMS World Publications).


A compound of the current invention may also be used in combination with known therapeutic processes, for example, the administration of hormones or radiation. In certain embodiments, a provided compound is used as a radiosensitizer, especially for the treatment of tumors which exhibit poor sensitivity to radiotherapy.


A compound of the current invention can be administered alone or in combination with one or more other therapeutic compounds, possible combination therapy taking the form of fixed combinations or the administration of a compound of the invention and one or more other therapeutic compounds being staggered or given independently of one another, or the combined administration of fixed combinations and one or more other therapeutic compounds. A compound of the current invention can besides or in addition be administered especially for tumor therapy in combination with chemotherapy, radiotherapy, immunotherapy, phototherapy, surgical intervention, or a combination of these. Long-term therapy is equally possible as is adjuvant therapy in the context of other treatment strategies, as described above. Other possible treatments are therapy to maintain the patient's status after tumor regression, or even chemopreventive therapy, for example in patients at risk.


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


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


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


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


The amount of one or more other therapeutic agent present in the compositions of this invention may be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent. Preferably the amount of one or more other therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent. In some embodiments, one or more other therapeutic agent is administered at a dosage of about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, or about 95% of the amount normally administered for that agent. As used herein, the phrase “normally administered” means the amount an FDA approved therapeutic agent is approved for dosing per the FDA label insert.


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


Exemplary Immuno-Oncology Agents


In some embodiments, one or more other therapeutic agent is an immuno-oncology agent. As used herein, the term “an immuno-oncology agent” refers to an agent which is effective to enhance, stimulate, and/or up-regulate immune responses in a subject. In some embodiments, the administration of an immuno-oncology agent with a compound of the invention has a synergic effect in treating a cancer.


An immuno-oncology agent can be, for example, a small molecule drug, an antibody, or a biologic or small molecule. Examples of biologic immuno-oncology agents include, but are not limited to, cancer vaccines, antibodies, and cytokines. In some embodiments, an antibody is a monoclonal antibody. In some embodiments, a monoclonal antibody is humanized or human.


In some embodiments, an immuno-oncology agent is (i) an agonist of a stimulatory (including a co-stimulatory) receptor or (ii) an antagonist of an inhibitory (including a co-inhibitory) signal on T cells, both of which result in amplifying antigen-specific T cell responses.


Certain of the stimulatory and inhibitory molecules are members of the immunoglobulin super family (IgSF). One important family of membrane-bound ligands that bind to co-stimulatory or co-inhibitory receptors is the B7 family, which includes B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6. Another family of membrane bound ligands that bind to co-stimulatory or co-inhibitory receptors is the TNF family of molecules that bind to cognate TNF receptor family members, which includes CD40 and CD40L, OX-40, OX-40L, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, TACI, APRIL, BCMA, LTβR, LIGHT, DcR3, HVEM, VEGI/TL1A, TRAMP/DR3, EDAR, EDA1, XEDAR, EDA2, TNFR1, Lymphotoxin α/TNFβ, TNFR2, TNFα, LTβR, Lymphotoxin α1β2, FAS, FASL, RELT, DR6, TROY, NGFR.


In some embodiments, an immuno-oncology agent is a cytokine that inhibits T cell activation (e.g., IL-6, IL-10, TGF-β, VEGF, and other immunosuppressive cytokines) or a cytokine that stimulates T cell activation, for stimulating an immune response.


In some embodiments, a combination of a compound of the invention and an immuno-oncology agent can stimulate T cell responses. In some embodiments, an immuno-oncology agent is: (i) an antagonist of a protein that inhibits T cell activation (e.g., immune checkpoint inhibitors) such as CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, Galectin 9, CEACAM-1, BTLA, CD69, Galectin-1, TIGIT, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, and TIM-4; or (ii) an agonist of a protein that stimulates T cell activation such as B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD70, CD27, CD40, DR3 and CD28H.


In some embodiments, an immuno-oncology agent is an antagonist of inhibitory receptors on NK cells or an agonists of activating receptors on NK cells. In some embodiments, an immuno-oncology agent is an antagonists of KIR, such as lirilumab.


In some embodiments, an immuno-oncology agent is an agent that inhibits or depletes macrophages or monocytes, including but not limited to CSF-1R antagonists such as CSF-1R antagonist antibodies including RG7155 (WO11/70024, WO11/107553, WO11/131407, WO13/87699, WO13/119716, WO13/132044) or FPA-008 (WO11/140249; WO13169264; WO14/036357).


In some embodiments, an immuno-oncology agent is selected from agonistic agents that ligate positive costimulatory receptors, blocking agents that attenuate signaling through inhibitory receptors, antagonists, and one or more agents that increase systemically the frequency of anti-tumor T cells, agents that overcome distinct immune suppressive pathways within the tumor microenvironment (e.g., block inhibitory receptor engagement (e.g., PD-L1/PD-1 interactions), deplete or inhibit Tregs (e.g., using an anti-CD25 monoclonal antibody (e.g., daclizumab) or by ex vivo anti-CD25 bead depletion), inhibit metabolic enzymes such as IDO, or reverse/prevent T cell energy or exhaustion) and agents that trigger innate immune activation and/or inflammation at tumor sites.


In some embodiments, an immuno-oncology agent is a CTLA-4 antagonist. In some embodiments, a CTLA-4 antagonist is an antagonistic CTLA-4 antibody. In some embodiments, an antagonistic CTLA-4 antibody is YERVOY (ipilimumab) or tremelimumab.


In some embodiments, an immuno-oncology agent is a PD-1 antagonist. In some embodiments, a PD-1 antagonist is administered by infusion. In some embodiments, an immuno-oncology agent is an antibody or an antigen-binding portion thereof that binds specifically to a Programmed Death-1 (PD-1) receptor and inhibits PD-1 activity. In some embodiments, a PD-1 antagonist is an antagonistic PD-1 antibody. In some embodiments, an antagonistic PD-1 antibody is OPDIVO (nivolumab), KEYTRUDA (pembrolizumab), or MEDI-0680 (AMP-514; WO2012/145493). In some embodiments, an immuno-oncology agent may be pidilizumab (CT-011). In some embodiments, an immuno-oncology agent is a recombinant protein composed of the extracellular domain of PD-L2 (B7-DC) fused to the Fc portion of IgGI, called AMP-224.


In some embodiments, an immuno-oncology agent is a PD-L1 antagonist. In some embodiments, a PD-L1 antagonist is an antagonistic PD-L1 antibody. In some embodiments, a PD-L1 antibody is MPDL3280A (RG7446; WO2010/077634), durvalumab (MEDI4736), BMS-936559 (WO2007/005874), and MSB0010718C (WO2013/79174).


In some embodiments, an immuno-oncology agent is a LAG-3 antagonist. In some embodiments, a LAG-3 antagonist is an antagonistic LAG-3 antibody. In some embodiments, a LAG3 antibody is BMS-986016 (WO10/19570, WO14/08218), or IMP-731 or PIP-321 (WO08/132601, WO009/44273).


In some embodiments, an immuno-oncology agent is a CD137 (4-1BB) agonist. In some embodiments, a CD137 (4-1BB) agonist is an agonistic CD137 antibody. In some embodiments, a CD137 antibody is urelumab or PF-05082566 (WO12/32433).


In some embodiments, an immuno-oncology agent is a GITR agonist. In some embodiments, a GITR agonist is an agonistic GITR antibody. In some embodiments, a GITR antibody is BMS-986153, BMS-986156, TRX-518 (WO006/105021, WO009/009116), or MK-4166 (WO11/028683).


In some embodiments, an immuno-oncology agent is an indoleamine (2,3)-dioxygenase (IDO) antagonist. In some embodiments, an IDO antagonist is selected from epacadostat (INCB024360, Incyte); indoximod (NLG-8189, NewLink Genetics Corporation); capmanitib (INC280, Novartis); GDC-0919 (Genentech/Roche); PF-06840003 (Pfizer); BMS:F001287 (Bristol-Myers Squibb); Phy906/KD108 (Phytoceutica); an enzyme that breaks down kynurenine (Kynase, Kyn Therapeutics); and NLG-919 (WO09/73620, WO009/1156652, WO11/56652, WO12/142237).


In some embodiments, an immuno-oncology agent is an OX40 agonist. In some embodiments, an OX40 agonist is an agonistic OX40 antibody. In some embodiments, an OX40 antibody is MEDI-6383 or MEDI-6469.


In some embodiments, an immuno-oncology agent is an OX40L antagonist. In some embodiments, an OX40L antagonist is an antagonistic OX40 antibody. In some embodiments, an OX40L antagonist is RG-7888 (WO06/029879).


In some embodiments, an immuno-oncology agent is a CD40 agonist. In some embodiments, a CD40 agonist is an agonistic CD40 antibody. In some embodiments, an immuno-oncology agent is a CD40 antagonist. In some embodiments, a CD40 antagonist is an antagonistic CD40 antibody. In some embodiments, a CD40 antibody is lucatumumab or dacetuzumab.


In some embodiments, an immuno-oncology agent is a CD27 agonist. In some embodiments, a CD27 agonist is an agonistic CD27 antibody. In some embodiments, a CD27 antibody is varlilumab.


In some embodiments, an immuno-oncology agent is MGA271 (to B7H3) (WO11/109400).


In some embodiments, an immuno-oncology agent is abagovomab, adecatumumab, afutuzumab, alemtuzumab, anatumomab mafenatox, apolizumab, atezolimab, avelumab, blinatumomab, BMS-936559, catumaxomab, durvalumab, epacadostat, epratuzumab, indoximod, inotuzumab ozogamicin, intelumumab, ipilimumab, isatuximab, lambrolizumab, MED14736, MPDL3280A, nivolumab, obinutuzumab, ocaratuzumab, ofatumumab, olatatumab, pembrolizumab, pidilizumab, rituximab, ticilimumab, samalizumab, or tremelimumab.


In some embodiments, an immuno-oncology agent is an immunostimulatory agent. For example, antibodies blocking the PD-1 and PD-L1 inhibitory axis can unleash activated tumor-reactive T cells and have been shown in clinical trials to induce durable anti-tumor responses in increasing numbers of tumor histologies, including some tumor types that conventionally have not been considered immunotherapy sensitive. See, e.g., Okazaki, T. et al. (2013) Nat. Immunol. 14, 1212-1218; Zou et al. (2016) Sci. Transl. Med. 8. The anti-PD-1 antibody nivolumab (Opdivo®, Bristol-Myers Squibb, also known as ONO-4538, MDX1106 and BMS-936558), has shown potential to improve the overall survival in patients with RCC who had experienced disease progression during or after prior anti-angiogenic therapy.


In some embodiments, the immunomodulatory therapeutic specifically induces apoptosis of tumor cells. Approved immunomodulatory therapeutics which may be used in the present invention include pomalidomide (Pomalyst®, Celgene); lenalidomide (Revlimid®, Celgene); ingenol mebutate (Picato®, LEO Pharma).


In some embodiments, an immuno-oncology agent is a cancer vaccine. In some embodiments, the cancer vaccine is selected from sipuleucel-T (Provenge®, Dendreon/Valeant Pharmaceuticals), which has been approved for treatment of asymptomatic, or minimally symptomatic metastatic castrate-resistant (hormone-refractory) prostate cancer; and talimogene laherparepvec (Imlygic®, BioVex/Amgen, previously known as T-VEC), a genetically modified oncolytic viral therapy approved for treatment of unresectable cutaneous, subcutaneous and nodal lesions in melanoma. In some embodiments, an immuno-oncology agent is selected from an oncolytic viral therapy such as pexastimogene devacirepvec (PexaVec/JX-594, SillaJen/formerly Jennerex Biotherapeutics), a thymidine kinase-(TK-) deficient vaccinia virus engineered to express GM-CSF, for hepatocellular carcinoma (NCT02562755) and melanoma (NCT00429312); pelareorep (Reolysin®, Oncolytics Biotech), a variant of respiratory enteric orphan virus (reovirus) which does not replicate in cells that are not RAS-activated, in numerous cancers, including colorectal cancer (NCT01622543); prostate cancer (NCT01619813); head and neck squamous cell cancer (NCT01166542); pancreatic adenocarcinoma (NCT00998322); and non-mall cell lung cancer (NSCLC) (NCT 00861627); enadenotucirev (NG-348, PsiOxus, formerly known as ColoAd1), an adenovirus engineered to express a full length CD80 and an antibody fragment specific for the T-cell receptor CD3 protein, in ovarian cancer (NCT02028117); metastatic or advanced epithelial tumors such as in colorectal cancer, bladder cancer, head and neck squamous cell carcinoma and salivary gland cancer (NCT02636036); ONCOS-102 (Targovax/formerly Oncos), an adenovirus engineered to express GM-CSF, in melanoma (NCT03003676); and peritoneal disease, colorectal cancer or ovarian cancer (NCT02963831); GL-ONC1 (GLV-1h68/GLV-1h153, Genelux GmbH), vaccinia viruses engineered to express beta-alactosidase (beta-gal)/beta-glucoronidase or beta-gal/human sodium iodide symporter (hNIS), respectively, were studied in peritoneal carcinomatosis (NCT01443260); fallopian tube cancer, ovarian cancer (NCT 02759588); or CG0070 (Cold Genesys), an adenovirus engineered to express GM-CSF, in bladder cancer (NCT02365818).


In some embodiments, an immuno-oncology agent is selected from JX-929 (SillaJen/formerly Jennerex Biotherapeutics), a TK- and vaccinia growth factor-deficient vaccinia virus engineered to express cytosine deaminase, which is able to convert the prodrug 5-fluorocytosine to the cytotoxic drug 5-fluorouracil; TG01 and TG02 (Targovax/formerly Oncos), peptide-based immunotherapy agents targeted for difficult-to-treat RAS mutations; and TILT-123 (TILT Biotherapeutics), an engineered adenovirus designated: Ad5/3-E2F-delta24-hTNFα-IRES-hIL20; and VSV-GP (ViraTherapeutics) a vesicular stomatitis virus (VSV) engineered to express the glycoprotein (GP) of lymphocytic choriomeningitis virus (LCMV), which can be further engineered to express antigens designed to raise an antigen-specific CD8+ T cell response.


In some embodiments, an immuno-oncology agent is a T-cell engineered to express a chimeric antigen receptor, or CAR. The T-cells engineered to express such chimeric antigen receptor are referred to as a CAR-T cells.


CARs have been constructed that consist of binding domains, which may be derived from natural ligands, single chain variable fragments (scFv) derived from monoclonal antibodies specific for cell-surface antigens, fused to endodomains that are the functional end of the T-cell receptor (TCR), such as the CD3-zeta signaling domain from TCRs, which is capable of generating an activation signal in T lymphocytes. Upon antigen binding, such CARs link to endogenous signaling pathways in the effector cell and generate activating signals similar to those initiated by the TCR complex.


For example, in some embodiments the CAR-T cell is one of those described in U.S. Pat. No. 8,906,682 (June; hereby incorporated by reference in its entirety), which discloses CAR-T cells engineered to comprise an extracellular domain having an antigen binding domain (such as a domain that binds to CD19), fused to an intracellular signaling domain of the T cell antigen receptor complex zeta chain (such as CD3 zeta). When expressed in the T cell, the CAR is able to redirect antigen recognition based on the antigen binding specificity. In the case of CD19, the antigen is expressed on malignant B cells. Over 200 clinical trials are currently in progress employing CAR-T in a wide range of indications. [https://clinicaltrials.gov/ct2/results?term=chimeric+antigen+receptors&pg=1].


In some embodiments, an immunostimulatory agent is an activator of retinoic acid receptor-related orphan receptor γ (RORγt). RORγt is a transcription factor with key roles in the differentiation and maintenance of Type 17 effector subsets of CD4+(Th17) and CD8+ (Tc17) T cells, as well as the differentiation of IL-17 expressing innate immune cell subpopulations such as NK cells. In some embodiments, an activator of RORγt is LYC-55716 (Lycera), which is currently being evaluated in clinical trials for the treatment of solid tumors (NCT02929862).


In some embodiments, an immunostimulatory agent is an agonist or activator of a toll-like receptor (TLR). Suitable activators of TLRs include an agonist or activator of TLR9 such as SD-101 (Dynavax). SD-101 is an immunostimulatory CpG which is being studied for B-cell, follicular and other lymphomas (NCT02254772). Agonists or activators of TLR8 which may be used in the present invention include motolimod (VTX-2337, VentiRx Pharmaceuticals) which is being studied for squamous cell cancer of the head and neck (NCT02124850) and ovarian cancer (NCT02431559).


Other immuno-oncology agents that may be used in the present invention include urelumab (BMS-663513, Bristol-Myers Squibb), an anti-CD137 monoclonal antibody; varlilumab (CDX-1127, Celldex Therapeutics), an anti-CD27 monoclonal antibody; BMS-986178 (Bristol-Myers Squibb), an anti-OX40 monoclonal antibody; lirilumab (IPH2102/BMS-986015, Innate Pharma, Bristol-Myers Squibb), an anti-KIR monoclonal antibody; monalizumab (IPH2201, Innate Pharma, AstraZeneca) an anti-NKG2A monoclonal antibody; andecaliximab (GS-5745, Gilead Sciences), an anti-MMP9 antibody; MK-4166 (Merck & Co.), an anti-GITR monoclonal antibody.


In some embodiments, an immunostimulatory agent is selected from elotuzumab, mifamurtide, an agonist or activator of a toll-like receptor, and an activator of RORγt.


In some embodiments, an immunostimulatory therapeutic is recombinant human interleukin 15 (rhIL-15). rhIL-15 has been tested in the clinic as a therapy for melanoma and renal cell carcinoma (NCT01021059 and NCT01369888) and leukemias (NCT02689453). In some embodiments, an immunostimulatory agent is recombinant human interleukin 12 (rhIL-12). In some embodiments, an IL-15 based immunotherapeutic is heterodimeric IL-15 (hetIL-15, Novartis/Admune), a fusion complex composed of a synthetic form of endogenous IL-15 complexed to the soluble IL-15 binding protein IL-15 receptor alpha chain (IL15:sIL-15RA), which has been tested in Phase 1 clinical trials for melanoma, renal cell carcinoma, non-small cell lung cancer and head and neck squamous cell carcinoma (NCT02452268). In some embodiments, a recombinant human interleukin 12 (rhIL-12) is NM-IL-12 (Neumedicines, Inc.), NCT02544724, or NCT02542124.


In some embodiments, an immuno-oncology agent is selected from those descripted in Jerry L. Adams et al., “Big opportunities for small molecules in immuno-oncology,” Cancer Therapy 2015, Vol. 14, pages 603-622, the content of which is incorporated herein by reference in its entirety. In some embodiments, an immuno-oncology agent is selected from the examples described in Table 1 of Jerry L. Adams et al. In some embodiments, an immuno-oncology agent is a small molecule targeting an immuno-oncology target selected from those listed in Table 2 of Jerry L. Adams ET. AL. In some embodiments, an immuno-oncology agent is a small molecule agent selected from those listed in Table 2 of Jerry L. Adams et al.


In some embodiments, an immuno-oncology agent is selected from the small molecule immuno-oncology agents described in Peter L. Toogood, “Small molecule immuno-oncology therapeutic agents,” Bioorganic & Medicinal Chemistry Letters 2018, Vol. 28, pages 319-329, the content of which is incorporated herein by reference in its entirety. In some embodiments, an immuno-oncology agent is an agent targeting the pathways as described in Peter L. Toogood.


In some embodiments, an immuno-oncology agent is selected from those described in Sandra L. Ross et al., “Bispecific T cell engager (BiTE®) antibody constructs can mediate bystander tumor cell killing”, PLoS ONE 12(8): e0183390, the content of which is incorporated herein by reference in its entirety. In some embodiments, an immuno-oncology agent is a bispecific T cell engager (BiTE®) antibody construct. In some embodiments, a bispecific T cell engager (BiTE®) antibody construct is a CD19/CD3 bispecific antibody construct. In some embodiments, a bispecific T cell engager (BiTE®) antibody construct is an EGFR/CD3 bispecific antibody construct. In some embodiments, a bispecific T cell engager (BiTE®) antibody construct activates T cells. In some embodiments, a bispecific T cell engager (BiTE®) antibody construct activates T cells, which release cytokines inducing upregulation of intercellular adhesion molecule 1 (ICAM-1) and FAS on bystander cells. In some embodiments, a bispecific T cell engager (BiTE®) antibody construct activates T cells which result in induced bystander cell lysis. In some embodiments, the bystander cells are in solid tumors. In some embodiments, the bystander cells being lysed are in proximity to the BiTE®-activated T cells. In some embodiment, the bystander cells comprises tumor-associated antigen (TAA) negative cancer cells. In some embodiment, the bystander cells comprise EGFR-negative cancer cells. In some embodiments, an immuno-oncology agent is an antibody which blocks the PD-L1/PD1 axis and/or CTLA4. In some embodiments, an immuno-oncology agent is an ex-vivo expanded tumor-infiltrating T cell. In some embodiments, an immuno-oncology agent is a bispecific antibody construct or chimeric antigen receptors (CARs) that directly connect T cells with tumor-associated surface antigens (TAAs).


Exemplary Immune Checkpoint Inhibitors


In some embodiments, an immuno-oncology agent is an immune checkpoint inhibitor as described herein.


The term “checkpoint inhibitor” as used herein relates to agents useful in preventing cancer cells from avoiding the immune system of the patient. One of the major mechanisms of anti-tumor immunity subversion is known as “T-cell exhaustion,” which results from chronic exposure to antigens that has led to up-regulation of inhibitory receptors. These inhibitory receptors serve as immune checkpoints in order to prevent uncontrolled immune reactions.


PD-1 and co-inhibitory receptors such as cytotoxic T-lymphocyte antigen 4 (CTLA-4, B and T Lymphocyte Attenuator (BTLA; CD272), T cell Immunoglobulin and Mucin domain-3 (Tim-3), Lymphocyte Activation Gene-3 (Lag-3; CD223), and others are often referred to as a checkpoint regulators. They act as molecular “gatekeepers” that allow extracellular information to dictate whether cell cycle progression and other intracellular signaling processes should proceed.


In some embodiments, an immune checkpoint inhibitor is an antibody to PD-1. PD-1 binds to the programmed cell death 1 receptor (PD-1) to prevent the receptor from binding to the inhibitory ligand PDL-1, thus overriding the ability of tumors to suppress the host anti-tumor immune response.


In one aspect, the checkpoint inhibitor is a biologic therapeutic or a small molecule. In another aspect, the checkpoint inhibitor is a monoclonal antibody, a humanized antibody, a fully human antibody, a fusion protein or a combination thereof. In a further aspect, the checkpoint inhibitor inhibits a checkpoint protein selected from CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands or a combination thereof. In an additional aspect, the checkpoint inhibitor interacts with a ligand of a checkpoint protein selected from CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands or a combination thereof. In an aspect, the checkpoint inhibitor is an immunostimulatory agent, a T cell growth factor, an interleukin, an antibody, a vaccine or a combination thereof. In a further aspect, the interleukin is IL-7 or IL-15. In a specific aspect, the interleukin is glycosylated IL-7. In an additional aspect, the vaccine is a dendritic cell (DC) vaccine.


Checkpoint inhibitors include any agent that blocks or inhibits in a statistically significant manner, the inhibitory pathways of the immune system. Such inhibitors may include small molecule inhibitors or may include antibodies, or antigen binding fragments thereof, that bind to and block or inhibit immune checkpoint receptors or antibodies that bind to and block or inhibit immune checkpoint receptor ligands. Illustrative checkpoint molecules that may be targeted for blocking or inhibition include, but are not limited to, CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, GAL9, LAG3, TIM3, VISTA, KIR, 2B4 (belongs to the CD2 family of molecules and is expressed on all NK, γδ, and memory CD8+ (αβ) T cells), CD160 (also referred to as BY55), CGEN-15049, CHK 1 and CHK2 kinases, A2aR, and various B-7 family ligands. B7 family ligands include, but are not limited to, B7-1, B7-2, B7-DC, B7-H1, B7-H2, B7-H3, B7-H4, B7-H5, B7-H6 and B7-H7. Checkpoint inhibitors include antibodies, or antigen binding fragments thereof, other binding proteins, biologic therapeutics, or small molecules, that bind to and block or inhibit the activity of one or more of CTLA-4, PDL1, PDL2, PD1, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD 160 and CGEN-15049. Illustrative immune checkpoint inhibitors include Tremelimumab (CTLA-4 blocking antibody), anti-OX40, PD-L1 monoclonal Antibody (Anti-B7-H1; MEDI4736), MK-3475 (PD-1 blocker), Nivolumab (anti-PD1 antibody), CT-011 (anti-PD1 antibody), BY55 monoclonal antibody, AMP224 (anti-PDL1 antibody), BMS-936559 (anti-PDL1 antibody), MPLDL3280A (anti-PDL1 antibody), MSB0010718C (anti-PDL1 antibody), and ipilimumab (anti-CTLA-4 checkpoint inhibitor). Checkpoint protein ligands include, but are not limited to PD-L1, PD-L2, B7-H3, B7-H4, CD28, CD86 and TIM-3.


In certain embodiments, the immune checkpoint inhibitor is selected from a PD-1 antagonist, a PD-L1 antagonist, and a CTLA-4 antagonist. In some embodiments, the checkpoint inhibitor is selected from the group consisting of nivolumab (Opdivo®), ipilimumab (Yervoy®), and pembrolizumab (Keytruda®). In some embodiments, the checkpoint inhibitor is selected from nivolumab (anti-PD-1 antibody, Opdivo®, Bristol-Myers Squibb); pembrolizumab (anti-PD-1 antibody, Keytruda®, Merck); ipilimumab (anti-CTLA-4 antibody, Yervoy®, Bristol-Myers Squibb); durvalumab (anti-PD-L1 antibody, Imfinzi®, AstraZeneca); and atezolizumab (anti-PD-L1 antibody, Tecentriq®, Genentech).


In some embodiments, the checkpoint inhibitor is selected from the group consisting of lambrolizumab (MK-3475), nivolumab (BMS-936558), pidilizumab (CT-011), AMP-224, MDX-1105, MEDI4736, MPDL3280A, BMS-936559, ipilimumab, lirlumab, IPH2101, pembrolizumab (Keytruda®), and tremelimumab.


In some embodiments, an immune checkpoint inhibitor is REGN2810 (Regeneron), an anti-PD-1 antibody tested in patients with basal cell carcinoma (NCT03132636); NSCLC (NCT03088540); cutaneous squamous cell carcinoma (NCT02760498); lymphoma (NCT02651662); and melanoma (NCT03002376); pidilizumab (CureTech), also known as CT-011, an antibody that binds to PD-1, in clinical trials for diffuse large B-cell lymphoma and multiple myeloma; avelumab (Bavencio®, Pfizer/Merck KGaA), also known as MSB0010718C), a fully human IgGI anti-PD-L1 antibody, in clinical trials for non-small cell lung cancer, Merkel cell carcinoma, mesothelioma, solid tumors, renal cancer, ovarian cancer, bladder cancer, head and neck cancer, and gastric cancer; or PDR001 (Novartis), an inhibitory antibody that binds to PD-1, in clinical trials for non-small cell lung cancer, melanoma, triple negative breast cancer and advanced or metastatic solid tumors. Tremelimumab (CP-675,206; Astrazeneca) is a fully human monoclonal antibody against CTLA-4 that has been in studied in clinical trials for a number of indications, including: mesothelioma, colorectal cancer, kidney cancer, breast cancer, lung cancer and non-small cell lung cancer, pancreatic ductal adenocarcinoma, pancreatic cancer, germ cell cancer, squamous cell cancer of the head and neck, hepatocellular carcinoma, prostate cancer, endometrial cancer, metastatic cancer in the liver, liver cancer, large B-cell lymphoma, ovarian cancer, cervical cancer, metastatic anaplastic thyroid cancer, urothelial cancer, fallopian tube cancer, multiple myeloma, bladder cancer, soft tissue sarcoma, and melanoma. AGEN-1884 (Agenus) is an anti-CTLA4 antibody that is being studied in Phase 1 clinical trials for advanced solid tumors (NCT02694822).


In some embodiments, a checkpoint inhibitor is an inhibitor of T-cell immunoglobulin mucin containing protein-3 (TIM-3). TIM-3 inhibitors that may be used in the present invention include TSR-022, LY3321367 and MBG453. TSR-022 (Tesaro) is an anti-TIM-3 antibody which is being studied in solid tumors (NCT02817633). LY3321367 (Eli Lilly) is an anti-TIM-3 antibody which is being studied in solid tumors (NCT03099109). MBG453 (Novartis) is an anti-TIM-3 antibody which is being studied in advanced malignancies (NCT02608268).


In some embodiments, a checkpoint inhibitor is an inhibitor of T cell immunoreceptor with Ig and ITIM domains, or TIGIT, an immune receptor on certain T cells and NK cells. TIGIT inhibitors that may be used in the present invention include BMS-986207 (Bristol-Myers Squibb), an anti-TIGIT monoclonal antibody (NCT02913313); OMP-313M32 (Oncomed); and anti-TIGIT monoclonal antibody (NCT03119428).


In some embodiments, a checkpoint inhibitor is an inhibitor of Lymphocyte Activation Gene-3 (LAG-3). LAG-3 inhibitors that may be used in the present invention include BMS-986016 and REGN3767 and IMP321. BMS-986016 (Bristol-Myers Squibb), an anti-LAG-3 antibody, is being studied in glioblastoma and gliosarcoma (NCT02658981). REGN3767 (Regeneron), is also an anti-LAG-3 antibody, and is being studied in malignancies (NCT03005782). IMP321 (Immutep S.A.) is an LAG-3-Ig fusion protein, being studied in melanoma (NCT02676869); adenocarcinoma (NCT02614833); and metastatic breast cancer (NCT00349934).


Checkpoint inhibitors that may be used in the present invention include OX40 agonists. OX40 agonists that are being studied in clinical trials include PF-04518600/PF-8600 (Pfizer), an agonistic anti-OX40 antibody, in metastatic kidney cancer (NCT03092856) and advanced cancers and neoplasms (NCT02554812; NCT05082566); GSK3174998 (Merck), an agonistic anti-OX40 antibody, in Phase 1 cancer trials (NCT02528357); MEDI0562 (Medimmune/AstraZeneca), an agonistic anti-OX40 antibody, in advanced solid tumors (NCT02318394 and NCT02705482); MEDI6469, an agonistic anti-OX40 antibody (Medimmune/AstraZeneca), in patients with colorectal cancer (NCT02559024), breast cancer (NCT01862900), head and neck cancer (NCT02274155) and metastatic prostate cancer (NCT01303705); and BMS-986178 (Bristol-Myers Squibb) an agonistic anti-OX40 antibody, in advanced cancers (NCT02737475).


Checkpoint inhibitors that may be used in the present invention include CD137 (also called 4-1BB) agonists. CD137 agonists that are being studied in clinical trials include utomilumab (PF-05082566, Pfizer) an agonistic anti-CD137 antibody, in diffuse large B-cell lymphoma (NCT02951156) and in advanced cancers and neoplasms (NCT02554812 and NCT05082566); urelumab (BMS-663513, Bristol-Myers Squibb), an agonistic anti-CD137 antibody, in melanoma and skin cancer (NCT02652455) and glioblastoma and gliosarcoma (NCT02658981).


Checkpoint inhibitors that may be used in the present invention include CD27 agonists. CD27 agonists that are being studied in clinical trials include varlilumab (CDX-1127, Celldex Therapeutics) an agonistic anti-CD27 antibody, in squamous cell head and neck cancer, ovarian carcinoma, colorectal cancer, renal cell cancer, and glioblastoma (NCT02335918); lymphomas (NCT01460134); and glioma and astrocytoma (NCT02924038).


Checkpoint inhibitors that may be used in the present invention include glucocorticoid-induced tumor necrosis factor receptor (GITR) agonists. GITR agonists that are being studied in clinical trials include TRX518 (Leap Therapeutics), an agonistic anti-GITR antibody, in malignant melanoma and other malignant solid tumors (NCT01239134 and NCT02628574); GWN323 (Novartis), an agonistic anti-GITR antibody, in solid tumors and lymphoma (NCT 02740270); INCAGN01876 (Incyte/Agenus), an agonistic anti-GITR antibody, in advanced cancers (NCT02697591 and NCT03126110); MK-4166 (Merck), an agonistic anti-GITR antibody, in solid tumors (NCT02132754) and MEDI1873 (Medimmune/AstraZeneca), an agonistic hexameric GITR-ligand molecule with a human IgGI Fc domain, in advanced solid tumors (NCT02583165).


Checkpoint inhibitors that may be used in the present invention include inducible T-cell co-stimulator (ICOS, also known as CD278) agonists. ICOS agonists that are being studied in clinical trials include MEDI-570 (Medimmune), an agonistic anti-ICOS antibody, in lymphomas (NCT02520791); GSK3359609 (Merck), an agonistic anti-ICOS antibody, in Phase 1 (NCT02723955); JTX-2011 (Jounce Therapeutics), an agonistic anti-ICOS antibody, in Phase 1 (NCT02904226).


Checkpoint inhibitors that may be used in the present invention include killer IgG-like receptor (KTR) inhibitors. KTR inhibitors that are being studied in clinical trials include lirilumab (IPH2102/BMS-986015, Innate Pharma/Bristol-Myers Squibb), an anti-KIR antibody, in leukemias (NCT01687387, NCT02399917, NCT02481297, NCT02599649), multiple myeloma (NCT02252263), and lymphoma (NCT01592370); IPH2101 (1-7F9, Innate Pharma) in myeloma (NCT01222286 and NCT01217203); and IPH4102 (Innate Pharma), an anti-KIR antibody that binds to three domains of the long cytoplasmic tail (KIR3DL2), in lymphoma (NCT02593045).


Checkpoint inhibitors that may be used in the present invention include CD47 inhibitors of interaction between CD47 and signal regulatory protein alpha (SIRPa). CD47/SIRPa inhibitors that are being studied in clinical trials include ALX-148 (Alexo Therapeutics), an antagonistic variant of (SIRPa) that binds to CD47 and prevents CD47/SIRPa-mediated signaling, in phase 1 (NCT03013218); TTI-621 (SIRPa-Fc, Trillium Therapeutics), a soluble recombinant fusion protein created by linking the N-terminal CD47-binding domain of SIRPa with the Fc domain of human IgGI, acts by binding human CD47, and preventing it from delivering its “do not eat” signal to macrophages, is in clinical trials in Phase 1 (NCT02890368 and NCT02663518); CC-90002 (Celgene), an anti-CD47 antibody, in leukemias (NCT02641002); and Hu5F9-G4 (Forty Seven, Inc.), in colorectal neoplasms and solid tumors (NCT02953782), acute myeloid leukemia (NCT02678338) and lymphoma (NCT02953509).


Checkpoint inhibitors that may be used in the present invention include CD73 inhibitors. CD73 inhibitors that are being studied in clinical trials include MEDI9447 (Medimmune), an anti-CD73 antibody, in solid tumors (NCT02503774); and BMS-986179 (Bristol-Myers Squibb), an anti-CD73 antibody, in solid tumors (NCT02754141).


Checkpoint inhibitors that may be used in the present invention include agonists of stimulator of interferon genes protein (STING, also known as transmembrane protein 173, or TMEM173). Agonists of STING that are being studied in clinical trials include MK-1454 (Merck), an agonistic synthetic cyclic dinucleotide, in lymphoma (NCT03010176); and ADU-S100 (MIW815, Aduro Biotech/Novartis), an agonistic synthetic cyclic dinucleotide, in Phase 1 (NCT02675439 and NCT03172936).


Checkpoint inhibitors that may be used in the present invention include CSF1R inhibitors. CSF1R inhibitors that are being studied in clinical trials include pexidartinib (PLX3397, Plexxikon), a CSF1R small molecule inhibitor, in colorectal cancer, pancreatic cancer, metastatic and advanced cancers (NCT02777710) and melanoma, non-small cell lung cancer, squamous cell head and neck cancer, gastrointestinal stromal tumor (GIST) and ovarian cancer (NCT02452424); and IMC-CS4 (LY3022855, Lilly), an anti-CSF-1R antibody, in pancreatic cancer (NCT03153410), melanoma (NCT03101254), and solid tumors (NCT02718911); and BLZ945 (4-[2((1R,2R)-2-hydroxycyclohexylamino)-benzothiazol-6-yloxyl]-pyridine-2-carboxylic acid methylamide, Novartis), an orally available inhibitor of CSF1R, in advanced solid tumors (NCT02829723).


Checkpoint inhibitors that may be used in the present invention include NKG2A receptor inhibitors. NKG2A receptor inhibitors that are being studied in clinical trials include monalizumab (IPH2201, Innate Pharma), an anti-NKG2A antibody, in head and neck neoplasms (NCT02643550) and chronic lymphocytic leukemia (NCT02557516).


In some embodiments, the immune checkpoint inhibitor is selected from nivolumab, pembrolizumab, ipilimumab, avelumab, durvalumab, atezolizumab, or pidilizumab.


EXEMPLIFICATION

Abbreviations

    • Ac: acetyl
    • AcOH: acetic acid
    • ACN: acetonitrile
    • Ad: adamantly
    • AIBN: 2,2′-azo bisisobutyronitrile
    • Anhyd: anhydrous
    • Aq: aqueous
    • B2Pin2: bis (pinacolato)diboron-4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(1,3,2-dioxaborolane)
    • BINAP: 2,2′-bis(diphenylphosphino)-1,1′-binaphthyl
    • BH3: Borane
    • Bn: benzyl
    • Boc: tert-butoxycarbonyl
    • Boc2O: di-tert-butyl dicarbonate
    • BPO: benzoyl peroxide
    • nBuOH: n-butanol
    • CDI: carbonyldiimidazole
    • COD: cyclooctadiene
    • d: days
    • DABCO: 1,4-diazobicyclo[2.2.2]octane
    • DAST: diethylaminosulfur trifluoride
    • dba: dibenzylideneacetone
    • DBU: 1,8-diazobicyclo[5.4.0]undec-7-ene
    • DCE: 1,2-dichloroethane
    • DCM: dichloromethane
    • DEA: diethylamine
    • DHP: dihydropyran
    • DIBAL-H: diisobutylaluminum hydride
    • DIPA: diisopropylamine
    • DIPEA or DIEA: N,N-diisopropylethylamine
    • DMA: N,N-dimethylacetamide
    • DME: 1,2-dimethoxyethane
    • DMAP: 4-dimethylaminopyridine
    • DMF: N,N-dimethylformamide
    • DMP: Dess-Martin periodinane
    • DMSO-dimethyl sulfoxide
    • DPPA: diphenylphosphoryl azide
    • dppf: 1,1′-bis(diphenylphosphino)ferrocene
    • EDC or EDCI: 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
    • ee: enantiomeric excess
    • ESI: electrospray ionization
    • EA: ethyl acetate
    • EtOAc: ethyl acetate
    • EtOH: ethanol
    • FA: formic acid
    • h or hrs: hours
    • HATU: N,N,N′,N′-tetramethyl-O-(7-azabenzotriazol-1-yl)uronium hexafluorophosphate
    • HCl: hydrochloric acid
    • HPLC: high performance liquid chromatography
    • HOAc: acetic acid
    • IBX: 2-iodoxybenzoic acid
    • IPA: isopropyl alcohol
    • KHMDS: potassium hexamethyldisilazide
    • K2CO3: potassium carbonate
    • LAH: lithium aluminum hydride
    • LDA: lithium diisopropylamide
    • m-CPBA: meta-chloroperbenzoic acid
    • M: molar
    • MeCN: acetonitrile
    • MeOH: methanol
    • Me2S: dimethyl sulfide
    • MeONa: sodium methylate
    • MeI: iodomethane
    • min: minutes
    • mL: milliliters
    • mM: millimolar
    • mmol: millimoles
    • MPa: mega pascal
    • MOMCl: methyl chloromethyl ether
    • MsCl: methanesulfonyl chloride
    • MTBE: methyl tert-butyl ether
    • nBuLi: n-butyllithium
    • NaNO2: sodium nitrite
    • NaOH: sodium hydroxide
    • Na2SO4: sodium sulfate
    • NBS: N-bromosuccinimide
    • NCS: N-chlorosuccinimide
    • NFSI: N-Fluorobenzenesulfonimide
    • NMO: N-rnethylmorpholine N-oxide
    • NMP: N-methylpyrrolidine
    • NMR: Nuclear Magnetic Resonance
    • ° C.: degrees Celsius
    • Pd/C: Palladium on Carbon
    • Pd(OAc)2: Palladium Acetate
    • PBS: phosphate buffered saline
    • PE: petroleum ether
    • POCl3: phosphorus oxychloride
    • PPh3: triphenylphosphine
    • PyBOP: (Benzotriazol-1-yloxy)tripyrrolidinophosphonium hexafluorophosphate
    • Rel: relative
    • R.T. or rt: room temperature
    • sat: saturated
    • SEMCl: chloromethyl-2-trimethylsilylethyl ether
    • SFC: supercritical fluid chromatography
    • SOCl2: sulfur dichloride
    • tBuOK: potassium tert-butoxide
    • TBAB: tetrabutylammonium bromide
    • TBAI: tetrabutylammonium iodide
    • TEA: triethylamine
    • Tf: trifluoromethanesulfonate
    • TfAA, TFMSA or Tf2O: trifluoromethanesulfonic anhydride
    • TFA: trifluoracetic acid
    • TIPS: triisopropylsilyl
    • THF: tetrahydrofuran
    • THP: tetrahydropyran
    • TLC: thin layer chromatography
    • TMEDA: tetramethylethylenediamine
    • pTSA: para-toluenesulfonic acid
    • wt: weight
    • Xantphos: 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene


General Synthetic Methods


The following examples are intended to illustrate the invention and are not to be construed as being limitations thereon. Temperatures are given in degrees centigrade. If not mentioned otherwise, all evaporations are performed under reduced pressure, preferably between about 15 mm Hg and 100 mm Hg (=20-133 mbar). The structure of final products, intermediates and starting materials is confirmed by standard analytical methods, e.g., microanalysis and spectroscopic characteristics, e.g., MS, IR, NMR. Abbreviations used are those conventional in the art.


All starting materials, building blocks, reagents, acids, bases, dehydrating agents, solvents, and catalysts utilized to synthesis the compounds of the present invention are either commercially available or can be produced by organic synthesis methods known to one of ordinary skill in the art (Houben-Weyl 4th Ed. 1952, Methods of Organic Synthesis, Thieme, Volume 21). Further, the compounds of the present invention can be produced by organic synthesis methods known to one of ordinary skill in the art as shown in the following examples.


All reactions are carried out under nitrogen or argon unless otherwise stated.


Proton NMR (1H NMR) is conducted in deuterated solvent. In certain compounds disclosed herein, one or more 1H shifts overlap with residual proteo solvent signals; these signals have not been reported in the experimental provided hereinafter.









TABLE 2





Analytical instruments
















LCMS
Shimadzu UFLC MS: LCMS-2020



Agilent Technologies 1200 series MS: Agilent Technologies



6110



Agilent Technologies 1200 series MS: LC/MSD VL


NMR
BRUKER AVANCE III/400; Frequency (MHz) 400.13;



Nucleus: 1H; Number of Transients: 8


Prep-HPLC
Gilson GX-281 systems: instruments GX-A, GX-B, GX-C,



GX-D, GX-E, GX-F, GX-G and GX-H


GCMS
SHIMADZU GCMS-QP2010 Ultra


Analytical
Agilent Technologies 1290 Infinity


cSFC



Prep-cSFC
Waters SFC Prep 80









For acidic LCMS data: LCMS was recorded on an Agilent 1200 Series LC/MSD or Shimadzu LCMS2020 equipped with electro-spray ionization and quadruple MS detector [ES+ve to give MILE] and equipped with Chromolith Flash RP-18e 25*2.0 mm, eluting with 0.0375 vol % TFA in water (solvent A) and 0.01875 vol % TFA in acetonitrile (solvent B). Other LCMS was recorded on an Agilent 1290 Infinity RRLC attached with Agilent 6120 Mass detector. The column used was BEH C18 50*2.1 mm, 1.7 micron. Column flow was 0.55 ml/min and mobile phase were used (A) 2 mM Ammonium Acetate in 0.1% Formic Acid in Water and (B) 0.1% Formic Acid in Acetonitrile.


For basic LCMS data: LCMS was recorded on an Agilent 1200 Series LC/MSD or Shimadzu LCMS 2020 equipped with electro-spray ionization and quadruple MS detector [ES+ve to give MH+] and equipped with Xbridge C18, 2.1×50 mm columns packed with 5 mm C18-coated silica or Kinetex EVO C18 2.1×30 mm columns packed with 5 mm C18-coated silica, eluting with 0.05 vol % NH3H2O in water (solvent A) and acetonitrile (solvent B).


HPLC Analytical Method: HPLC was carried out on X Bridge C18 150*4.6 mm, 5 micron. Column flow was 1.0 ml/min and mobile phase were used (A) 0.1% Ammonia in water and (B) 0.1% Ammonia in Acetonitrile.


Prep HPLC Analytical Method: The compound was purified on Shimadzu LC-20AP and UV detector. The column used was X-BRIDGE C18 (250*19) mm, 5. Column flow was 16.0 ml/min. Mobile phase were used (A) 0.1% Formic Acid in Water and (B) Acetonitrile Basic method used (A) 5 mM ammonium bicarbonate and 0.1% NH3 in Water and (B) Acetonitrile or (A) 0.1% Ammonium Hydroxide in Water and (B) Acetonitrile. The UV spectra were recorded at 202 nm & 254 nm.


NMR Method: The 1H NMR spectra were recorded on a Bruker Ultra Shield Advance 400 MHz/5 mm Probe (BBFO). The chemical shifts are reported in part-per-million.


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


Example 1. 6′-Amino-1-methyl-5′-(1-(1-phenylethyl)-1H-pyrazol-4-yl)-[3,3′-bipyridin]-2(1H)-one (I-1)



embedded image


Step 1: 1-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2-one

To a solution of 3-bromo-1-methyl-pyridin-2-one (1.00 g, 5.32 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (2.70 g, 10.6 mmol) in dioxane (20 mL) was added potassium acetate (1.57 g, 16.0 mmol) and Pd(dppf)Cl2.CH2Cl2 (217 mg, 266 umol). The mixture was stirred at 100° C. for 12 hours. The reaction mixture was diluted with water 50 mL and extracted with ethyl acetate 90 mL (30 mL×3). The combined organic layers were washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase HPLC (0.1% HCl condition) to give the title compound (331 mg, 15% yield) as black oil. 1H NMR (400 MHz, DMSO-d6) δ=7.82 (dd, J=2.4, 6.8 Hz, 1H), 7.72 (dd, J=2.4, 6.8 Hz, 1H), 6.16 (t, J=6.8 Hz, 1H), 3.37 (s, 3H), 1.23 (s, 12H).


Step 2: 1-(1-phenylethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole

A mixture of 1-bromoethylbenzene (5.00 g, 27.0 mmol), 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (5.24 g, 27.0 mmol) and cesium carbonate (26.4 g, 81.1 mmol) in DMF (45 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25° C. for 12 hours under N2 atmosphere. The reaction mixture was diluted with water 60 mL and extracted with ethyl acetate 150 mL (50 mL×3). The combined organic layers were washed with Brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=1:0 to 100:1) to give the title compound (7.71 g, 40% yield) as a yellow oil. H NMR (400 MHz, DMSO-d6) δ=8.05 (s, 1H), 7.95 (s, 1H), 7.61 (s, 1H), 7.31 (d, J=6.4 Hz, 2H), 7.27-7.25 (m, 2H), 5.65-5.60 (m, 1H), 1.80 (d, J=7.2 Hz, 3H), 1.23 (s, 12H).


Step 3: 5-bromo-3-[1-(1-phenylethyl)pyrazol-4-yl]pyridin-2-amine

To a solution of 5-bromo-3-iodo-pyridin-2-amine (2.50 g, 8.36 mmol) and 1-(1-phenylethyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazole (2.99 g, 10.0 mmol) in a mixture of toluene (35 mL) and ethanol (17.5 mL) was added Pd(PPh3)2Cl2 (264 mg, 376 umol) and sodium carbonate (2.39 g, 22.6 mmol). The mixture was stirred at 90° C. for 12 hours. The reaction mixture was diluted with water 60 mL and extracted with ethyl acetate 150 mL (50 mL×3). The combined organic layers were washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=1:0 to 1:5) to give the title compound (2.00 g, 59% yield) was obtained as a yellow oil. 1H NMR (400 MHz, CDCl3) δ=8.02 (d, J=2.4 Hz, 1H), 7.75 (s, 1H), 7.60 (s, 1H), 7.49 (d, J=2.4 Hz, 1H), 7.40-7.30 (m, 3H), 7.28 (d, J=1.6 Hz, 1H), 7.26 (s, 1H), 5.56 (q, J=7.0 Hz, 1H), 1.95 (d, J=7.2 Hz, 3H).


Step 4: 3-[6-amino-5-[1-(1-phenylethyl)pyrazol-4-yl]-3-pyridyl]-1-methyl-pyridin-2-one

To a solution of 5-bromo-3-[1-(1-phenylethyl)pyrazol-4-yl]pyridin-2-amine (390 mg, 1.14 mmol) and 1-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2-one (280 mg, 1.19 mmol) in dioxane (1.2 mL) and water (0.3 mL) was added Pd(dppf)Cl2—CH2Cl2 (92.8 mg, 113.63 umol) and K2CO3 (471 mg, 3.41 mmol). The mixture was stirred at 90° C. for 12 hours. The reaction mixture was diluted with water (30 mL) and extracted with ethyl acetate 60 mL (20 mL×3). The combined organic layers were washed with brine 20 mL, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase HPLC (0.1% HCl condition) to give the title compound (55.9 mg, 12% yield) as a yellow gum. 1H NMR (400 MHz, CD3OD) δ=8.48-8.42 (m, 1H), 8.37 (s, 1H), 8.31 (d, J=1.6 Hz, 1H), 8.03 (s, 1H), 7.90 (d, J=7.2 Hz, 1H), 7.76 (d, J=6.0 Hz, 1H), 7.40-7.29 (m, 5H), 6.50 (t, J=6.8 Hz, 1H), 5.75 (q, J=6.8 Hz, 1H), 3.64 (s, 3H), 2.00-1.93 (d, J=6.8 Hz, 3H); LC-MS (ESI, m/z): [M+1]+=372.1.


Example 2. 3-[6-Amino-5-(1H-pyrazol-4-yl)-3-pyridyl]-1-methyl-pyridin-2-one (I-2)



embedded image


Step 1: 5-bromo-3-(1H-pyrazol-4-yl)pyridin-2-amine

To a solution of 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (1.30 g, 6.69 mmol) in a mixture of toluene (28 mL) and ethanol (14 mL) was added 5-bromo-3-iodo-pyridin-2-amine (2.00 g, 6.69 mmol), sodium carbonate (1.91 g, 18.1 mmol) and Pd(PPh3)2Cl2 (211 mg, 0.301 mmol). The mixture was stirred at 80° C. for 12 hours. The reaction mixture was diluted with water (60 mL) and extracted with ethyl acetate 120 mL (40 mL×3). The combined organic layers were washed with brine (60 mL), dried over by anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chomatography (SiO2, Petroleum ether/Ethyl acetate=1:0 to 1:4) to give the title compound (264 mg, 9% yield) as a white solid. LC-MS (ESI, m/z): [M+1]+=240.9.


Step 2: 3-[6-amino-5-(1H-pyrazol-4-yl)-3-pyridyl]-1-methyl-pyridin-2-one

To a solution of 5-bromo-3-(1H-pyrazol-4-yl)pyridin-2-amine (80 mg, 0.334 mmol) and 1-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2-one (118 mg, 0.501 mmol) in a mixture of dioxane (2.4 mL) and water (0.8 mL) was added Pd(dppf)Cl2—CH2Cl2 (27.3 mg, 0.033 mmol) and K2CO3 (138 mg, 1.00 mmol). The mixture was stirred at 90° C. for 12 hours under N2 atmosphere. The reaction mixture was diluted with water (30 mL) and extracted with ethyl acetate 60 mL (20 mL×3). The combined organic layers were washed with brine (20 mL), dried over by anhydrous sodium sulfate, concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase flash (0.1% HCl) and the residue was purified by prep-HPLC (column: Luna C18 150*25*5 um; mobile phase: [water (0.075% TFA)-ACN]; B %: 1%-30%, 9 min). Then the product was added HCl (5 mL) to give the title compound (4.62 mg, 4% yield) as a grey solid. 1H-NMR (400 MHz, CD3OD) δ=8.46 (s, 1H), 8.32 (s, 1H), 8.17 (s, 2H), 7.90 (d, J=6.4 Hz, 1H), 7.76 (d, J=6.4 Hz, 1H), 6.51 (t, J=6.4 Hz, 1H), 3.65 (s, 3H); LC-MS (ESI, m/z): [M+1]+=268.1.


Example 3. 2-((1-benzyl-1H-pyrazol-4-yl)methyl)-8-hydroxyisoquinolin-1(2H)-one (I-3) and 2-((1H-pyrazol-4-yl)methyl)-8-hydroxyisoquinolin-1(2H)-one (I-4)



embedded image


Step 1: 2-((1-benzyl-1H-pyrazol-4-yl)methyl)-8-bromoisoquinolin-1(2H)-one

A solution of 1-benzyl-4-(chloromethyl)pyrazole (1 g, 4.84 mmol), K2CO3 (803 mg, 5.81 mmol), KI (80.3 mg, 484 umol) and 18-CROWN-6 (25.6 mg, 96.8 umol) in toluene (10 mL) was stirred at 25° C. for 0.5 hour, then a solution of 8-bromo-2H-isoquinolin-1-one (1.30 g, 5.81 mmol) in toluene (10 mL) was added to the above solution and stirred at 120° C. for 12 hours. On completion, filtered to combine the organic phase and evaporated the solvent. The residue was purified by column chromatography (SiO2, PE/EA=10/1 to 2/1) to give the title compound (0.6 g, 31% yield) as a yellow solid.


Step 2: 2-((1-benzyl-1H-pyrazol-4-yl)methyl)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)isoquinolin-1(2H)-one

To a solution of 2-[(1-benzylpyrazol-4-yl)methyl]-8-bromo-isoquinolin-1-one (1 g, 2.54 mmol), KOAc (747 mg, 7.61 mmol) and Pin2B2 (1.29 g, 5.07 mmol) in DMSO (10 mL) was added Pd(dppf)Cl2 (92.8 mg, 126.8 umol), then the mixture was stirred at 90° C. for 12 hours. On completion, water (15 mL) was added to the above solution and the mixture was extracted with ethyl acetate (3×20 mL), combined the organic phase and dried over Na2SO4, concentrated under reduced pressure to give a residue. The residue was purified by column chromatography or by prep-TLC (SiO2, PE:EA=1:1) to give the title compound (1 g, 89% yield) as a yellow oil.


Step 3: 2-((1-benzyl-1H-pyrazol-4-yl)methyl)-8-hydroxyisoquinolin-1(2H)-one

To a solution of 2-[(1-benzylpyrazol-4-yl)methyl]-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)isoquinolin-1-one (200 mg, 453 umol) in THF (8 mL) was added NaOH (1 M, 1.36 mL), followed by H2O2 (435 uL, 30% purity) at 0° C., the mixture was stirred at 20° C. for 2 hours. On completion, water (10 mL) was added to the solution and the mixture was extracted with ethyl acetate (3×6 mL), combined the organic phase and evaporated the solvent. Half of the crude was used for the next step directly and the other was purified by prep-HPLC (column: Phenomenex Synergi C18 150*25*10 um; mobile phase: [water (0.05% HCl)-ACN]; B %: 45%-65%, 10 min) to give the title compound (17 mg, 10% yield) as a white solid. 1H NMR (400 MHz, CD30D) δ=7.83 (s, 1H), 7.64 (s, 2H), 7.52 (m, 1H), 7.36-7.31 (m, 4H), 7.23-7.21 (m, 2H), 7.00 (m, 1H), 6.83 (d, J=8 Hz, 1H), 6.20 (d, J=5.6 Hz, 1H), 5.31 (s, 2H), 5.06 (s, 2H); LC-MS (ESI, m/z): [M+1]*=332.1.


Step 4: 2-((1H-pyrazol-4-yl)methyl)-8-hydroxyisoquinolin-1(2H)-one

To a solution of 2-[(1-benzylpyrazol-4-yl)methyl]-8-hydroxy-isoquinolin-1-one (100 mg, 301.8 umol) in EtOH (10 mL) was added Pd(OH)2 (200 mg) and cyclohexene (5 mL), then the mixture was stirred under H2 (15 psi) at 80° C. for 6 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*25*10 um; mobile phase: [water (0.05% HCl)-ACN]; B %: 23%-43%, 10 min) to give the title product (30 mg, 36% yield) as a white solid. 1H NMR (400 MHz, CD3OD) δ=8.21 (s, 2H), 7.56-7.52 (m, 1H), 7.43 (d, J=7.2 Hz, 1H), 7.02 (d, J=7.2 Hz, 1H), 6.84 (d, J=8.0 Hz, 1H), 6.66 (d, J=8.0 Hz, 1H); LC-MS (ESI, m/z): [M+1]+=242.3.


Example 4. Tert-butyl-2-((8-hydroxy-1-oxoisoquinolin-2(1H)-yl)methyl)pyrrolidine-1-carboxylate (I-6), 8-hydroxy-2-(pyrrolidin-2-ylmethyl)isoquinolin-1(2H)-one (I-7), and 8-hydroxy-2-((1-(pyridin-2-yl)pyrrolidin-2-yl)methyl)isoquinolin-1(2H)-one (I-8)



embedded image


Step 1: tert-butyl 2-((tosyloxy)methyl)pyrrolidine-1-carboxylate

To a solution of tert-butyl 2-(hydroxymethyl)pyrrolidine-1-carboxylate (3.00 g, 14.9 mmol) in DCM (30 mL) was added Et3N (3.02 g, 29.8 mmol, 4.15 mL) and TosCl (3.69 g, 19.3 mmol), then the mixture was stirred at 25° C. for 12 hours. On completion, the mixture was concentrated in vacuo. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=20/1 to 3/1) to give the title compound (3.00 g, 57% yield) as yellow oil. LC-MS (ESI, m/z): [M+1]+=356.1.


Step 2: tert-butyl 2-((8-bromo-1-oxoisoquinolin-2(1H)-yl)methyl)pyrrolidine-1-carboxylate

A solution of tert-butyl 2-(p-tolylsulfonyloxymethyl)pyrrolidine-1-carboxylate (1.00 g, 2.81 mmol), K2CO3 (777 mg, 5.63 mmol), KI (46.7 mg, 281 umol), 18-CROWN-6 (22.3 mg, 84.4 umol) in toluene (10 mL) was stirred at 25° C. for 0.5 hour, then a solution of 8-bromo-2H-isoquinolin-1-one (756 mg, 3.38 mmol) in toluene (10 mL) was added to the above solution and stirred at 120° C. for 12 hours. On completion, water (20 mL) was added to the above solution and the mixture was extracted with ethyl acetate (3×20 mL), combined the organic phase and evaporated the solvent in vacuo. The residue was purified by reversed phase flash (0.1% FA) to give the title compound (800 mg, 70% yield) as yellow solid. LC-MS (ESI, m/z): [M+1]+=407.3.


Step 3: tert-butyl 2-((1-oxo-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)isoquinolin-2(1H)-yl)methyl)pyrrolidine-1-carboxylate

A mixture of tert-butyl 2-[(8-bromo-1-oxo-2-isoquinolyl)methyl]pyrrolidine-1-carboxylate (150 mg, 368 umol), KOAc (108 mg, 1.10 mmol), Pin2B2 (187 mg, 736 umol) and Pd(dppf)Cl2—CH2Cl2 (15.0 mg, 18.4 umol) in DMSO (2 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 105° C. for 12 hours under N2 atmosphere. On completion, the reaction mixture was partitioned between H2O (10 mL) and EA (10 mL). The organic phase was separated, washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, PE:EA=1:1) to give the title compound (90.0 mg, 43% yield) as a yellow liquid. LC-MS (ESI, m/z): [M+1]+=455.2.


Step 4: tert-butyl 2-((8-hydroxy-1-oxoisoquinolin-2(1H)-yl)methyl)pyrrolidine-1-carboxylate

A mixture of tert-butyl 2-[[1-oxo-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-isoquinolyl]methyl]pyrrolidine-1-carboxylate (50.0 mg, 110 umol), H2O2 (124 mg, 1.10 mmol, 105 uL, 30% purity), NaOH (1 M, 330 uL) in THE (8 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 0° C. for 2 hours under N2 atmosphere. On completion, the reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give the title compound (15.0 mg, 38.9% yield) as yellow solid. 1H NMR (400 MHz, CDCl3) δ=12.94-12.89 (m, 1H), 7.54-7.33 (m, 1H), 6.83 (dd, J=7.2, 18.0 Hz, 2H), 6.43 (d, J=7.6 Hz, 1H), 4.36-3.75 (m, 3H), 3.70-3.09 (m, 2H), 1.91-1.71 (m, 4H), 1.47-1.22 (m, 9H); LC-MS (ESI, m/z): [M+1]+=345.1.


Step 5: 8-hydroxy-2-(pyrrolidin-2-ylmethyl)isoquinolin-1(2H)-one

A mixture of tert-butyl 2-[(8-hydroxy-1-oxo-2-isoquinolyl)methyl]pyrrolidine-1-carboxylate (100 mg, 290 umol) in TFA (0.2 mL) and DCM (1.8 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25° C. for 2 hours under N2 atmosphere. On completion, the reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give the title compound (45.0 mg, 53% yield) as red liquid. 1H NMR (400 MHz, DMSO-d6) δ=12.9 (s, 1H), 8.27 (s, 1H), 7.59 (s, 1H), 7.51-7.47 (m, 1H), 7.11-7.07 (m, 1H), 6.84 (d, J=8.0 Hz, 1H), 6.74 (d, J=7.6 Hz, 1H), 4.12-4.05 (m, 1H), 4.12-4.04 (m, 1H), 4.25-4.04 (m, 1H), 3.72-3.63 (m, 1H), 3.10 (s, 1H), 3.02-2.94 (m, 1H), 2.04-1.95 (m, 1H), 1.92-1.74 (m, 1H), 1.65-1.53 (m, 1H); LC-MS (ESI, m/z): [M+1]+=245.1.


Step 6: 8-hydroxy-2-((1-(pyridin-2-yl)pyrrolidin-2-yl)methyl)isoquinolin-1(2H)-one

A mixture of 8-hydroxy-2-(pyrrolidin-2-ylmethyl)isoquinolin-1-one (10.0 mg, 40.9 umol), 2-chloropyrimidine (9.38 mg, 81.8 umol), DBU (37.3 mg, 245 umol, 37.0 uL) in DMSO (1 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 120° C. for 12 hours under N2 atmosphere. On completion, the reaction mixture was purified by prep-HPLC to give the title compound (6.00 mg, 43% yield) as a white solid. 1H NMR (400 MHz, CDCl3) δ=12.91 (s, 1H), 8.26-8.14 (m, 2H), 7.42 (t, J=8.0 Hz, 1H), 7.05 (d, J=7.6 Hz, 1H), 6.86-6.77 (m, 2H), 6.46-6.35 (m, 2H), 4.50-4.42 (m, 1H), 4.42-4.34 (m, 1H), 3.94 (dd, J=4.0, 12.8 Hz, 1H), 3.57 (dd, J=4.8, 6.0 Hz, 1H), 3.46 (s, 1H), 2.12-2.02 (m, 1H), 2.01-1.86 (m, 3H); LC-MS (ESI, m/z): [M+1]+=323.1.


Example 5. Tert-butyl 3-((8-hydroxy-1-oxoisoquinolin-2(1H)-yl)methyl)pyrrolidine-1-carboxylate (I-9), 8-hydroxy-2-(pyrrolidin-3-ylmethyl)isoquinolin-1(2H)-one (I-10), and 8-hydroxy-2-((1-(pyridin-2-yl)pyrrolidin-3-yl)methyl)isoquinolin-1(2H)-one (I-5)



embedded image


Step 1: tert-butyl 3-((tosyloxy)methyl)pyrrolidine-1-carboxylate

To a solution of tert-butyl 3-(hydroxymethyl)pyrrolidine-1-carboxylate (3.00 g, 14.9 mmol) in DCM (30 mL) was added Et3N (3.02 g, 29.8 mmol, 4.15 mL) and TosCl (3.41 g, 17.8 mmol), then the mixture was stirred at 25° C. for 12 hours. On completion, the mixture was concentrated in vacuo. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=20/1 to 3/1) to give the title compound (3.20 g, 60% yield) as yellow oil.


Step 2: tert-butyl 3-((8-bromo-1-oxoisoquinolin-2(1H)-yl)methyl)pyrrolidine-1-carboxylate

A mixture of tert-butyl 3-(p-tolylsulfonyloxymethyl)pyrrolidine-1-carboxylate (1.00 g, 2.81 mmol), K2CO3 (466 mg, 3.38 mmol), KI (46.7 mg, 281 umol), 18-CROWN-6 (22.3 mg, 84.4 umol) in toluene (10 mL) was stirred at 25° C. for 0.5 hour, then a solution of 8-bromo-2H-isoquinolin-1-one (756 mg, 3.38 mmol) in toluene (10 mL) was added to the above solution and stirred at 120° C. for 12 hours. On completion, water (20 mL) was added to the above solution and the mixture was extracted with ethyl acetate (3×20 mL), combined the organic phase and evaporated the solvent in vacuo. The residue was purified by reversed phase flash (FA) to give the title compound (700 mg, 61% yield) as white solid. LC-MS (ESI, m/z): [M+1]+=407.1.


Step 3: tert-butyl 3-((1-oxo-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)isoquinolin-2(1H)-yl)methyl)pyrrolidine-1-carboxylate

To a solution of tert-butyl 3-[(8-bromo-1-oxo-2-isoquinolyl)methyl]pyrrolidine-1-carboxylate (500 mg, 1.23 mmol), KOAc (361 mg, 3.68 mmol) and Pin2B2 (623 mg, 2.46 mmol) in DMSO (5 mL) was added Pd(dppf)Cl2 (89.8 mg, 122 umol), then the mixture was stirred at 90° C. for 12 hours. On completion, water (6 mL) was added to the above solution and the mixture was extracted with ethyl acetate (3×5 mL), combined the organic phase and evaporated the solvent to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=20/1 to 5/1) to give the title compound (150 mg, 27% yield) as a yellow solid. LC-MS (ESI, m/z): [M+1]*=455.2.


Step 4: tert-butyl 3-((8-hydroxy-1-oxoisoquinolin-2(1H)-yl)methyl)pyrrolidine-1-carboxylate

A mixture of tert-butyl 3-[[1-oxo-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-isoquinolyl]methyl]pyrrolidine-1-carboxylate (150 mg, 330 umol), H2O2 (374 mg, 3.30 mmol, 317 uL, 30% purity), NaOH (1 M, 990 uL) in THE (8 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 0° C. for 2 hours under N2 atmosphere. On completion, the reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give the title compound (100 mg, 86% yield) as yellow solid. 1H NMR (400 MHz, CDCl3) δ=12.82 (s, 1H), 7.67-7.45 (m, 1H), 7.08-6.79 (m, 3H), 6.55 (s, 1H), 4.31-3.74 (m, 2H), 3.55 (d, J=7.2 Hz, 2H), 3.41-3.10 (m, 2H), 2.89-2.70 (m, 1H), 2.13-1.92 (m, 1H), 1.82-1.70 (m, 1H), 1.65-1.46 (m, 10H); LC-MS (ESI, m/z): [M+Na]*=367.2.


Step 5: 8-hydroxy-2-(pyrrolidin-3-ylmethyl)isoquinolin-1(2H)-one

A mixture of tert-butyl 3-[(8-hydroxy-1-oxo-2-isoquinolyl)methyl]pyrrolidine-1-carboxylate (100 mg, 290 umol) in TFA (0.2 mL) DCM (1.8 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25° C. for 2 hours under N2 atmosphere. On completion, the reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give the title compound (40.0 mg, 47% yield) as red liquid. 1H NMR (400 MHz, CDCl3) δ=12.86-12.50 (m, 1H), 8.55 (s, 1H), 7.54 (s, 1H), 7.11 (d, J=7.4 Hz, 1H), 6.95 (d, J=17.0 Hz, 2H), 6.56 (d, J=7.4 Hz, 1H), 4.10 (d, J=6.8 Hz, 1H), 4.03 (d, J=8.4 Hz, 1H), 3.52-3.40 (m, 1H), 3.36-3.22 (m, 2H), 3.16 (s, 1H), 3.10-2.96 (m, 1H), 2.27-2.14 (m, 1H), 1.96-1.82 (m, 1H); LC-MS (ESI, m/z): [M+1]+=245.6.


Step 6: 8-hydroxy-2-((1-(pyridin-2-yl)pyrrolidin-3-yl)methyl)isoquinolin-1(2H)-one

A mixture of 8-hydroxy-2-(pyrrolidin-3-ylmethyl)isoquinolin-1-one (20.0 mg, 81.8 umol), 2-fluoropyridine (15.9 mg, 163 umol, 14.0 uL), DBU (74.7 mg, 491 umol, 74.0 uL) in DMSO (1 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 110° C. for 12 hours under N2 atmosphere. On completion, the reaction mixture was purified by prep-HPLC to give the title compound (11.6 mg, 37% yield) as white solid. 1H NMR (400 MHz, CDCl3) δ=12.86 (s, 1H), 8.17 (d, J=5.2 Hz, 1H), 7.53 (s, 1H), 7.49-7.43 (m, 1H), 7.01-6.90 (m, 3H), 6.54 (d, J=7.2 Hz, 2H), 6.39-6.33 (m, 1H), 4.19-4.10 (m, 1H), 4.02-3.93 (m, 1H), 3.72-3.61 (m, 2H), 3.52-3.44 (m, 1H), 3.39-3.32 (m, 1H), 3.01-2.89 (m, 1H), 2.24-2.14 (m, 1H), 1.96-1.86 (m, 1H); LC-MS (ESI, m/z): [M+1]+=322.1.


Example 6. 5-Chloro-1-phenylquinazolin-4(1H)-one (I-11)



embedded image


Step 1: 2-chloro-6-(phenylamino)benzamide

To a solution of aniline (563 mg, 6.05 mmol, 552 uL) in THE (10 mL) at 20° C. was added NaH (345 mg, 8.64 mmol, 60% purity), the reaction mixture was stirred at 20° C. for 0.5 hour. Then a solution of 2-chloro-6-fluorobenzamide (1.00 g, 5.76 mmol) in THF (5 mL) was added to the reaction mixture and stirred at 50° C. for 12 hours. On completion, the reaction mixture was quenched by H2O (10 mL) and filtered and concentrated under reduced pressure to give a residue. The residue was purified by reversed-phase flash (0.1% NH3H2O) to give the title compound (100 mg, 6% yield) as yellow oil. LC-MS (ESI, m/z): [M+1]+=247.1.


Step 2: 5-chloro-1-phenylquinazolin-4(1H)-one

A mixture of 2-anilino-6-chloro-benzamide (50.0 mg, 202 umol), trimethoxymethane (1.38 g, 12.9 mmol, 1.42 mL) and hydrochloric acid (7.39 mg, 202 umol, 7.25 uL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 110° C. for 4 hours under N2 atmosphere. On completion, the reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give the title compound (20.0 mg, 33% yield) as white solid. 1H NMR (400 MHz, CDCl3) δ=8.20 (s, 1H), 7.73-7.60 (m, 3H), 7.52-7.46 (m, 1H), 7.45-7.39 (m, 3H), 7.27 (s, 1H), 6.78 (m, J=1.2, 8.4 Hz, 1H); LC-MS (ESI, m/z): [M+1]+=257.0.




embedded image


Example 7. 5-Chloro-1-(piperidin-4-yl)quinazolin-4(1H)-one (I-12)
Step 1: tert-butyl 4-((2-carbamoyl-3-chlorophenyl)amino)piperidine-1-carboxylate

A mixture of 2-chloro-6-fluoro-benzamide (1.00 g, 5.76 mmol), tert-butyl 4-aminopiperidine-1-carboxylate (1.27 g, 6.34 mmol), DIPEA (2.23 g, 17.2 mmol) in DMSO (10 mL) was degassed and purged with N2 for 3 times and the mixture was stirred at 160° C. for 12 hours under N2 atmosphere. On completion, the mixture was partitioned between H2O (20 ml) and EA (20 mL). The organic phase was separated, washed with brine 20 ml, dried over Na2S04, filtered and concentrated under reduced pressure to give a residue. The residue was purified by pre-HPLC (FA condition) to give the title compound (320 mg, 15% yield) as yellow solid. LC-MS (ESI, m/z): [M+1]+=354.1.


Step 2: tert-butyl 4-(5-chloro-4-oxoquinazolin-1(4H)-yl)piperidine-1-carboxylate

A mixture of tert-butyl 4-(2-carbamoyl-3-chloro-anilino)piperidine-1-carboxylate (50.0 mg, 141 umol), trimethoxymethane (968 mg, 9.12 mmol), hydrochloric acid (5.15 mg, 141 umol, 5.05 uL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100° C. for 2 hours under N2 atmosphere. On completion, the mixture was concentrated in vacuo to give the title compound (40.0 mg, 65% yield) as yellow solid. LC-MS (ESI, m/z): [M+1]+=364.1.


Step 3: 5-chloro-1-(piperidin-4-yl)quinazolin-4(1H)-one

A mixture of tert-butyl 4-(5-chloro-4-oxo-quinazolin-1-yl)piperidine-1-carboxylate (40.0 mg, 109 umol) in TFA (0.1 mL) DCM (0.9 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25° C. for 2 hours under N2 atmosphere. On completion, the mixture was concentrated in vacuo. The residue was purified by prep-HPLC to give the title compound (10.0 mg, 29% yield) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ=8.50 (m, 1H), 8.36 (m, 1H), 7.88 (m, J=8.8 Hz, 1H), 7.75 (m, J=8.0 Hz, 1H), 7.55 (d, J=8.0 Hz, 1H), 4.66 (m, 1H), 3.23 (m, J=6.0 Hz, 2H), 2.91 (m, 2H), 2.17-1.99 (m, 4H); LC-MS (ESI, m/z): [M+1]+=264.2.


Example 8. 2-Hydroxy-N-[3-(4-piperidyl)-1H-indazol-6-yl]benzamide (I-23)



embedded image


embedded image


Step 1: 2-hydroxybenzoyl chloride

A mixture of 2-hydroxybenzoic acid (500 mg, 3.62 mmol), SOCl2 (2.15 g, 18.1 mmol) in DCE (15 mL) was degassed and purged with N2 for 3 times. Then the mixture was stirred at 85° C. for 2 hours under N2 atmosphere. On completion, the reaction mixture was concentrated to give the title compound (400 mg, crude) as a brown solid.


Step 2: tert-butyl 3-bromo-6-nitro-indazole-1-carboxylate

To a solution of 3-bromo-6-nitro-1H-indazole (500 mg, 2.07 mmol), tert-butoxycarbonyl tert-butyl carbonate (1.13 g, 5.16 mmol), TEA (522 mg, 5.16 mmol) in DCM (15 mL) was added DMAP (126 mg, 1.03 mmol). The mixture was stirred at 25° C. for 12 hours. After the reaction mixture was washed twice with 1 M HCl, washed with water and dried over MgSO4, concentrated and purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=15/1 to 5/1) to give the title compound (400 mg, 48% yield) as a white solid. 1H NMR (400 MHz, CD3Cl) δ=9.02 (s, 1H), 8.17 (d, J=8.8 Hz, 1H), 7.72 (d, J=8.8 Hz, 1H), 1.68 (s, 9H).


Step 3: tert-butyl 3-(1-tert-butoxycarbonyl-3,6-dihydro-2H-pyridin-4-yl)-6-nitro-indazole-1-carboxylate

A mixture of tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1-carboxylate (474 mg, 1.53 mmol), tert-butyl 3-bromo-6-nitro-indazole-1-carboxylate (350 mg, 1.02 mmol), Pd(dppf)Cl2.CH2Cl2 (41.8 mg, 51.2 umol) and Cs2CO3 (2 M, 1.02 mL) in dioxane (14 mL) was degassed and purged with N2 for 3 times. Then the mixture was stirred at 80° C. for 12 hours under N2 atmosphere. On completion, the reaction mixture was concentrated and purified by prep-TLC (SiO2, Petroleum ether/Ethyl acetate=3:1) to give the title compound (320 mg, 56% yield) as a yellow solid. LC-MS (ESI, m/z): [M+Na]*=467.2.


Step 4: tert-butyl 6-amino-3-(1-tert-butoxycarbonyl-4-piperidyl)indazole-1-carboxylate

To a solution of tert-butyl 3-(1-tert-butoxycarbonyl-3,6-dihydro-2H-pyridin-4-yl)-6-nitro-indazole-1-carboxylate (320 mg, 720 umol) in EtOH (40 mL) was added Pd/C (10%, 100 mg) under N2 atmosphere. The suspension was degassed and purged with H2 for 5 times. The mixture was stirred under H2 (15 Psi) at 25° C. for 12 hours. The reaction mixture was filtered, concentrated and purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=5/1 to 1/1) to give the title compound (100 mg, 32% yield) as a white solid. LC-MS (ESI, m/z): [M+1]+=417.2.


Step 5: tert-butyl 3-(1-tert-butoxycarbonyl-4-piperidyl)-6-[(2-hydroxybenzoyl)amino]indazole-1-carboxylate

A mixture of tert-butyl 6-amino-3-(1-tert-butoxycarbonyl-4-piperidyl)indazole-1-carboxylate (20 mg, 48.0 umol), 2-hydroxybenzoyl chloride (11.3 mg, 72.0 umol) in DCM (1 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25° C. for 12 hours under N2 atmosphere. The reaction mixture was quenched by H2O (1 mL) at 25° C. and diluted with DCM (5 mL) and extracted with DCM 10 mL (5 mL×2). The combined organic layers were washed with brine (5 mL), dried over Na2SO4, filtered and concentrated to give the title compound (10 mg, crude) as a yellow solid. LC-MS (ESI, m/z): [M+1]+=537.4.


Step 6: 2-hydroxy-N-[3-(4-piperidyl)-1H-indazol-6-yl]benzamide

To a solution of tert-butyl 3-(1-tert-butoxycarbonyl-4-piperidyl)-6-[(2-hydroxybenzoyl)amino]indazole-1-carboxylate (10 mg, 18.6 umol) in DCM (1 mL) was added HCl/dioxane (4 M, 4.66 uL). The mixture was stirred at 25° C. for 0.5 hour. The reaction mixture was concentrated and purified by prep-HPLC (column: Phenomenex Synergi C18 150*25 mm*10 um; mobile phase: [water (0.05% HCl)-ACN]; B %: 6%-36%, 10 min) to give the title compound (2.24 mg, 36% yield) as a brown oil. 1H NMR (400 MHz, DMSO-d6) δ=12.70 (s, 1H), 11.80 (s, 1H), 10.53 (s, 1H), 8.95-8.74 (m, 2H), 8.15 (s, 1H), 8.00 (dd, J=1.2, 6.4 Hz, 1H), 7.81 (d, J=8.4 Hz, 1H), 7.50-7.40 (m, 1H), 7.27 (d J=8.8 Hz, 1H), 7.08-6.94 (m, 2H), 3.42-3.35 (m, 4H), 3.09-3.07 (m, 2H), 2.17-2.09 (m, 4H); LC-MS (ESI, m/z): [M+1]*=337.2.


Example 9. 3-(piperazin-1-yl)-5-(pyrimidin-2-yl)pyridin-2-amine (I-14)



embedded image


Step 1: tert-butyl 4-(5-bromo-2-nitro-3-pyridyl)piperazine-1-carboxylate

A mixture of 5-bromo-3-fluoro-2-nitro-pyridine (250 mg, 1.13 mmol), tert-butyl piperazine-1-carboxylate (421 mg, 2.26 mmol), TEA (229 mg, 2.26 mmol) in DMF (3 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 90° C. for 12 hours under N2 atmosphere. The residue was diluted with water (100 mL) and extracted with ethyl acetate (100 mL×2). The combined organic layers were washed with brine (100 mL), dried over sodium sulfate anhydrous, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 1:1) to give a title compound (500 mg, 97% yield) as a yellow solid. LC-MS (ESI, m/z): [M-55]+=331.0


Step 2: tert-butyl 4-(2-nitro-5-pyrimidin-2-yl-3-pyridyl)piperazine-1-carboxylate

A mixture of tert-butyl 4-(5-bromo-2-nitro-3-pyridyl)piperazine-1-carboxylate (300 mg, 774 umol), tributyl(pyrimidin-2-yl)stannane (572 mg, 1.55 mmol), CuI (7.38 mg, 38.74 umol), Pd(PPh3)4(44.7 mg, 38.7 umol) and LiCl (32.8 mg, 774 umol) in toluene (10 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 110° C. for 12 hr under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 1:1) to give a title compound (200 mg, 66% yield) as a yellow solid. LC-MS (ESI, m/z): [M+1]*=387.2


Step 3: tert-butyl 4-(2-amino-5-pyrimidin-2-yl-3-pyridyl)piperazine-1-carboxylate

To a solution of tert-butyl 4-(2-nitro-5-pyrimidin-2-yl-3-pyridyl)piperazine-1-carboxylate (100 mg, 258 umol) in THE (10 mL) was added PtO2 (29.4 mg, 129 umol) under N2. The suspension was degassed under vacuum and purged with H2 several times. The mixture was stirred under H2 (15 psi) at 25° C. for 1 hour. The reaction mixture was concentrated under reduced pressure to give a title compound (90 mg, crude) as a yellow solid. LC/MS (ESI, m/z): [M+1]+=357.2


Step 4: 3-piperazin-1-yl-5-pyrimidin-2-yl-pyridin-2-amine (SMA-217)

A mixture of tert-butyl 4-(2-amino-5-pyrimidin-2-yl-3-pyridyl)piperazine-1-carboxylate (90 mg, 252 umol) in DCM (10 mL) TFA (1 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25° C. for 2 hr under N2 atmosphere. The residue was diluted with water 10 mL and extracted with ethyl acetate (10 mL×2). The combined organic layers were washed with brine (10 mL), dried over sodium sulphate anhydrous, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Xtimate C18 150*25 mm*5 um; mobile phase: [water (0.05% ammonia hydroxide v/v)-ACN]; B %: 5%-35%, 10 min) to give a title compound (40 mg) as a white solid. 1H NMR (400 MHz, CD3OD) δ=8.87-8.66 (m, 3H), 8.24 (d, J=2.0 Hz, 1H), 7.28 (t, J=4.9 Hz, 1H), 3.06 (d, J=4.0 Hz, 4H), 2.98 (d, J=4.2 Hz, 4H). LC-MS (ESI, m/z): [M+1]*=257.0.


Example 10. 5-(3-chlorophenyl)-3-(piperazin-1-yl)pyridin-2-amine (1-15)



embedded image


Step 1: tert-butyl 4-(5-(3-chlorophenyl)-2-nitropyridin-3-yl)piperazine-1-carboxylate

A mixture of tert-butyl 4-(5-bromo-2-nitro-3-pyridyl)piperazine-1-carboxylate (100 mg, 258 umol), 2-(3-chlorophenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (123 mg, 517 umol), Pd(dppf)Cl2—CH2Cl2 (10.5 mg, 12.9 umol) and Cs2CO3 (2 M in water, 387 uL) in dioxane (3 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 90° C. for 12 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (neutral condition) give the title compound (100 mg, 89% yield) was obtained as a yellow solid. LC-MS (ESI, m/z): [M+1]+=419.2.


Step 2: tert-butyl 4-(2-amino-5-(3-chlorophenyl)pyridin-3-yl)piperazine-1-carboxylate

To a solution of tert-butyl 4-[5-(3-chlorophenyl)-2-nitro-3-pyridyl]piperazine-1-carboxylate (40 mg, 95.49 umol) in THE (1 mL) was added PtO2 (10.8 mg, 47.7 umol) under N2. The suspension was degassed under vacuum and purged with H2 several times. The mixture was stirred under H2 (15 psi) at 25° C. for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give the title compound (30 mg, crude) as a yellow solid. LC-MS (ESI, m/z): [M+1]*=389.2.


Step 3: 5-(3-chlorophenyl)-3-(piperazin-1-yl)pyridin-2-amine

To a solution of tert-butyl 4-[2-amino-5-(3-chlorophenyl)-3-pyridyl] piperazine-1-carboxylate (30 mg, 77.1 umol) in TFA (0.5 mL) and DCM (5 mL) was stirred at 25° C. for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*25*10 um; mobile phase: [water (0.225% FA)-ACN]; B %: 16%-40%, 8 min) give the title compound (20 mg, 77% yield) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ=8.22 (s, 1H), 8.09 (d, J=2.2 Hz, 1H), 7.69 (t, J=1.8 Hz, 1H), 7.59 (d, J=7.8 Hz, 1H), 7.48-7.40 (m, 2H), 7.33 (dd, J=1.1, 7.8 Hz, 1H), 6.03 (s, 2H), 3.27 (d, J=4.6 Hz, 4H), 3.07 (s, 4H); LC-MS (ESI, m/z): [M+1]+=289.1.


Example 11. 2-Hydroxy-N-((1-(pyridin-2-yl)piperidin-4-yl)methyl)benzamide (I-16)



embedded image


Step 1: benzyl ((1-(pyridin-2-yl)piperidin-4-yl)methyl)carbamate

Benzyl N-(4-piperidylmethyl)carbamate (1 g, 4.03 mmol) and DIPEA (560 mg, 4.33 mmol) in 2-fluoropyridine (5.65 g, 58.2 mmol) was heat to reflux at 130° C. and stirred for 12 hours. On completion, evaporated the solvent to give the title compound (1.3 g, crude) as a yellow solid. LC-MS (ESI, m/z): [M+1]+=326.1.


Step 2: (1-(pyridin-2-yl)piperidin-4-yl)methanamine

To a solution of benzyl N-[[1-(2-pyridyl)-4-piperidyl]methyl]carbamate (0.5 g, 1.54 mmol) in MeOH (5 mL) was added Pd/C (50 mg, 1.54 mmol, 10% purity), then the mixture was stirred under H2 atmosphere(15 psi) at 25° C. for 12 hours. On completion, filtered and evaporated the solvent to give the title compound (0.2 g, crude) as a yellow solid. LC-MS (ESI, m/z): [M+1]+=192.2.


Step 3: 2-hydroxy-N-((1-(pyridin-2-yl)piperidin-4-yl)methyl)benzamide (SMA-219)

To a solution of [1-(2-pyridyl)-4-piperidyl]methanamine (0.2 g, 1.05 mmol) and 2-hydroxybenzoic acid (144 mg, 1.05 mmol) in DMF (1 mL) was added Et3N (317 mg, 3.14 mmol) and HATU (795 mg, 2.09 mmol), then the mixture was stirred at 25° C. for 12 hours. On completion, the mixture was send to purify by prep-HPLC(TFA:column: Shim-pack C18 150*25*10 um; mobile phase: [water (0.1% TFA)-ACN]; B %: 13%-43%, 10 min) and prep-HPLC (HCl: column: Phenomenex Synergi C18 150*30 mm*4 um; mobile phase: [water (0.05% HCl)-ACN]; B %: 14%-34%, 11 min) to give the title compound (24 mg, 7% yield) as a white yellow solid. 1H NMR (400 MHz, CD3OD) δ=7.83 (s, 1H), 7.64 (s, 2H), 7.52 (m, 1H), 7.36-7.31 (m, 4H), 7.23-7.21 (m, 2H), 7.00 (m, 1H), 6.83 (d, J=8 Hz, 1H), 6.20 (d, J=5.6 Hz, 1H), 5.31 (s, 2H), 5.06 (s, 2H), LC-MS (ESI, m/z): [M+1]+=312.1.


Example 12. 3-(6-Amino-5-(piperazin-1-yl)pyridazin-3-yl)pyridin-4-ol (I-17)



embedded image


embedded image


Step 1: tert-butyl 4-(3-amino-6-chloro-pyridazin-4-yl)piperazine-1-carboxylate

To a solution of 4-bromo-6-chloro-pyridazin-3-amine (2 g, 9.59 mmol) in DMSO (40 mL) was added tert-butyl piperazine-1-carboxylate (8.94 g, 47.9 mmol). Then the mixture was stirred at 80° C. for 12 hours. The reaction mixture was diluted with ethyl acetate (50 mL) and H2O (50 mL) and then filtered. The filter cake was dried under the reduced pressure to give the title compound (3 g, crude) as a white solid. LC-MS (ESI, m/z): [M+1]+=314.1


Step 2: (4-methoxypyridin-3-yl)boronic acid

To a solution of 3-bromo-4-methoxy-pyridine (1 g, 5.32 mmol) in dioxane (6 mL) was added 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (2.70 g, 10.6 mmol), Pd(dppf)Cl2 (389 mg, 531 umol) and KOAc (1.57 g, 15.9 mmol). Then the mixture was stirred at 110° C. for 2 hours. The reaction mixture was filtered and concentrated to give the title compound (1 g, crude) as a black solid. LC-MS (ESI, m/z): [M+1]+=154.0


Step 3: tert-butyl 4-[3-amino-6-(4-methoxy-3-pyridyl)pyridazin-4-yl]piperazine-1-carboxylate

To a solution 4-methoxy-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (149 mg, 637 umol) in dioxane (5 mL) was added tert-butyl 4-(3-amino-6-chloro-pyridazin-4-yl)piperazine-1-carboxylate (200 mg, 637 umol), Cs2CO3 (2 M, 956 uL) and BrettPhos Pd G3 (57.7 mg, 63.7 umol). Then the mixture was stirred at 100° C. for 12 hours. The reaction mixture was filtered and purified directly. The residue was purified by reversed phase flash (NH3.H2O) to give the title compound (150 mg, 60% yield) as a brown solid. LC-MS (ESI, m/z): [M+1]+=387.2


Step 4: 3-(6-amino-5-piperazin-1-yl-pyridazin-3-yl)pyridin-4-ol

To a solution of tert-butyl 4-[3-amino-6-(4-methoxy-3-pyridyl)pyridazin-4-yl]piperazine-1-carboxylate (70 mg, 181 umol) in MeCN (3 mL) was added TMSI (181 mg, 905 umol). Then the mixture was stirred at 80° C. for 12 hours. The reaction mixture was filtered and concentrated. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25 mm*5 um; mobile phase: [water (0.05% ammonia hydroxide v/v)-ACN]; B %: 1%-20%, 10 min) to give the title compound (20 mg, 2% yield) as a yellow. LC-MS (ESI, m/z): [M+1]+=273.1


Step 5: tert-butyl 4-[3-amino-6-(4-hydroxy-3-pyridyl)pyridazin-4-yl]piperazine-1-carboxylate

To a solution of 3-(6-amino-5-piperazin-1-yl-pyridazin-3-yl)pyridin-4-ol (20 mg, 73.4 umol) in DMF (2 mL) was added Boc2O (48.0 mg, 220 umol) and TEA (22.3 mg, 220 umol) DMAP (4.49 mg, 36.7 umol). The mixture was stirred at 25° C. for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (TFA condition) to give the title compound (5 mg, 17% yield) as a white solid. LC-MS (ESI, m/z): [M+1-Boc]+=373.0


Step 6: 3-(6-amino-5-piperazin-1-yl-pyridazin-3-yl)pyridin-4-ol (SMA-244)

To a solution of tert-butyl 4-[3-amino-6-(4-hydroxy-3-pyridyl)pyridazin-4-yl]piperazine-1-carboxylate (5 mg, 13.4 umol) in TFA (0.1 mL) DCM (1 mL) was stirred at 25° C. for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*25*10 um; mobile phase: [water (0.05% HCl)-ACN]; B %: 0%-10%, 10 min) to give the title compound (2 mg, 46% yield) as a white solid. 1H NMR (400 MHz, CD3OD) δ=9.01 (s, 1H), 8.46 (d, J=6.8 Hz, 1H), 7.96 (s, 1H), 7.31 (d, J=7.0 Hz, 1H), 3.57 (s, 8H). LC-MS (ESI, m/z): [M+1]+=273.0.


Example 13. 2-(6-Amino-5-(4-(4-butoxyphenyl)piperazin-1-yl)pyridazin-3-yl)phenol (I-18)
Step 1: tert-butyl 4-(4-butoxyphenyl)piperazine-1-carboxylate



embedded image


To a solution of tert-butyl 4-(4-hydroxyphenyl)piperazine-1-carboxylate (0.5 g, 1.80 mmol) and 1-bromobutane (0.25 g, 1.80 mmol) in DMF (20 mL) was added K2CO3 (0.32 g, 2.34 mmol), then the reaction mixture was stirred at 100° C. for 12 hours. The reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (2×20 mL). The combined organic layers were washed with brine (2×30 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give the title compound (0.6 g, crude) as a white solid. LC-MS (ESI, m/z): [M+1]+=335.3.


Step 2: 1-(4-butoxyphenyl)piperazine

To a solution of tert-butyl 4-(4-butoxyphenyl)piperazine-1-carboxylate (0.6 g, 1.79 mmol) in DCM (6 mL) was added HCl/dioxane (4 M, 4.48 mL), then the reaction mixture was stirred at 25° C. for 12 hours. The reaction mixture was concentrated under reduced pressure to give the title compound (0.48 g, crude) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 9.79-9.77 (m, 1H), 7.18 (d, J=7.6 Hz, 2H), 6.95-6.92 (m, 2H), 3.93 (t, J=6.4 Hz, 2H), 3.46 (s, 4H), 3.34 (s, 4H), 1.70-1.63 (m, 2H), 1.44-1.39 (m, 2H), 0.92 (t, J=7.6 Hz, 3H). LC-MS (ESI, m/z): [M+1]+=235.1.


Step 3: 4-(4-(4-butoxyphenyl)piperazin-1-yl)-6-chloropyridazin-3-amine

To a solution of 1-(4-butoxyphenyl)piperazine (0.48 g, 1.77 mmol, HCl) in DMSO (10 mL) was added DIEA (1.14 g, 8.85 mmol) and 4-bromo-6-chloro-pyridazin-3-amine (0.44 g, 2.12 mmol), then the reaction mixture was stirred at 100° C. for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Synergi Max-RP 250*50 mm*10 um; mobile phase: [water (0.1% TFA)-ACN]; B %: 30ACN %-60ACN %, 15 min) to give the title compound (0.5 g, 77% yield for three steps) as a off-white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 7.30 (s, 1H), 7.09 (d, J=8.8 Hz, 2H), 6.93-6.90 (m, 2H), 3.96-3.87 (m, 2H), 3.34-3.30 (m, 8H), 1.70-1.65 (m, 2H), 1.45-1.39 (m, 2H), 0.94-0.90 (m, 3H). LC-MS (ESI, m/z): [M+1]+=362.0.


Step 4: 2-(6-amino-5-(4-(4-butoxyphenyl)piperazin-1-yl)pyridazin-3-yl)phenol (SMA-252-001F)

A mixture of 4-[4-(4-butoxyphenyl)piperazin-1-yl]-6-chloro-pyridazin-3-amine (0.3 g, 0.83 mmol), (2-hydroxyphenyl)boronic acid (0.23 g, 1.66 mmol), BrettPhos Pd G3 (0.08 g, 0.083 mmol) and K2CO3 (0.34 g, 2.49 mmol) in dioxane (5 mL) and water (1 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 85° C. for 12 h under N2 atmosphere. The reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (2×20 mL). The combined organic layers were washed with brine (2×30 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified purified by reversed-phase HPLC (0.1% FA condition), then purified by prep-HPLC (column: Phenomenex Gemini 150*25 mm*10 um; mobile phase: [water (0.225% FA)-ACN]; B %: 30%-63%, 10 min) to give the title compound (14 mg, 4% yield) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ=14.28-14.16 (m, 1H), 7.94 (d, J=6.8 Hz, 1H), 7.57 (s, 1H), 7.27 (s, 1H), 6.96-6.84 (m, 6H), 6.33 (s, 2H), 3.90 (t, J=6.8 Hz, 2H), 3.25 (s, 8H), 1.68-1.63 (m, 2H), 1.45-1.40 (m, 2H), 0.93 (t, J=7.2 Hz, 3H); LC-MS (ESI, m/z): [M+1]+=420.2.




embedded image


Example 14. 2-(7H-Pyrrolo[2,3-c]pyridazin-3-yl)phenol (I-19)
Step 1: 6-chloro-4-((trimethylsilyl)ethynyl)pyridazin-3-amine

To a mixture of 4-bromo-6-chloro-pyridazin-3-amine (200 mg, 959 umol), CuI (5.48 mg, 28.7 umol), Pd(PPh3)2Cl2 (60.6 mg, 86.3 umol) and TEA (194 mg, 1.92 mmol) in DMF (4 mL) was added trimethylsilylacetylene (141 mg, 1.44 mmol) under nitrogen. The reaction was stirred at 100° C. for 5 hours. On completed, the mixture was washed with water (40 mL), extracted with EA (2×20 mL). The organic layer was dried with Na2SO4, filtrated and concentrated in vacuo to give a residue. The residue was purified by silica gel chromatography (PE:EA=4:1) to give the title compound (200 mg, 92% yield) as an brown solid. 1H NMR (400 MHz, DMSO-d6) δ=7.56 (s, 1H), 6.80 (s, 2H), 0.26 (s, 9H); LC-MS (ESI, m/z): [M+1]*=226.0& 228.0.


Step 2: 3-chloro-7H-pyrrolo[2,3-c]pyridazine

To a mixture of 6-chloro-4-(2-trimethylsilylethynyl)pyridazin-3-amine (220 mg, 974 umol), t-BuOK (185 mg, 1.66 mmol) and N-methylmethanamine; hydrochloride (79.4 mg, 974 umol) in DMF (4 mL) was stirred at 25° C. for 0.5 hour. Then the mixture was stirred at 120° C. for 2 hours. On completion, the mixture was quenched by aqueous NH4Cl (20 mL) and filtrated. The filtrate was washed by brine and dried over by Na2SO4, then filtrated and the filtrate was concentrated in vacuo. The residue was purified by silica gel chromatography (PE:EA=1:1) to give the title compound (70.0 mg, 46% yield) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) S=12.61 (s, 1H), 8.13-7.83 (m, 2H), 6.54 (d, J=4.0 Hz, 1H); LC-MS (ESI, m/z): [M+1]+=154.1 & 156.1.


Step 3: 2-(7H-pyrrolo[2,3-c]pyridazin-3-yl)phenol

A mixture of 3-chloro-7H-pyrrolo[2,3-c]pyridazine (70.0 mg, 455 umol), 2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenol (200 mg, 911 umol), K2CO3 (188 mg, 1.37 mmol) and [2-(2-aminophenyl)phenyl]-methylsulfonyloxy-palladium; dicyclohexyl-[3,6-dimethoxy-2-(2,4,6-triisopropylphenyl)phenyl]phosphane (41.3 mg, 45.5 umol) in dioxane (2 mL) and H2O (1 mL) was stirred at 100° C. for 12 hours. On completion, the mixture was filtrated and the filtrate was concentrated in vacuo. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25*5 um; mobile phase: [water (10 mm NH4HCO3)-ACN]; B %: 20%-50%, 10 min) to give the title compound (20.0 mg, 20% yield) as a brown solid. 1H NMR (400 MHz, DMSO-d6) δ=8.67 (s, 1H), 8.08-8.00 (m, 2H), 7.33-7.25 (m, 1H), 7.01-6.91 (m, 2H), 6.66 (d, J=3.2 Hz, 1H); LC-MS (ESI, m/z): [M+1]*=212.0.


Example 15. 2-(5-(Piperidin-4-yl)-7H-pyrrolo[2,3-c]pyridazin-3-yl)phenol (I-20)



embedded image


Step 1: 5-bromo-3-chloro-7H-pyrrolo[2,3-c]pyridazine

To a solution of 3-chloro-7H-pyrrolo[2,3-c]pyridazine (0.4 g, 2.60 mmol) in DMF (10 mL) was added NBS (556 mg, 3.13 mmol). The mixture was stirred at 25° C. for 12 hours. The reaction mixture was diluted with H2O (50 mL) and filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=5/1 to 1/1) to get the title compound (0.45 g, 70% yield) as a red solid. LC-MS (ESI, m/z): [M+1]+=233.9.


Step 2: 5-bromo-3-chloro-7-tosyl-7H-pyrrolo[2,3-c]pyridazine

5-bromo-3-chloro-7H-pyrrolo[2,3-c]pyridazine (0.3 g, 1.29 mmol) in DMF (10 mL) was added NaH (154 mg, 3.87 mmol, 60% purity) at 0° C. The reaction mixture was stirred at 0° C. for 0.5 hour before TosCl (492 mg, 2.58 mmol) was added and the resulting mixture solution was stirred at 0° C. for additional 1 hour. The reaction was quenched with saturated ammonia chloride aqueous solution and extracted with EtOAc (10 mL, three times). The combined organics were washed with brine dried over anhydrous Na2SO4 and concentrated in vacuo to give a crude product. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 1/1) to get the title compound (300 mg, 57% yield) as a yellow solid. LC-MS (ESI, m/z): [M+1]+=387.9.


Step 3: tert-butyl 4-(3-chloro-7-tosyl-7H-pyrrolo[2,3-c]pyridazin-5-yl)-5,6-dihydropyridine-1(2H)-carboxylate

A mixture of 5-bromo-3-chloro-7-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridazine (300 mg, 775 umol), tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1-carboxylate (215 mg, 698 umol), Pd(dppf)Cl2—CH2Cl2 (63.3 mg, 77.6 umol), Cs2CO3 (2 M, 776 uL) in dioxane (6 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 85° C. for 4 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 3/1) to get the title compound (190 mg, 49% yield) as a yellow solid. LC-MS (ESI, m/z): [M+1]+=489.4.


Step 4: tert-butyl 4-(3-(2-hydroxyphenyl)-7-tosyl-7H-pyrrolo[2,3-c]pyridazin-5-yl)-5,6-dihydropyridine-1(2H)-carboxylate

A mixture of tert-butyl 4-[3-chloro-7-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridazin-5-yl]—3,6-dihydro-2H-pyridine-1-carboxylate (0.1 g, 204 umol), (2-hydroxyphenyl)boronic acid (84.6 mg, 613 umol), K2CO3 (2 M, 409 uL), BrettPhos Pd G3 (18.5 mg, 20.4 umol) in dioxane (4 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 85° C. for 12 hours under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*25*10 um; mobile phase: [water (0.10% TFA)-ACN]; B %: 40%-60%, 10 min) to get the title compound (20 mg, 48.41 umol, 24% yield) as a yellow solid. LC-MS (ESI, m/z): [M+1]+=393.0.


Step 5: tert-butyl 4-(3-(2-hydroxyphenyl)-7-tosyl-7H-pyrrolo[2,3-c]pyridazin-5-yl)piperidine-1-carboxylate

To a solution of tert-butyl 4-[3-(2-hydroxyphenyl)-7H-pyrrolo[2,3-c]pyridazin-5-yl]—3,6-dihydro-2H-pyridine-1-carboxylate (20 mg, 50.9 umol) in EtOH (10 mL) was added Pd/C (20 mg, 50.9 umol, 10% purity) under N2. The suspension was degassed under vacuum and purged with H2 several times. The mixture was stirred under H2 (15 psi) at 25° C. for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give the title compound (20 mg, crude) as a yellow solid. LC-MS (ESI, m/z): [M+1]+=395.5.


Step 6: 2-(5-(piperidin-4-yl)-7H-pyrrolo[2,3-c]pyridazin-3-yl)phenol

To a solution of tert-butyl 4-[3-(2-hydroxyphenyl)-7H-pyrrolo [2, 3-c] pyridazin-5-yl]piperidine-1-carboxylate (20 mg, 50.7 umol) in dioxane (2 mL) was added HCl/dioxane (4 M, 2 mL). The mixture was stirred at 25° C. for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*25*10 um; mobile phase: [water (0.05% HCl)-ACN]; B %: 0%-20%, 12 min) to get the title compound (5 mg, 25% yield) as a yellow solid. 1H NMR (400 MHz, CD3OD) δ=8.86 (s, 1H), 8.42 (s, 1H), 7.73 (dd, J=1.3, 7.7 Hz, 1H), 7.59-7.42 (m, 1H), 7.22-7.07 (m, 2H), 3.58 (d, J=12.8 Hz, 2H), 3.52-3.40 (m, 1H), 3.31-3.23 (m, 2H), 2.35 (d, J=13.7 Hz, 2H), 2.19-2.05 (m, 2H); LC-MS (ESI, m/z): [M+1]+=294.9.


Example 16. 2-Hydroxy-N-(1H-indazol-6-yl)benzamide (I-21)



embedded image


Step 1: pent-4-ynyl 4-methylbenzenesulfonate

A mixture of 2-hydroxybenzoic acid (0.30 g, 2.17 mmol), 1H-indazol-6-amine (347 mg, 2.61 mmol), DIPEA (561 mg, 4.34 mmol), HATU (1.24 g, 3.26 mmol) in DMF (3 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25° C. for 12 hours under N2 atmosphere. On completion, the reaction mixture was partitioned between H2O (10 mL) and Ethyl acetate (100 mL). The organic phase was separated, washed with brine 30 mL (15 mL×2). The extracts was dried over Na2SO4, filtered and concentrated to get the crude residue. The crude residue was purified by prep-HPLC (column: Waters Xbridge 150*25 5 um; mobile phase: [water (10 mM NH4HCO3)-ACN]; B %: 15%-45%, 10 min) to give the title compound (50 mg, 9% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ=12.98 (s, 1H), 11.80 (s, 1H), 10.62 (s, 1H), 8.22 (s, 1H), 8.04-7.95 (m, 2H), 7.73 (d, J=8.4 Hz, 1H), 7.44 (t, J=8.0 Hz, 1H), 7.26 (d, J=8.8 Hz, 1H), 7.04-6.91 (m, 2H); LC-MS (ESI, m/z): [M+1]+=254.1.


Example 17. 2-Hydroxy-N-(6-(piperidin-4-yl)-1H-indazol-4-yl)benzamide (I-22)



embedded image


Step 1: 2-hydroxybenzoyl chloride

A mixture of 2-hydroxybenzoic acid (200 mg, 1.45 mmol), thionyl chloride(861 mg, 7.24 mmol) in 1,2-dichloro-ethane (4 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 85° C. for 2 hours under N2 atmosphere. On completion, the reaction mixture was filtered and concentrated under reduced pressure to give the crude title compound (260 mg, crude). LC-MS (ESI, m/z): [M+1]+=153.0.


Step 2: tert-butyl 4-(4-nitro-1H-indazol-6-yl)-5,6-dihydropyridine-1(2H)-carboxylate

A mixture of 4-bromo-6-nitro-1H-indazole (500 mg, 2.07 mmol), tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1-carboxylate (1.92 μg, 6.20 mmol), Cs2CO3 (2.02 g, 6.20 mmol), BrettPhos Pd G3 (187 mg, 206 umol) in dioxane (20 mL) and H2O (4 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 110° C. for 36 hours under N2 atmosphere. On completion, the reaction mixture was partitioned between H2O (10 mL) and EA (10 mL). The organic phase was separated, washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=20/1 to 1/1) to give the title compound (360 mg, 42% yield) as a black solid. LC-MS (ESI, m/z): [M+1]*=345.1.


Step 3: tert-butyl 6-(1-(tert-butoxycarbonyl)-1,2,3,6-tetrahydropyridin-4-yl)-4-nitro-1H-indazole-1-carboxylate

A mixture of tert-butyl 4-(6-nitro-1H-indazol-4-yl)-3,6-dihydro-2H-pyridine-1-carboxylate (340 mg, 987 umol), Boc2O (430 mg, 1.97 mmol, 453 uL), TEA (279 mg, 2.76 mmol, 384 uL) in THE (5 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25° C. for 12 hours under N2 atmosphere. On completion, the reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=30/1 to 5/1) to give the title compound (170 mg, 39% yield) as a yellow solid. LC-MS (ESI, m/z): [M+Na]=467.2.


Step 4: tert-butyl 4-amino-6-(1-(tert-butoxycarbonyl)piperidin-4-yl)-1H-indazole-1-carboxylate

To a solution of tert-butyl 4-(1-tert-butoxycarbonyl-3,6-dihydro-2H-pyridin-4-yl)-6-nitro-indazole-1-carboxylate (170 mg, 382 umol) in EtOH (4 mL) was added Pd/C (80.3 mg, 76.4 umol, 10% purity) under N2 atmosphere. The suspension was degassed and purged with H2 for 5 times. The mixture was stirred under H2 (15 Psi) at 25° C. for 12 hours. On completion, the reaction mixture was filtered and concentrated in vacuo to give the title compound (120 mg, 54% yield) as white solid. LC-MS (ESI, m/z): [M+Na]=439.2.


Step 5: tert-butyl 6-(1-(tert-butoxycarbonyl)piperidin-4-yl)-4-(2-hydroxybenzamido)-1H-indazole-1-carboxylate

A mixture of tert-butyl 6-amino-4-(1-tert-butoxycarbonyl-4-piperidyl)indazole-1-carboxylate (60.0 mg, 144 umol), 2-hydroxybenzoyl chloride (33.8 mg, 216 umol) in DCM (1 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25° C. for 12 hours under N2 atmosphere. On completion, the reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC to give the title compound (15.0 mg, 19% yield) as a white solid. LC-MS (ESI, m/z): [M+1]*=537.2.


Step 6: 2-hydroxy-N-(6-(piperidin-4-yl)-1H-indazol-4-yl)benzamide

A mixture of tert-butyl 4-(1-tert-butoxycarbonyl-4-piperidyl)-6-[(2-hydroxybenzoyl)amino]indazole-1-carboxylate (15.0 mg, 27.9 umol) in TFA (0.2 mL) DCM (1 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25° C. for 2 hours under N2 atmosphere. On completion, the reaction mixture was filtered and the filtrate was concentrated in vacuo. The residue was purified by prep-HPLC to give the title compound (4.00 mg, 38% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ=11.99 (d, J=2.8 Hz, 1H), 10.86 (s, 1H), 9.26-9.00 (m, 1H), 8.99-8.76 (m, 1H), 9.00-8.76 (m, 1H), 8.08-8.00 (m, 2H), 7.76 (s, 1H), 7.50-7.43 (m, 1H), 7.16-7.09 (m, 2H), 7.05-6.98 (m, 1H), 3.39 (d, J=12.4 Hz, 3H), 3.11-2.94 (m, 4H), 2.04-1.92 (m, 5H); LC-MS (ESI, m/z): [M+1]*=337.1.




embedded image


Example 18. 2-Hydroxy-N-[3-(4-piperidyl)-1H-indazol-6-yl]benzamide (I-23)
Step 1: 2-hydroxybenzoyl chloride

A mixture of 2-hydroxybenzoic acid (500 mg, 3.62 mmol), SOCl2 (2.15 g, 18.1 mmol) in DCE (15 mL) was degassed and purged with N2 for 3 times. Then the mixture was stirred at 85° C. for 2 hours under N2 atmosphere. On completion, the reaction mixture was concentrated to give the title compound (400 mg, crude) as a brown solid.


Step 2: tert-butyl 3-bromo-6-nitro-indazole-1-carboxylate

To a solution of 3-bromo-6-nitro-1H-indazole (500 mg, 2.07 mmol), tert-butoxycarbonyl tert-butyl carbonate (1.13 g, 5.16 mmol), TEA (522 mg, 5.16 mmol) in DCM (15 mL) was added DMAP (126 mg, 1.03 mmol). The mixture was stirred at 25° C. for 12 hours. After the reaction mixture was washed twice with 1 M HCl, washed with water and dried over MgSO4, concentrated and purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=15/1 to 5/1) to give the title compound (400 mg, 48% yield) as a white solid. 1H NMR (400 MHz, CD3Cl) δ=9.02 (s, 1H), 8.17 (d, J=8.8 Hz, 1H), 7.72 (d, J=8.8 Hz, 1H), 1.68 (s, 9H).


Step 3: tert-butyl 3-(1-tert-butoxycarbonyl-3,6-dihydro-2H-pyridin-4-yl)-6-nitro-indazole-1-carboxylate

A mixture of tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1-carboxylate (474 mg, 1.53 mmol), tert-butyl 3-bromo-6-nitro-indazole-1-carboxylate (350 mg, 1.02 mmol), Pd(dppf)Cl2.CH2Cl2 (41.8 mg, 51.2 umol) and Cs2CO3 (2 M, 1.02 mL) in dioxane (14 mL) was degassed and purged with N2 for 3 times. Then the mixture was stirred at 80° C. for 12 hours under N2 atmosphere. On completion, the reaction mixture was concentrated and purified by prep-TLC (SiO2, Petroleum ether/Ethyl acetate=3:1) to give the title compound (320 mg, 56% yield) as a yellow solid. LC-MS (ESI, m/z): [M+Na]*=467.2.


Step 4: tert-butyl6-amino-3-(1-tert-butoxycarbonyl-4-piperidyl)indazole-1-carboxylate

To a solution of tert-butyl 3-(1-tert-butoxycarbonyl-3,6-dihydro-2H-pyridin-4-yl)-6-nitro-indazole-1-carboxylate (320 mg, 720 umol) in EtOH (40 mL) was added Pd/C (10%, 100 mg) under N2 atmosphere. The suspension was degassed and purged with H2 for 5 times. The mixture was stirred under H2 (15 Psi) at 25° C. for 12 hours. The reaction mixture was filtered, concentrated and purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=5/1 to 1/1) to give the title compound (100 mg, 32% yield) as a white solid. LC-MS (ESI, m/z): [M+1]+=417.2.


Step 5: tert-butyl3-(1-tert-butoxycarbonyl-4-piperidyl)-6-[(2-hydroxybenzoyl)aminol indazole-1-carboxylate

A mixture of tert-butyl 6-amino-3-(1-tert-butoxycarbonyl-4-piperidyl)indazole-1-carboxylate (20 mg, 48.0 umol), 2-hydroxybenzoyl chloride (11.3 mg, 72.0 umol) in DCM (1 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25° C. for 12 hours under N2 atmosphere. The reaction mixture was quenched by H2O (1 mL) at 25° C. and diluted with DCM (5 mL) and extracted with DCM 10 mL (5 mL×2). The combined organic layers were washed with brine (5 mL), dried over Na2SO4, filtered and concentrated to give the title compound (10 mg, crude) as a yellow solid. LC-MS (ESI, m/z): [M+1]+=537.4.


Step 6: 2-hydroxy-N-[3-(4-piperidyl)-1H-indazol-6-yl]benzamide

To a solution of tert-butyl 3-(1-tert-butoxycarbonyl-4-piperidyl)-6-[(2-hydroxybenzoyl)amino]indazole-1-carboxylate (10 mg, 18.6 umol) in DCM (1 mL) was added HCl/dioxane (4 M, 4.66 uL). The mixture was stirred at 25° C. for 0.5 hour. The reaction mixture was concentrated and purified by prep-HPLC (column: Phenomenex Synergi C18 150*25 mm*10 um; mobile phase: [water (0.05% HCl)-ACN]; B %: 6%-36%, 10 min) to give the title compound (2.24 mg, 36% yield) as a brown oil. 1H NMR (400 MHz, DMSO-d6) δ=12.70 (s, 1H), 11.80 (s, 1H), 10.53 (s, 1H), 8.95-8.74 (m, 2H), 8.15 (s, 1H), 8.00 (dd, J=1.2, 6.4 Hz, 1H), 7.81 (d, J=8.4 Hz, 1H), 7.50-7.40 (m, 1H), 7.27 (d J=8.8 Hz, 1H), 7.08-6.94 (m, 2H), 3.42-3.35 (m, 4H), 3.09-3.07 (m, 2H), 2.17-2.09 (m, 4H); LC-MS (ESI, m/z): [M+1]+=337.2.


Example 19. 2-(4-Piperidyl)-3H-pyrrolo [2, 3-c]cinnoline (I-24)



embedded image


Step 1: 4-iodocinnolin-3-amine

A mixture of cinnolin-3-amine (50 mg, 344 umol), NIS (77.4 mg, 344 umol) in DMF (2.0 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25° C. for 12 hours under N2 atmosphere. The reaction mixture was diluted with brine (30 mL) and extracted with EA (30 mL×3). The combined organic layers were washed with brine (30 mL×3), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. Then the residue was purified by column chromatography (SiO2, DCM/MeOH=40/1 to 20/1) to give the title compound (90 mg, 86% yield) as a yellow solid. LC-MS (ESI+) m/z 271.9 (M+H)+.


Step 2: Tert-butyl 4-(3H-pyrrolo[2,3-c]cinnolin-2-yl)piperidine-1-carboxylate

A mixture of 4-iodocinnolin-3-amine (90 μmg, 332 umol), tert-butyl 4-ethynylpiperidine-1-carboxylate (69.4 mg, 332 umol), CuI (12.6 mg, 66.4 umol), Pd(PPh3)2Cl2 (23.3 mg, 33.2 umol), CsF (201 mg, 1.33 mmol) and 4A molecular sieve (30 mg) in DMSO (3 mL) was degassed and purged with N2 for 3 times. Then the mixture was stirred at 50° C. for 2 hrs under N2 atmosphere. The reaction mixture was diluted with brine (30 mL) and extracted with EA (30 mL×3). The combined organic layers were washed with brine 90 mL (30 mL×3), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. Then the residue was purified by column chromatography (SiO2, DCM/MeOH=40/1 to 20/1) to give the title compound (90 mg, 71% yield) as a brown solid. 1H NMR (400 MHz, DMSO-d6) δ=12.76 (d, J=1.2 Hz, 1H), 8.44-8.37 (m, 1H), 8.32 (d, J=8.0 Hz, 1H), 7.80-7.75 (m, 1H), 7.74-7.68 (m, 1H), 4.14-4.05 (m, 2H), 3.17-3.06 (m, 1H), 3.04-2.83 (m, 2H), 2.13-2.03 (m, 2H), 1.76-1.62 (m, 2H), 1.43 (s, 9H); LC-MS (ESI, m/z): [M+1]*=353.2.


Step 3: 2-(4-piperidyl)-3H-pyrrolo[2,3-c]cinnoline

A mixture of tert-butyl 4-(3H-pyrrolo[2,3-c]cinnolin-2-yl)piperidine-1-carboxylate (44 mg, 124 umol), TFA (3.08 g, 27.01 mmol) in DCM (10 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25° C. for 12 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. Then the residue was purified by prep-HPLC (column: Waters X bridge 150*25 mm*5 um; mobile phase: [water (10 mm NH4HCO3)-ACN]; B %: 1%-30%, 10 min) to give the title compound (13.3 mg, 42% yield) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ=12.96-12.49 (m, 1H), 8.39 (d, J=8.4 Hz, 1H), 8.33 (d, J=7.6 Hz, 1H), 7.79-7.74 (m, 1H), 7.73-7.67 (m, 1H), 6.99-6.92 (m, 1H), 3.13-3.02 (m, 2H), 3.02-2.92 (m, 1H), 2.71-2.58 (m, 2H), 2.11-1.94 (m, 2H), 1.67 (dq, J=3.2, 12.0 Hz, 2H); LC-MS (ESI, m/z): [M+1]+=253.0.




embedded image


Example 20. 4-(Piperidin-4-ylethynyl)isoquinolin-3-amine (I-25)
Step 1: 4-iodoisoquinolin-3-amine

To a solution of isoquinolin-3-amine (100 mg, 694 umol) in DCM (2 mL) and EtOH (1 mL) was adde NIS (125 mg, 555 umol). Then the mixture was stirred at 0-25° C. for 2 hours. On completion, the residue was diluted with ethyl acetate (30 mL) and extracted with water (20 mL). The combined organic layers were washed with drine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=5:1) to give the title compound (80 mg, 38% yield) as a yellow solid. LC-MS (ESI, m/z): [M+1]+=271.0.


Step 2: tert-butyl 4-[2-(3-amino-4-isoquinolyl)ethynyl]piperidine-1-carboxylate

To a solution of 4-iodoisoquinolin-3-amine (80 mg, 296 umol) in DMSO (4 mL) was added CuI (11.3 mg, 59.25 umol), 4A molecular sieve (30 mg), CsF (178 mg, 1.18 mmol), Pd(PPh3)2Cl2 (20.8 mg, 29.6 umol) and tert-butyl 4-ethynylpiperidine-1-carboxylate (125 mg, 592 umol), then the mixture was stirred at 85° C. for 12 hours. On completion, the residue was diluted with ethyl acetate (30 mL) and extracted with water (20 mL). The combined organic layers were washed with drine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue, The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=4:1) to give the title compound (40 mg, 27% yield, 70% purity) as a yellow oil. LC-MS (ESI, m/z): [M+1]+=352.3.


Step 3: 4-[2-(4-piperidyl)ethynyl]isoquinolin-3-amine

To a solution of tert-butyl 4-[2-(3-amino-4-isoquinolyl)ethynyl]piperidine-1-carboxylate (40 mg, 79.7 umol) in DCM (1 mL) was added HCl/dioxane (4 M, 700 uL), then the mixture was stirred at 25° C. for 12 hours. On completion, the reaction mixture was concentrated under reduced pressure to remove solvent DCM. The residue was diluted with DMF and water, the residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*25*10 um; mobile phase: [water (0.05% HCl)-ACN]; B %: 7%-27%, 11 min) to give the title compound (8.65 mg, 43% yield) as a brown gum. 1H NMR (400 MHz, DMSO-d6) δ=9.01-9.36 (m, 3H) 8.02 (dd, J=8.00, 4.80 Hz, 1H) 7.84-7.90 (m, 1H) 7.72-7.83 (m, 1H) 7.33-7.46 (m, 1H) 3.16-3.31 (m, 3H) 3.07 (br dd, J=7.20, 2.80 Hz, 2H) 2.12-2.24 (m, 2H) 1.90-2.04 (m, 2H); LC-MS (ESI, m/z): [M+1]*=252.5.


Example 21. 8-Chloro-4-(piperidin-4-ylethynyl)isoquinolin-3-amine (I-27)



embedded image


Step 1: methyl 2,2-diethoxyethanimidate

To a solution of 2,2-diethoxyacetonitrile (11.0 g, 85.2 mmol) in MeOH (30 mL) was added NaOMe (460 mg) and then the mixture was stirred at 25° C. for 3 hours. On completion, the reaction mixture was quenched by addition dry ice. The reaction mixture was concentrated under reduced pressure to remove methanol and then the residue was diluted with ethyl acetate (40 mL) and filtered and concentrated under reduced pressure to give the title compound (12 g, crude) as a yellow oil. 1H NMR (400 MHz, DMSO-d6) δ=7.87-8.04 (m, 1H), 4.67-4.90 (m, 1H), 3.65-3.69 (m, 3H), 3.52 (q, J=7.2 Hz, 4H), 1.15 (t, J=7.2 Hz, 6H).


Step 2: N-[(2-chlorophenyl)methyl]-2,2-diethoxy-acetamidine

To a solution of (2-chlorophenyl)methanamine (3 g, 21.2 mmol) in MeOH (30 mL) was added methyl 2,2-diethoxyethanimidate (4.44 g, 27.5 mmol), then the mixture was stirred at 70° C. for 12 hours. On completion, the reaction mixture was concentrated under reduced pressure to remove methanol. The residue was diluted with ethyl acetate (100 mL) and extracted with water (80 mL). The combined organic layers were washed with brine (80 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, DCM: MeOH=50:1) to give the title compound (4 g, 51% yield, 73% purity) as a yellow oil. LC-MS (ESI, m/z): [M+1]+=271.1


Step 3: 8-chloroisoquinolin-3-amine

To a solution of N-[(2-chlorophenyl) methyl]-2, 2-diethoxy-acetamidine (4.00 g, 14.8 mmol) in H2SO4 (10 mL), the mixture was stirred at 80° C. for 12 hours. On completion, the mixture was diluted with 100 mL water and 12 N NaOH aqueous solution was added until PH=12. The mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (FA condition) to give the title compound (80 mg, 52% yield, 81% purity) as a yellow solid. LC-MS (ESI, m/z): [M+1]+=179.1.


Step 4: 8-chloro-4-iodo-isoquinolin-3-amine

To a solution of 8-chloroisoquinolin-3-amine (300 mg, 1.68 mmol) in DCM (6 mL) and EtOH (3 mL) was added NIS (302 mg, 1.34 mmol), then the mixture was stirred at 5-25° C. for 12 hours. On completion, the residue was diluted with ethyl acetate (30 mL) and extracted with water (20 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=2:1) to give the title compound (300 mg, 54% yield) as a yellow solid. LC-MS (ESI, m/z): [M+1]+=305.0.


Step 5: tert-butyl 4-[2-(3-amino-8-chloro-4-isoquinolyl) ethynyl]piperidine-1-carboxylate

To a solution of 8-chloro-4-iodo-isoquinolin-3-amine (120 mg, 394 umol) in DMSO (6 mL) was added CuI (15.0 mg, 78.8 umol), Pd(PPh3)2Cl2 (27.7 mg, 39.4 umol), 4A molecular sieve (30 mg, 394 umol) and tert-butyl 4-ethynylpiperidine-1-carboxylate (165 mg, 788 umol) and CsF (239 mg, 1.58 mmol), then the mixture was stirred at 85° C. for 12 hours. On completion, the residue was diluted with ethyl acetate (30 mL) and extracted with water (20 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=4:1) to give the title compound (120 mg, 67% yield) as a yellow solid. LC-MS (ESI, m/z): [M+1-Boc]=386.2


Step 6: 8-chloro-4-[2-(4-piperidyl)ethynyl]isoquinolin-3-amine

To a solution of tert-butyl 4-[2-(3-amino-8-chloro-4-isoquinolyl)ethynyl]piperidine-1-carboxylate (100 mg, 259 umol) in THE (1 mL) was added 4M HCl/dioxane (1 mL), then the mixture was stirred at 35° C. for 12 hours. On completion, the reaction mixture was concentrated under reduced pressure to remove HCl/dioxane. The residue was diluted with DMF. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*25*10 um; mobile phase: [water (0.05% HCl)-ACN]; B %: 16%-36%, 12 min) to give the title compound (53.8 mg, 62% yield) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ=9.12-9.33 (m, 2H), 9.07 (s, 1H), 7.79 (d, J=8.8 Hz, 1H), 7.56-7.68 (m, 1H), 7.40 (d, J=6.8 Hz, 1H), 3.15-3.33 (m, 3H), 2.95-3.11 (m, 2H), 2.09-2.26 (m, 2H), 1.89-2.04 (m, 2H); LC-MS (ESI, m/z): [M+1]*=286.1.




embedded image


Example 22. 2-(6-Amino-5-(piperazin-1-yl)pyridazin-3-yl)-6-chlorophenol (I-28)
Step 1: tert-butyl 4-[(4-ethynylphenyl)methyl]piperazine-1-carboxylate

To a solution of 4-ethynylbenzaldehyde (1 g, 7.68 mmol) in DCM (20 mL) was added tert-butyl piperazine-1-carboxylate (2.86 g, 15.37 mmol), NaBH(OAc)3 (3.26 g, 15.4 mmol), then the mixture was stirred at 25° C. for 16 hours. On completion, the reaction mixture was quenched by addition water (50 mL). The residue was diluted with ethyl acetate (80 mL). The combined organic layers were washed with brine (20 mL×3), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=20:1) to give the title compound (1.8 g, 78% yield) as a yellow solid. LC-MS (ESI, m/z): [M+1]+=301.2


Step 2: (1E)-2-methoxybenzaldehyde oxime

To a solution of 2-methoxybenzaldehyde (2 g, 14.7 mmol) in EtOH (20 mL) was added NH2OH.HCl (1.4 g, 20.2 mmol) and TEA (1.92 g, 19.0 mmol). Then the mixture was stirred at 95° C. for 5 hours. On completion, the reaction mixture was concentrated under reduced pressure to remove EtOH. The residue was diluted with water (70 mL) and extracted with ethyl acetate 80 mL (20 mL×4). The combined organic layers were washed with brine (40 mL) and dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=5:1) to give the title compound (1.8 g, 79% yield) as a white solid. LC-MS (ESI, m/z): [M+1]+=152.1


Step 3: (1Z)—N-hydroxy-2-methoxy-benzimidoyl chloride

To a solution of (1E)-2-methoxybenzaldehyde oxime (1 g, 6.62 mmol) in DMF (4 mL) was added NCS (972 mg, 7.28 mmol). Then the mixture was stirred at 20° C. for 2 hours. On completion, the residue was diluted with water (70 mL) and extracted with ethyl acetate (20 mL×4). The combined organic layers were washed with brine (40 mL) and dried over NaSO4. The extracts were filtered and concentrated under reduced pressure to give the title compound (1 g, crude) as a white solid.


Step 4: tert-butyl 4-[[4-[3-(2-methoxyphenyl) isoxazol-5-yl]phenyl]methyl]piperazine-1-carboxylate

To a solution of (1Z)—N-hydroxy-2-methoxy-benzimidoyl chloride (324 mg, 1.75 mmol) in DMF (5 mL) was added TEA (212 mg, 2.10 mmol) and tert-butyl 4-[(4-ethynylphenyl)methyl]piperazine-1-carboxylate (525 mg, 1.75 mmol). Then the mixture was stirred at 60° C. for 12 hours. On completion, the residue was diluted with water (70 mL) and extracted with ethyl acetate (20 mL×4). The combined organic layers were washed with brine (40 mL) and dried over NaSO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=3:1) to give the title compound (500 mg, 51% yield) as a yellow oil. LC-MS (ESI, m/z): [M+1]+=450.3


Step 5: tert-butyl 4-[[4-[(Z)-1-amino-3-(2-methoxyphenyl)-3-oxo-prop-1-enyl]phenyl]methyl]piperazine-1-carboxylate

To a solution of tert-butyl 4-[[4-[3-(2-methoxyphenyl) isoxazol-5-yl]phenyl]methyl]piperazine-1-carboxylate (150 mg, 334 umol) in MeOH (3 mL) was added raney nickel (28.6 mg, 334 umol), NH2NH2-H2O (83.5 mg, 1.67 mmol). Then the mixture was stirred at 25° C. for 12 hours. On completion, the mixture was filtered and concentrated under reduced pressure to give the title compound (100 mg, crude) as a yellow solid. LC-MS (ESI, m/z): [M+1]+=452.3


Step 6: tert-butyl 4-[[4-[3-(2-methoxyphenyl)-1H-pyrazol-5-yl]phenyl]ethyl]piperazine-1-carboxylate

To a solution of tert-butyl 4-[[4-[(Z)-1-amino-3-(2-methoxyphenyl)-3-oxo-prop-1-enyl] phenyl]methyl]piperazine-1-carboxylate (155 mg, 343 umol) in DMF (3 mL) was added NH2NH2.H2O (85.8 mg, 1.71 mmol), then the mixture was stirred at 100° C. for 2 hours under microwave. On completion, the residue was diluted with ethyl acetate (30 mL) and extracted with water (20 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give the title compound (100 mg, crude) as colorless oil. LC-MS (ESI, m/z): [M+1]+=449.2


Step 7: 2-[5-[4-(piperazin-1-ylmethyl)phenyl]-1H-pyrazol-3-yl]phenol

To a solution of tert-butyl 4-[[4-[3-(2-methoxyphenyl)-1H-pyrazol-5-yl]phenyl]methyl]piperazine-1-carboxylate (50 mg, 111.47 umol) in DCM (1 mL) was added BBr3 (1 M, 445 uL). Then the mixture was stirred at 30° C. for 12 hours. On completion, the mixture was diluted with DCM, water and a litter DMF and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*30 mm*4 um; mobile phase: [water (0.05% HCl)-ACN]; B %: 12%-32%, 12 min) to give the title compound (6.28 mg, 16% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ=9.80 (s, 2H), 7.96 (d, J=7.6 Hz, 2H), 7.76 (d, J=7.2 Hz, 3H), 7.33 (s, 1H), 7.20 (t, J=7.2 Hz, 1H), 6.99 (d, J=8.0 Hz, 1H), 6.92 (t, J=7.2 Hz, 1H), 4.45 (s, 2H) 2.96-3.71 (m, 9H); LC-MS (ESI, m/z): [M+1]+=335.2.




embedded image


Example 23. 2-(1H-indazol-3-yl)phenol (I-29)
Step 1: 2-(1H-indazol-3-yl)phenol

A mixture of 3-bromo-1H-indazole (200 mg, 1.02 mmol), (2-hydroxyphenyl)boronic acid (140 mg, 1.02 mmol), Pd(PPh3)4 (58.65 mg, 50.7 umol) in NaHCO3 (0.5 mL) and IPA (2 mL) was stirred at 150° C. for 20 minutes under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to remove solvent, diluted with H2O (60 mL) and extracted with DCM (30 mL×3). The combined organic layers were washed with brine (20 mL), dried over by anhydrous sodium sulfate, concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*30 mm*4 um; mobile phase: [water (0.05% HCl)-ACN]; B %: 52%-72%, 12 min) to give the title compound (39 mg, 15% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ=13.43 (d, J=1.2 Hz, 1H), 10.74 (s, 1H), 8.13 (d, J=8.4 Hz, 1H), 7.94 (dd, J=1.5, 7.6 Hz, 1H), 7.63 (d, J=8.4 Hz, 1H), 7.46 (td, J=0.9, 7.6 Hz, 1H), 7.30-7.19 (m, 2H), 7.04-6.97 (m, 2H); LC-MS (ESI, m/z): [M+1]+=211.1.


Example 24. 2-(1H-Pyrazolo[4,3-c]pyridin-3-yl)phenol (I-30)



embedded image


Step 1: 2-(1H-pyrazolo[4,3-c]pyridin-3-yl)phenol

A mixture of 3-bromo-1H-pyrazolo[4,3-c]pyridine (200 mg, 1.01 mmol), (2-hydroxyphenyl)boronic acid (139 mg, 1.01 mmol), Pd(PPh3)4 (58.3 mg, 50.5 umol) in NaHCO3 (0.5 mL) and IPA (2 mL) was stirred at 150° C. for 20 minutes under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to remove solvent and diluted with H2O (60 mL) and extracted with DCM (30 mL×3). The combined organic layers were washed with brine (20 mL), dried over by sodium sulphate anhydrous, concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*30 mm*4 um; mobile phase: [water (0.05% HCl)-ACN]; B %: 5%-25%, 12 min) to give the title compound (15 mg, 6% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ=15.00-14.51 (m, 1H), 10.51 (s, 1H), 9.62 (s, 1H), 8.55 (d, J=6.8 Hz, 1H), 8.14-8.10 (m, 1H), 7.76 (dd, J=1.6, 7.6 Hz, 1H), 7.38 (td, J=1.6, 7.8 Hz, 1H), 7.17-7.12 (m, 1H), 7.03-6.97 (m, 1H); LC-MS (ESI, m/z): [M+1]+=212.1.




embedded image


Example 25. 2-(6-Amino-5-piperazin-1-yl-pyridazin-3-yl)-5-chloro-phenol (I-31)
Step 1: tert-butyl 4-[3-amino-6-(5-chloro-2-methoxy-phenyl)pyridazin-4-yl]piperazine-1-carboxylate

Tert-butyl-4-(3-amino-6-chloro-pyridazin-4-yl)piperazine-1-carboxylate (200 mg, 637 umol) in dioxane (5 mL) and H2O (1 mL) was added K2CO3 (264 mg, 1.91 mmol), BrettPhos Pd G3 (28.9 mg, 31.9 umol) and (5-chloro-2-methoxy-phenyl)boronic acid (356 mg, 1.91 mmol). The mixture was stirred at 85° C. for 12 hours under N2 atmosphere. The reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (50 mL×3). The combined organic layers were washed with brine (50 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether:Ethyl acetate=1:2) to give the title compound (170 mg, 36% yield) as a yellow oil. LC-MS (ESI, m/z): [M+1]+=420.4.


Step 2: 2-(6-amino-5-piperazin-1-yl-pyridazin-3-yl)-5-chloro-phenol

To a solution of tert-butyl 4-[3-amino-6-(5-chloro-2-methoxy-phenyl)pyridazin-4-yl]piperazine-1-carboxylate (100 mg, 238 umol) in DCM (2 mL) was added BBr3 (1 M, 714 uL) at −78° C. The mixture was stirred at −78° C. for 2 hours. The reaction mixture was added water (5 mL) and DMF (2 mL), concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25 mm*5 um; mobile phase: [water (10 mm NH4HCO3)-ACN]; B %: 14%-44%, 10 min) to give the title compound (16.0 mg, 22% yield) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ=9.58 (d, J=1.2 Hz, 2H), 7.73 (s, 1H), 7.58-7.71 (m, 3H), 7.20 (s, 1H), 7.04 (dd, J=8.0, 2.0 Hz, 1H), 3.42-3.46 (m, 4H), 3.33 (m, 4H); LC-MS (ESI, m/z): [M+1]+=306.2.




embedded image


Example 26. 2-(6-Amino-5-(piperazin-1-yl)pyridazin-3-yl)-6-chlorophenol (I-32)
Step 1: tert-butyl 4-[3-amino-6-(3-chloro-2-methoxy-phenyl)pyridazin-4-yl]piperazine-1-carboxylate

A mixture of tert-butyl 4-(3-amino-6-chloro-pyridazin-4-yl)piperazine-1-carboxylate (0.2 g, 637 umol), (3-chloro-2-methoxy-phenyl)boronic acid (356 mg, 1.91 mmol), K2CO3 (176 mg, 1.27 mmol), BrettPhos Pd G3 (57 mg, 63.7 umol) in dioxane (5 mL) and H2O (1 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 85° C. for 12 hours under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=1/1 to 0/1) to give the title compound (0.2 g, 59% yield) as a yellow liquid. LC-MS (ESI, m/z): [M+1]+=420.2.


Step 2: 2-(6-amino-5-piperazin-1-yl-pyridazin-3-yl)-6-chloro-phenol

To a solution of tert-butyl 4-[3-amino-6-(3-chloro-2-methoxy-phenyl)pyridazin-4-yl]piperazine-1-carboxylate (50 mg, 119 umol) in DCM (3 mL) was added BBr3 (1 M, 952 uL). The mixture was stirred at 25° C. for 12 hours. The reaction mixture was filtered to give a residue. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25 mm*5 um; mobile phase: [water (10 mm NH4HCO3)-ACN]; B %: 12%-42%, 10 min) to give the title compound (10 mg, 31.4 umol, 26% yield) as a yellow solid. 1H NMR (400 MHz, CD3OD) δ=7.80 (m, 1H), 7.56 (s, 1H), 7.38 (dd, J=1.4, 7.8 Hz, 1H), 6.91 (t, J=7.8 Hz, 1H), 3.16 (d, J=4.6 Hz, 4H), 3.12-3.05 (m, 4H). LC-MS (ESI, m/z): [M+1]+=306.5.




embedded image


Example 27. 2-(6-Amino-5-piperazin-1-yl-pyridazin-3-yl)-4-chloro-phenol (I-33)
Step 1: tert-butyl 4-[3-amino-6-(5-chloro-2-methoxy-phenyl)pyridazin-4-yl]piper azine-1-carboxylate

Tert-butyl-4-(3-amino-6-chloro-pyridazin-4-yl)piperazine-1-carboxylate (200 mg, 637 umol) in dioxane (5 mL) and H2O (1 mL) was added K2CO3 (264 mg, 1.91 mmol), BrettPhos Pd G3 (28.9 mg, 31.9 umol) and (5-chloro-2-methoxy-phenyl)boronic acid (356 mg, 1.91 mmol). The mixture was stirred at 85° C. for 12 hours under N2 atmosphere. The reaction mixture was diluted with water (50 mL) and extracted with Ethyl acetate (50 mL×3). The combined organic layers were washed with brine (50 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether:Ethyl acetate=1:2) to give the title compound (170 mg, 36% yield) as a yellow oil. LC-MS (ESI, m/z): [M+1]+=420.4.


Step 2: 2-(6-amino-5-piperazin-1-yl-pyridazin-3-yl)-4-chloro-phenol

To a solution of tert-butyl 4-[3-amino-6-(5-chloro-2-methoxy-phenyl)pyridazin-4-yl]piperazine-1-carboxylate (100 mg, 238 umol) in DCM (2 mL) was added BBr3 (1 M, 714.45 uL) at −78° C.. The mixture was stirred at −78° C. for 2 hours. The reaction mixture was added water (5 mL) and DMF (2 mL), concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25 mm*5 um; mobile phase: [water (10 mm NH4HCO3)-ACN]; B %: 14%-44%, 10 min) to give the title compound (16.0 mg, 22% yield) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ=14.25-14.83 (m, 1H), 8.06 (d, J=2.8 Hz, 1H), 7.54 (s, 1H), 7.26 (dd, J=8.8, 2.8 Hz, 1H), 6.91 (d, J=8.4 Hz, 1H), 6.22-6.37 (m, 2H), 3.00-3.04 (m, 4H), 2.91 (d, J=4.4 Hz, 4H); LC-MS (ESI, m/z): [M+1]*=306.2.




embedded image


Example 28. 2-(6-Amino-5-piperazin-1-yl-pyridazin-3-yl)-4-methyl-phenol (I-34)
Step 1: tert-butyl 4-[3-amino-6-(2-hydroxy-5-methyl-phenyl)pyridazin-4-yl]piperazine-1-carboxylate

Tert-butyl 4-(3-amino-6-chloro-pyridazin-4-yl)piperazine-1-carboxylate (200 mg, 637 umol) in dioxane (5 mL) and H2O (1 mL) was added K2CO3 (264 mg, 1.91 mmol), BrettPhos Pd G3 (28.9 mg, 31.9 umol) and (2-hydroxy-5-methyl-phenyl)boronic acid (291 mg, 1.91 mmol). The mixture was stirred at 85° C. for 12 hours under N2 atmosphere. The reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (50 mL×3). The combined organic layers were washed with brine (50 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether: Ethyl acetate=I/O to 1/3) to give the title compound (200 mg, 76% yield) as a yellow solid. LC-MS (ESI, m/z): [M+1]*=386.4.


Step 2: tert-butyl 4-[3-amino-6-(1H-pyrazol-5-yl)pyridazin-4-yl]-3,6-dihydro-2H-pyridine-1-carboxylate

To a solution of tert-butyl 4-[3-amino-6-(2-hydroxy-5-methyl-phenyl)pyridazin-4-yl]piperazine-1-carboxylate (100 mg, 259 umol) in DCM (5 mL) was added TFA (906 mg, 7.94 mmol) at 0° C. The mixture was stirred at 0-25° C. for 2 hours. The mixture was added DCM (10 ml), little water and DMF (3 ml), filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Xtimate C18 10u 250 mm*80 mm; mobile phase: [water (0.05% ammonia hydroxide v/v)-ACN]; B %: 10%-40%, 10 min) to give the title compound (16.0 mg, 21% yield) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ=13.81 (s, 1H), 8.66-8.89 (m, 1H), 7.77 (d, J=1.6 Hz, 1H), 7.57 (s, 1H), 7.06 (dd, J=8.0, 1.6 Hz, 1H), 6.80 (d, J=8.0 Hz, 1H), 6.48 (s, 2H), 3.31-3.34 (m, 4H), 3.28 (m, 4H), 2.30 (s, 3H); LC-MS (ESI, m/z): [M+1]+=286.3.


Example 29. 4-(4-Piperidyl)-6-(1H-pyrazol-5-yl)pyridazin-3-amine (I-35)



embedded image


Step 1: tert-butyl 4-(3-amino-6-chloro-pyridazin-4-yl)-3,6-dihydro-2H-pyridine-1-carboxylate

To a mixture of 4-bromo-6-chloro-pyridazin-3-amine (500 mg, 2.40 mmol), tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1-carboxylate (1.11 g, 3.60 mmol), K2CO3 (995 mg, 7.20 mmol) and Pd(PPh3)4 (277 mg, 240 umol) in a mixture of dioxane (20 mL) and H2O (4 mL) was degassed and purged with N2 for 3 times. The mixture was stirred at 85° C. for 12 hours under N2 atmosphere. The reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (50 mL×3). The combined organic layers were washed with brine (50 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether: Ethyl acetate=20/1 to 1/2) to give the title compound (650 mg, 82% yield) as a yellow solid. LC-MS (ESI, m/z): [M+1]+=311.1.


Step 2: tert-butyl 4-[3-amino-6-(1H-pyrazol-5-yl)pyridazin-4-yl]-3,6-dihydro-2H-pyridine-1-carboxylate

To a solution of tert-butyl 4-(3-amino-6-chloro-pyridazin-4-yl)-3,6-dihydro-2H-pyridine-1-carboxylate (300 mg, 965 umol), 1H-pyrazol-5-ylboronic acid (162 mg, 1.45 mmol), Pd(PPh3)4 (112 mg, 96.5 umol), Na2CO3 (2 M, 7.24 mL) in dioxane (10 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 120° C. for 1 hour under microwave irradiation. The reaction mixture was diluted with water (60 mL) and extracted with ethyl acetate (60 mL×3). The combined organic layers were washed with brine (60 mL×2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (TFA condition) to give the title compound (34 mg, 10% yield) as a brown solid. LC-MS (ESI, m/z): [M+1]+=343.1.


Step 3: tert-butyl 4-[3-amino-6-(1H-pyrazol-5-yl)pyridazin-4-yl]piperidine-1-carboxylate

To a solution of tert-butyl 4-[3-amino-6-(1H-pyrazol-5-yl)pyridazin-4-yl]-3,6-dihydro-2H-pyridine-1-carboxylate (34 mg, 99.3 umol) in EtOH (3 mL) was added Pd/C (20 mg, 10% purity). The mixture was stirred at 20° C. for 12 hours under H2. The reaction mixture was added EtOH (10 mL), filtered and concentrated under reduced pressure to give the title compound (19 mg, crude) as a yellow solid. LC-MS (ESI+) m/z=345.3 (M+H)*.


Step 4: 4-(4-piperidyl)-6-(1H-pyrazol-5-yl)pyridazin-3-amine(SMA-472-001H)

The solution of tert-butyl 4-[3-amino-6-(1H-pyrazol-5-yl)pyridazin-4-yl]piperidine-1-carboxylate (19 mg, 55.2 umol) in TFA (1 mL) was stirred at 25° C. for 12 hours. The reaction mixture was added water (5 mL) and DMF (2 mL), concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*25*10 um; mobile phase: [water (0.05% HCl)-ACN]; B %: 0%-10%, 12 min) to give the title compound (9.31 mg, 60% yield) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ=9.08-9.26 (m, 2H), 8.82 (m, 2H), 8.03 (s, 1H), 7.94 (d, J=2.2 Hz, 1H), 6.86 (d, J=2.0 Hz, 1H), 3.41 (d, J=12.4 Hz, 2H), 3.22 (t, J=11.6 Hz, 1H), 2.97-3.09 (m, 2H), 1.98-2.08 (m, 2H), 1.85-1.96 (m, 2H); LC-MS (ESI, m/z): [M+1]*=245.2.


Example 30. 4-(1-(Piperidin-4-ylmethyl)-1H-pyrazol-4-yl)-6-(thiazol-2-yl)pyridazin-3-amine (I-36)



embedded image


Step 1: tert-butyl 4-[[4-(3-amino-6-thiazol-2-yl-pyridazin-4-yl) pyrazol-1-yl]methyl]piperidine-1-carboxylate

To a solution of tert-butyl 4-[[4-(3-amino-6-chloro-pyridazin-4-yl) pyrazol-1-yl]methyl]piperidine-1-carboxylate (100 mg, 255 umol) in THF (4 mL) was added APhos-Pd-G3 (29.4 mg, 25.5 umol) and tributyl(thiazol-2-yl)stannane (286 mg, 764 umol), then the mixture was stirred at 90° C. for 12 hours. On completion, the residue was diluted with ethyl acetate (30 mL) and extracted with water (20 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=4:1) to give the title compound (70 mg, crude) as a white solid. LC-MS (ESI, m/z): [M+1]+=442.3.


Step 2: 4-[1-(4-piperidylmethyl)pyrazol-4-yl]-6-thiazol-2-yl-pyridazin-3-amine

To a solution of tert-butyl 4-[[4-(3-amino-6-thiazol-2-yl-pyridazin-4-yl) pyrazol-1-yl]methyl]piperidine-1-carboxylate (70 mg, 159 umol) in TFA (1 mL) and DCM (1 mL), then the mixture was stirred at 25° C. for 2 hours. On completion, the reaction mixture was concentrated under reduced pressure to remove DCM and TFA. The mixture was diluted with DMF. The residue was purified by prep-HPLC (column: Shim-pack C18 150*25*10 um; mobile phase: [water (0.1% TFA) -ACN]; B %: 25%, 10 min) to give the title compound (5.96 mg, 10% yield) as a yellow oil.



1H NMR (400 MHz, DMSO-d6) δ=8.94-9.36 (m, 2H) 8.57 (s, 1H) 8.30 (s, 1H) 8.15 (s, 1H) 8.07 (d, J=2.4 Hz, 1H) 8.00 (d, J=3.2 Hz, 1H) 4.02-4.21 (m, 2H) 3.16-3.33 (m, 2H) 2.75-2.93 (m, 2H) 1.64-1.82 (m, 2H) 1.38-1.57 (m, 2H) 1.03-1.30 (m, 1H); LC-MS (ESI, m/z): [M+1]+=342.2.


Example 31. SMARCA2 BromoScan and Human HTRF Inhibition

SMARCA2 BromoScan


Protocol Description


Bromodomain assays. T7 phage strains displaying bromodomains were grown in parallel in 24-well blocks in an E. coli host derived from the BL21 strain. E. coli were grown to log-phase and infected with T7 phage from a frozen stock (multiplicity of infection=0.4) and incubated with shaking at 32° C. until lysis (90-150 minutes). The lysates were centrifuged (5,000×g) and filtered (0.2 μm) to remove cell debris. Streptavidin-coated magnetic beads were treated with biotinylated small molecule or acetylated peptide ligands for 30 minutes at room temperature to generate affinity resins for bromodomain assays. The liganded beads were blocked with excess biotin and washed with blocking buffer (SeaBlock (Pierce), 1% BSA, 0.05% Tween 20, 1 mM DTT) to remove unbound ligand and to reduce non-specific phage binding. Binding reactions were assembled by combining bromodomains, liganded affinity beads, and test compounds in 1× binding buffer (17% SeaBlock, 0.33×PBS, 0.04% Tween 20, 0.02% BSA, 0.004% Sodium azide, 7.4 mM DTT). Test compounds were prepared as 1000λ stocks in 100% DMSO. Kds were determined using an 11-point 3-fold compound dilution series with one DMSO control point. All compounds for Kd measurements are distributed by acoustic transfer (non-contact dispensing) in 100% DMSO. The compounds were then diluted directly into the assays such that the final concentration of DMSO was 0.09%. All reactions performed in polypropylene 384-well plates. Each was a final volume of 0.02 ml. The assay plates were incubated at room temperature with shaking for 1 hour and the affinity beads were washed with wash buffer (1× PBS, 0.05% Tween 20). The beads were then re-suspended in elution buffer (1× PBS, 0.05% Tween 20, 2 μM non-biotinylated affinity ligand) and incubated at room temperature with shaking for 30 minutes. The bromodomain concentration in the eluates was measured by qPCR.


Compound Handling: An 11-point 3-fold serial dilution of each test compound was prepared in 100% DMSO at 1000× final test concentration. All compounds for Kd measurements are distributed by acoustic transfer (non-contact dispensing) in 100% DMSO. The compounds were then diluted directly into the assays such that the final concentration of DMSO was 0.09%. Most Kds were determined using a compound top concentration=10,000 nM. If the initial Kd determined was <0.169 nM (the lowest concentration tested), the measurement was repeated with a serial dilution starting at a lower top concentration.


Binding constants (Kds) were calculated with a standard dose-response curve using the Hill equation. Curves were fitted using a non-linear least square fit with the Levenberg-Marquardt algorithm.












SMARCA2 HTRF Assay


Materials

















PerkinElmer EnVision 2104 Multilabel Reader



OptiPlate 384-well Microplate, PerkinElmer 6007290



Mab Anti 6His Tb cryptate Gold, Cisbio 61HI2TLB, Lot 10A



His-tagged SMARCA2-17, Viva 20181219



SMA-611-001H, Wuxi EW12802-518



5× Epigenetics Buffer 1 Kit, PerkinElmer AL008C



SMA-788-001H, Wuxi EW16490-111









Methods


Sample handling: Prepared 1× Epigenetics Buffer according to manufacturer's instructions. Prepared 32 nM SMARCA2-17 (4X) with Epigenetics Buffer; Prepared 4× Tb working solution: dilute Mab Anti 6His Tb cryptate Gold in PPI-Terbium Detection buffer to 0.21 ng/l (100× dilution from stock). Prepared SMARCA2-mAb-Tb working solution by mixing equal volume of 32 nM SMARCA2 with Tb working solution. Started a half-hour timer. In a 96-well plate, prepared serial dilution of compound DMSO solutions. Placed 24 μl/well of Epigenetics Buffer to a second 96-well plate. Transferred 1 μl of the 100× compound DMSO solution to the second 96-well plate to make 4× compound working solution. Prepared 80 nM SMA-611-001H/SMA-788-001H (4×) with Epigenetics Buffer. Added 5 μl/well of SMA-611-001H into the 384-well plate. Added 5 μl/well of SMA-788-001H into the 384-well plate. Added 5 μl/well of 4× compound working solution into the 384-well plate, mix well. Added 10 l/well of SMARCA2-mAb-Tb working solution into 384-well plate. Incubated the above mixtures at room temperature for 30 min. Data collected, processed and on EnVision. Calculated 520 nm/495 nm readouts ratio from raw data.


SMARCA2 in vitro activity for compounds of the invention are presented in Table 2. The letter codes for SMARCA2 external Kd (nM) include: A (<100 nM), B (100-500 nM), C (501-1000 nM), and D (>1000 nM). The letter codes for SMARCA2 average IC50 (μM) include: A (<1 μM), B (1-100 μM), C (101-500 μM), and D (>500 μM).









TABLE 2







SMARCA2 BromoScan and HTRF Results.










BromoScan
SMARCA2



Panel - SMARCA2:
binding Human HTRF:


I-#
External Kd (nM)
Average IC50 (μM)





I-1 
D



I-2 
D



I-3 
D



I-4 
D



I-5 
D



I-6 
D



I-7 
D



I-8 
D



I-9 
D



I-10
D



I-11
D



I-12
D



I-13
D



I-14
D



I-15
D



I-16
D



I-17
D



I-18
D



I-19
A
B


I-20
A



I-21
D



I-22
D



I-23
D



I-24
C



I-25

C


I-26

C


I-27
D



I-28
D



I-29
D



I-30
D



I-31
D



I-32
A



I-33
B



I-34
D



I-35
D



I-36




I-37
D



I-38
D










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

Claims
  • 1. A compound of formula I-IV:
  • 2. The compound of claim 1, wherein said compound is any one of Formula I-a, I-b, I-c, I-d, I-e, II-a, II-b, III-a, III-b, III-c, IV-a:
  • 3. The compound of claim 1 or 2, wherein each Rx is independently hydrogen, R6, halogen, —CN, —NO2, —OR, —SR, —NR2, —SiR3, —S(O)2R, —S(O)2NR2, —S(O)R, —C(O)R, —C(O)OR, —C(O)NR2, —C(O)N(R)OR, —C(R)2N(R)C(O)R, —C(R)2N(R)C(O)NR2, —OC(O)R, —OC(O)NR2, —OP(O)R2, —OP(O)(OR)2, —OP(O)(OR)NR2, —OP(O)(NR2)2, —N(R)C(O)OR, —N(R)C(O)R, —N(R)C(O)NR2, —N(R)S(O)2R, —N(R)P(O)R2, —N(R)P(O)(OR)2, —N(R)P(O)(OR)NR2, —N(R)P(O)(NR2)2, or —N(R)S(O)2R.
  • 4. The compound of any one of claims 1-3, wherein each Ry is independently hydrogen, R6, halogen, —CN, —NO2, —OR, —SR, —NR2, —SiR3, —S(O)2R, —S(O)2NR2, —S(O)R, —C(O)R, —C(O)OR, —C(O)NR2, —C(O)N(R)OR, —C(R)2N(R)C(O)R, —C(R)2N(R)C(O)NR2, —OC(O)R, —OC(O)NR2, —OP(O)R2, —OP(O)(OR)2, —OP(O)(OR)NR2, —OP(O)(NR2)2, —N(R)C(O)OR, —N(R)C(O)R, —N(R)C(O)NR2, —N(R)S(O)2R, —N(R)P(O)R2, —N(R)P(O)(OR)2, —N(R)P(O)(OR)NR2, —N(R)P(O)(NR2)2, or —N(R)S(O)2R.
  • 5. The compound of any one of claims 1-4, wherein Rz is hydrogen, —NR2, —C(O)NR2, —CF3, or
  • 6. The compound of any one of claims 1-5, wherein each R6 is independently an optionally substituted group selected from C1-6 aliphatic, phenyl, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • 7. The compound of any one of claims 1-6, wherein each Ring P is independently phenyl, a 4-10 membered saturated or partially unsaturated mono- or bicyclic carbocyclic or heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • 8. The compound of any one of claims 1-7, wherein each Lx is independently a covalent bond or a C1-3 bivalent straight or branched saturated or unsaturated hydrocarbon chain wherein 1-3 methylene units of the chain are independently and optionally replaced with —O—, —C(O)—, —C(S)—, —C(R)2—, —CH(R)—, —C(F)2—, —N(R)—, —S(O)2—, —CH═CH—, or —C≡C—.
  • 9. The compound of any one of claims 1-8, wherein said compound is selected from any one of the compounds depicted in Table 1, or a pharmaceutically acceptable salt thereof.
  • 10. A pharmaceutical composition comprising a compound of any one of claims 1-9, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • 11. The pharmaceutical composition according to claim 10, further comprising an additional therapeutic agent.
  • 12. A method of inhibiting one or more of SMARCA2, SMARCA4, and PB1 protein in a patient or biological sample comprising administering to said patient, or contacting said biological sample with a compound according to any one of claims 1-9, or a pharmaceutical composition thereof.
  • 13. A method of treating one or more SMARCA2-mediated, SMARCA4-mediated, or PB1-mediated disorder, disease, or condition in a patient comprising administering to said patient a compound according to any one of claims 1-9, or a pharmaceutical composition thereof.
  • 14. The method according to claim 13, further comprising administration of an additional therapeutic agent.
  • 15. The method according to claim 13, wherein the one or more SMARCA2-mediated, SMARCA4-mediated, or PB1-mediated disorder, disease or condition is selected from a cancer, a neurodegenerative disease, a viral disease, an autoimmune disease, an inflammatory disorder, a hereditary disorder, a hormone-related disease, a metabolic disorder, a condition associated with organ transplantation, an immunodeficiency disorder, a destructive bone disorder, a proliferative disorder, an infectious disease, a condition associated with cell death, thrombin-induced platelet aggregation, liver disease, a pathologic immune condition involving T cell activation, a cardiovascular disorder, and a CNS disorder.
  • 16. The method according to claim 15, wherein the cancer is selected from lung cancer, breast cancer, pancreatic cancer, colorectal cancer, melanoma, leukemia, and malignant rhabdoid tumors (MRT).
PCT Information
Filing Document Filing Date Country Kind
PCT/US2020/036921 6/10/2020 WO
Provisional Applications (1)
Number Date Country
62859325 Jun 2019 US