QUINOLINE cGAS ANTAGONIST COMPOUNDS

Abstract
The present disclosure provides compounds that are cGAS antagonists, methods of preparation of the compounds, pharmaceutical compositions comprising the compounds, and their use in medical therapy.
Description
1. BACKGROUND

Cyclic GMP-AMP synthase (cGAS) (UniProtKB—Q8N884) is an enzyme that acts as a DNA sensor to elicit an immune response to pathogens via activation of the stimulator of interferon genes (STING) receptor. Aberrant activation of cGAS by self-DNA is shown to underlie debilitating and sometimes fatal autoimmune diseases. Knockout studies in animal models have indicated that inhibiting cGAS is a promising approach for therapeutic intervention. Additionally, recent studies have shown that the cGAS-STING pathway plays a key role in the innate immune response to tumors, and stimulation of the pathway is a promising strategy being tested clinically for cancer immunotherapy.


An ongoing need exists in the art for effective treatments of human autoimmune and auto-inflammatory diseases, such as systemic lupus erythematosus (SLE), scleroderma, psoriasis, Aicardi Goutibres syndrome (AGS), Sjogren's syndrome, rheumatoid arthritis, inflammatory bowel diseases, multiple sclerosis, diabetes, cardiovascular, and neurodegenerative diseases. Accordingly, there remains a need to find potent and selective small molecule inhibitors of cGAS useful for the treatment of these and other debilitating human diseases associated with the aberrant activation of cGAS.


2. SUMMARY

The present disclosure relates to small molecule cGAS antagonist compounds, methods of preparation of the compounds, pharmaceutical compositions comprising the compounds, and their use in medical therapy. In particular, the present disclosure provides quinoline cGAS antagonist compounds, which find utility as inhibitors of cGAS. An advantage of the compounds provided herein is that a broad range of pharmacological activities is possible, consistent with the inhibition of cGAS. In addition, the disclosure provides methods of using the compounds described herein for the treatment of inflammatory, allergic, autoimmune, and infectious diseases. The compounds can also be used for the treatment of senescence- or age-related diseases, such as neurodegenerative diseases, cardiovascular diseases, liver and renal diseases, cancer, and premature aging.


It has now been found that compounds of the disclosure, and pharmaceutically acceptable compositions thereof, are effective for the inhibition of cGAS. Such compounds of the disclosure have the general formula I.




embedded image


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


Compounds of the present disclosure, and pharmaceutically acceptable compositions thereof, are useful for treating a variety of diseases, disorders, or conditions, associated with aberrant activation of cGAS. Such diseases, disorders, or conditions include those described herein.


Compounds provided by the present disclosure are also useful for the study of cGAS enzymes in biological and pathological phenomena and the comparative evaluation of new cGAS antagonists or other regulators of cGAS, signaling pathways, and cytokine levels in vitro or in vivo.







3. DETAILED DESCRIPTION

In the following disclosure, certain specific details are set forth in order to provide a thorough understanding of various embodiments. However, one skilled in the art will understand that the methods and uses described herein may be practiced without these details. In other instances, well-known structures have not been shown or described in detail to avoid unnecessarily obscuring descriptions of the embodiments. Unless the context requires otherwise, throughout the specification and claims which follow, the word “comprise” and variations thereof, such as, “comprises” and “comprising” are to be construed in an open, inclusive sense, that is, as “including, but not limited to.” Further, headings provided herein are for convenience only and do not interpret the scope or meaning of the claimed invention. Reference throughout this specification to “one embodiment” or “an embodiment” means that a particular feature, structure, or characteristic described in connection with the embodiment is included in at least one embodiment. Thus, the appearances of the phrases “in one embodiment” or “in an embodiment” in various places throughout this specification are not necessarily all referring to the same embodiment. Furthermore, the particular features, structures, or characteristics may be combined in any suitable manner in one or more embodiments. Also, as used in this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the content clearly dictates otherwise. It should also be noted that the term “or” is generally employed in its sense including “and/or” unless the content clearly dictates otherwise.


3.1. Definitions

Compounds of the present disclosure 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 disclosure, 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 to 6 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1 to 5 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1 to 4 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1 to 3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1 to 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 to 12 ring members and 0 to 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


In some embodiments, bicyclo[1.1.1.]pentane is a phenyl isostere.


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 an oxygen, sulfur, nitrogen, phosphorus, or silicon atom in a heterocyclic ring.


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 4 to 14 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 disclosure, “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 1 to 5 heteroatoms. The term “heteroatom” in the context of “heteroaryl” particularly includes, but is not limited 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 monocyclic or bicyclic. A heteroaryl ring may include one or more oxo (═O) or thioxo (═S) substituent. 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- to 10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably 1 to 4, 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 may have 0 to 3 heteroatoms selected from oxygen, sulfur or nitrogen.


A heterocyclic ring can be attached to a provided compound 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 monocyclic, bicyclic, bridged bicyclic, or spirocyclic. A heterocyclic ring may include one or more oxo (═O) or thioxo (═S) substituent. 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 disclosure may contain “substituted” moieties. In general, the term “substituted” 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 one or more substitutable position of the group, and when more than one position in any given structure is 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 disclosure 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)0-6R; —(CH2)0-6OR; —O(CH2)0-6R; —O—(CH2)0-6C(O)OR; —(CH2)0-6CH(OR)2; —(CH2)0-6SR; —(CH2)0-6Ph, which Ph may be substituted with R; —(CH2)0-6O(CH2)0-1Ph which Ph may be substituted with R; —CH═CHPh, which Ph may be substituted with R; —(CH2)0-6O(CH2)0-1-pyridyl which pyridyl may be substituted with R; —NO2; —CN; —N3; —(CH2)0-6N(R)2; —(CH2)0-6N(R)C(O)R; —N(R)C(S)R; —(CH2)0-6N(R)C(O)NR2; —N(R)C(S)NR2; —(CH2)0-6N(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)0-6C(O)R; —C(S)R; —(CH2)0-6C(O)OR; —(CH2)0-6C(O)SR; —(CH2)0-6C(O)OSiR3; —(CH2)0-6OC(O)R; —OC(O)(CH2)0-6SR, —(CH2)0-6SC(O)R; —(CH2)0-6C(O)NR2; —C(S)NR2; —C(S)SR; —SC(S)SR, —(CH2)0-6OC(O)NR2; —C(O)N(OR)R; —C(O)C(O)R; —C(O)CH2C(O)R; —C(NOR)R; —(CH2)0-6SSR; —(CH2)0-6S(O)2R; —(CH2)0-6S(O)2OR; —(CH2)0-6OS(O)2R; —S(O)2NR2; —(CH2)0-6S(O)R; —N(R)S(O)2NR2; —N(R)S(O)2R; —N(OR)R; —C(NH)NR2; —P(O)2R; —P(O)R2; —P(O)(OR)2; —OP(O)(R)OR; —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 R may be substituted as defined below and is independently hydrogen, C1-6 aliphatic, —CH2Ph, —O(CH2)0-1Ph, —CH2-(5- to 6-membered heteroaryl ring), or a 5- to 6-membered saturated, partially unsaturated, or aryl ring having 0 to 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- to 12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0 to 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- to 6-membered saturated, partially unsaturated, or aryl ring having 0 to 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- to 6-membered saturated, partially unsaturated, or aryl ring having 0 to 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- to 6-membered saturated, partially unsaturated, or aryl ring having 0 to 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- to 6-membered saturated, partially unsaturated, or aryl ring having 0 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.


Suitable substituents on a substitutable nitrogen of an “optionally substituted” group include —R, —NR2, —C(O)R, —C(O)OR, —C(O)C(O)R, —C(O)CH2C(O)R, —S(O)2R, —S(O)2NR2, —C(S)NR2, —C(NH)NR2, or —N(R)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- to 6-membered saturated, partially unsaturated, or aryl ring having 0 to 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- to 12-membered saturated, partially unsaturated, or aryl monocyclic or bicyclic ring having 0 to 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- to 6-membered saturated, partially unsaturated, or aryl ring having 0 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.


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


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, which is incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of this disclosure 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 disclosure. Unless otherwise stated, all tautomeric forms of the compounds of the disclosure are within the scope of the disclosure. 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 disclosure. Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present disclosure.


As used herein, the term “antagonist” is defined as a compound that binds to and/or inhibits cGAS with measurable affinity. In certain embodiments, a cGAS antagonist has an IC50 and/or binding constant of less than about 30 μM or less than about 2 μM.


3.2. Compounds of the Present Disclosure

Compounds of the present disclosure, and compositions thereof, are useful as cGAS antagonists. In some embodiments, a provided compound inhibits cGAS.


The present disclosure provides a compound of formula I:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • R1 is halogen, —OR, —NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —NRSO2R, —SR, —SO2R, —SO2NR2, —S(O)R, or RA, particularly —NR2 or RA;
    • each R is independently hydrogen or an optionally substituted group selected from C1-6 aliphatic; benzyl; phenyl; a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carbocyclic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur; and a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur, or: two R groups on the same nitrogen are optionally taken together with their intervening atoms to form an optionally substituted 4- to 7-membered saturated, partially unsaturated, or heteroaryl ring having 0 to 3 heteroatoms, in addition to the nitrogen, independently selected from nitrogen, oxygen, phosphorus, and sulfur;
    • R2 is halogen, —OR, —NR2, —NRNR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —NRSO2R, —SR, —SO2R, —SO2NR2, —S(O)R, or RB, particularly —NR2 or RB;
    • each R3 is independently halogen, —OR, —NR2, —SR, or RC, particularly halogen, such as chloro;
    • RA, RB, and RC, independently, are an optionally substituted group selected from C1-6 aliphatic; phenyl; a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur; and a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur; and n is 1, 2, 3, or 4.


In certain embodiments, the present disclosure provides a compound of formula I, wherein:

    • R1 is RA, wherein RA is an optionally substituted 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur (particularly imidazole or pyrazole, including 1-imidazole or 4-pyrazole);
    • R2 is RB, wherein RB is a substituted 4- to 7-membered saturated heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur (particularly pyrrolidine, piperidine, morpholine, and piperazine), e.g., substituted with one or two groups selected from halogen, ═O, ═CH2, C1-4alkyl, C1-4haloalkyl, C3-6cycloalkyl, —OH, —(CH2)1-2—OH, —OC1-4alkyl, —(CH2)0-2CO2H, —(CH2)0-2O(CH2)1-5CO2H, —(CH2)0-2O(CH2)1-5CO2C1-4alkyl, —(CH2)0-2CO2C1-4alkyl, —NH2, —(CH2)0-2CONH2, —(CH2)0-2CONRa—(CH2)1-2CO2H, —(CH2)0-2SO3H, —(CH2)0-2SO2NH2, —(CH2)0-2SO2NHC1-4alkyl, or —(CH2)0-2NRaSO2C1-4alkyl, wherein Ra, independently for each occurrence, is H or C1-4alkyl; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In other embodiments, the present disclosure provides a compound of formula I, wherein:

    • R1 is RA, wherein RA is an optionally substituted 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur (particularly imidazole or pyrazole, including 1-imidazole or 4-pyrazole);
    • R2 is —NR2, wherein one R is H or lower alkyl (particularly methyl), and the other R is substituted C1-6 aliphatic, including optionally substituted —C1-5alkylene-X; wherein X is selected from —OH, —OC1-4alkyl, —O—C1-4alkylene-CO2H, —O—C1-4alkylene-CO2C1-4alkyl, —CO2H, —CO2C1-4alkyl, —NH2, —CONH2, —CONHC1-4alkyl, —CONRa—(CH2)1-2—OH, —CONRa—(CH2)1-2—COOH, —CONRa—(CH2)1-2—CONH2, —SO3H, —SO2NH2, —SO2NHC1-4alkyl, —NRaSO2C1-4alkyl, —P(O)(OH)2, and —OP(O)(H)(OH), wherein Ra, independently for each occurrence, is H or C1-4alkyl; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In some embodiments, the present disclosure provides a compound of formula I, wherein:

    • R1 is —NR2, wherein one R is H or lower alkyl (particularly methyl), and the other R is substituted C1-6 aliphatic, including optionally substituted —C1-4alkylene-X; wherein X is selected from —OH, —OC1-4alkyl, —CO2H, —O—C1-4alkylene-CO2H, —O—C1-4alkylene-CO2C1-4alkyl, —CO2C1-4alkyl, —CONH2, and —CONHC1-4alkyl;
    • R2 is RB, wherein RB is an optionally substituted 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur (particularly imidazole or pyrazole, including 1-imidazole or 4-pyrazole); and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In other embodiments, the present disclosure provides a compound of formula I, wherein:

    • R1 is RA, wherein RA is an optionally substituted 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur (particularly imidazole or pyrazole, including 1-imidazole or 4-pyrazole);
    • R2 is substituted phenyl (particularly substituted with one or two groups selected from halogen, C1-4alkyl, —OH, —OC1-4alkyl, —CO2H, —CO2C1-4alkyl, —(CH2)0-2O(CH2)1-5CO2H, —(CH2)0-2O(CH2)1-5CO2C1-4alkyl, —NH2, and —CONH2); and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In other embodiments, the present disclosure provides a compound of formula I, wherein:

    • R1 is RA, wherein RA is an optionally substituted 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur (particularly imidazole or pyrazole, including 1-imidazole or 4-pyrazole);
    • R2 is RB, wherein RB is an optionally substituted C1-6 aliphatic, preferably a substituted C1-6aliphatic (particularly —(CR2)1-4NR(CR2)0-6CO2R); and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, the disclosure provides a compound, wherein the compound is of formula I*:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • R1 is




embedded image




    •  or an optionally substituted 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur;

    • R2 is —NRaR5,







embedded image




    • each R3 is independently halogen, —OR, —NR2, —SR, or RC;

    • Ring B1 is phenyl, preferably substituted phenyl; a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur;

    • Ring B2 is phenyl; a 4- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur;

    • each R is independently hydrogen or an optionally substituted group selected from C1-6 aliphatic; benzyl; phenyl; a 4- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; and a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur,

    • R4 is hydrogen or an optionally substituted C1-6 aliphatic;

    • R5 is —(CR2)0-4OR, —(CR2)0-5CO2R, —(CR2)0-5CONR2, —(CR2)0-4C(O)NR(CR2)0-4CO2R, —(CR2)0-4C(O)NR(CR2)0-4CONR2, —(CR2)0-4NRC(O)R, —(CR2)0-4SO3R, —(CR2)0-4SO2NR2, —(CR2)0-4OSO2NR2, —(CR2)0-4NRSO2R, —(CR2)0-4NRSO2OR, —(CR2)0-4OP(OR)2, —(CR2)0-4OP(O)(OR)2, —(CR2)0-4P(O)(OR)2, —(CR2)0-4OP(O)(H)OR, or RB;

    • each R6 is independently halogen, —COR, —(CR2)0-4CO2R, —(CR2)0-4CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-4SO3R, —(CR2)0-4SO2NR2, —(CR2)0-4OSO2NR2, —(CR2)0-4NRSO2R, —(CR2)0-4NRSO2OR, —(CR2)0-4OP(OR)2, —(CR2)0-4OP(O)(OR)2, —(CR2)0-4P(O)(OR)2, —(CR2)0-4OP(O)(H)OR, —B(OR)2, or RB;

    • RB and RC, independently, are an optionally substituted group selected from C1-6 aliphatic; phenyl; a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; and a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur;

    • each Ra is independently H or C1-6alkyl;

    • each m is 0, 1, 2, 3, or 4;

    • n is 1, 2, 3, or 4;

    • q is 0, 1, or 2.





In some embodiments, R1 is an optionally substituted 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.


In some embodiments, R1 is imidazole. In some embodiments, the imidazole is unsubstituted. In some embodiments, R1 is




embedded image


In some embodiments, R1 is pyrazole. In some embodiments, the pyrazole is unsubstituted. In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R4 is C1-6 alkyl.


In some embodiments, at least one occurrence of R3 is halogen. In some embodiments, at least one occurrence of R3 is chloro. In some embodiments, n is 2 and both R3 are chloro. In some embodiments, n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In some embodiments, R2 is —NRaR.


In some embodiments, R5 is —(CR2)2-4OR, —(CR2)0-5CO2R, —(CR2)0-5CONR2, —(CR2)0-4C(O)NR(CR2)0-4CO2R, —(CR2)0-4C(O)NR(CR2)0-4CONR2, —(CR2)0-4NRC(O)R, —(CR2)0-4SO3R, —(CR2)0-4SO2NR2, —(CR2)0-4OSO2NR2, —(CR2)0-4NRSO2R, —(CR2)0-4NRSO2OR, —(CR2)0-4OP(OR)2, —(CR2)0-4OP(O)(OR)2, —(CR2)0-4P(O)(OR)2, or —(CR2)0-4OP(O)(H)OR.


In some embodiments, R5 is —(CR2)2-4OR, —(CR2)1-5CO2R, —(CR2)1-5CONR2, —(CR2)0-4C(O)NR(CR2)0-4CO2R, —(CR2)0-4C(O)NR(CR2)0-4CONR2, —(CR2)0-4NRC(O)R, —(CR2)0-4SO3R, —(CR2)0-4SO2NR2, —(CR2)0-4OSO2NR2, —(CR2)0-4NRSO2R, —(CR2)0-4NRSO2OR, —(CR2)0-4OP(OR)2, —(CR2)0-4OP(O)(OR)2, —(CR2)0-4P(O)(OR)2, or —(CR2)0-4OP(O)(H)OR.


In some embodiments, R5 is —(CH2)2-4OH, —(CH2)2-4O(CH2)1-5CO2H, —(CH2)2-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-3CH(CH2OH)2, —(CH2)2-4OC1-4alkyl, —(CH2)1-5CO2H, —(CH2)1-5CO2C1-4alkyl, —(CH2)1-4CONH2, —(CH2)1-4CONHC1-4alkyl, —(CH2)1-4CON(C1-4alkyl)2, —(CH2)1-4C(O)NRa(CH2)1-4CO2H, —(CH2)1-4C(O)NRa(CH2)1-4CONH2, —(CH2)1-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)1-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —(CH2)0-4NRaC(O)(CH2)1-5CO2H, —(CH2)0-4NRaC(O)(CH2)1-5CO2C1-4alkyl, —(CH2)2-4SO3H, —(CH2)2-4SO3C1-4alkyl, —(CH2)2-4SO2NH2, —(CH2)2-4SO2NHC1-4alkyl, —(CH2)2-4SO2N(C1-4alkyl)2, —(CH2)2-4OSO2NH2, —(CH2)2-4NRaSO2C1-4alkyl, —(CH2)2-4NRaSO3H, —(CH2)1-4OP(OH)2, —(CH2)1-4P(O)(OH)2, —(CH2)1-4P(O)(OH)(OC1-4alkyl), or —(CH2)2-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-6alkyl.


In some embodiments, R5 is —(CH2)2-4OH, —(CH2)2-4O(CH2)1-5CO2H, —(CH2)2-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-3CH(CH2OH)2, —(CH2)1-5CO2H, —(CH2)1-5CONH2, —(CH2)1-4C(O)NRa(CH2)1-4CO2H, —(CH2)1-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4NRaC(O)(CH2)1-5CO2H, —(CH2)0-4NRaC(O)(CH2)1-5CO2C1-4alkyl, —(CH2)2-4SO3H, —(CH2)2-4SO2NH2, —(CH2)2-4SO2NRaC1-4alkyl, —(CH2)2-4NRaSO2C1-4alkyl, —(CH2)1-4P(O)(OH)2, or —(CH2)2-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl.


In some embodiments, R2 is




embedded image


In some embodiments, m is 1, 2, 3, or 4.


In some embodiments, Ring B1 is substituted phenyl. In some embodiments, the phenyl is substituted with one or more carboxyl groups.


In some embodiments, Ring B1 is a 3-carboxyphenyl group that is optionally substituted with one or more R6 groups.


In some embodiments, Ring B1 is a 3,5-dicarboxyphenyl group that is optionally substituted with one or more R6 groups.


In some embodiments, Ring B1 is a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carbocyclic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur.


In some embodiments, Ring B1 is a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.


In some embodiments, R2 is




embedded image


wherein X is absent (i.e., a bond in the ring), —CR2—, —NR—, —O—, —S—, or —SO2—; and p is 0, 1, or 2. In some embodiments, X is —CR2—, —NR—, or —O—. In some embodiments, p is 1. In some embodiments, m is 1, 2, 3, or 4.


In some embodiments, each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, C3-6cycloalkyl, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CO2C1-4haloalkyl, —(CH2)0-4CONH2, —(CH2)0-4CONHC1-4alkyl, —(CH2)0-4CON(C1-4alkyl)2, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-4OC(O)C1-4alkyl, —NH2, —(CH2)1-4NH2, —(CH2)0-4NHC1-4alkyl, —(CH2)0-4N(C1-4alkyl)2, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4SO2NHC1-4alkyl, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, —(CH2)0-4OP(OH)(OC1-4alkyl), or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl.


In some embodiments, each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, C3-6cycloalkyl, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-4OC(O)C1-4alkyl, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0- 4OP(OH)2, or —(CR2)0-4OP(O)(H)OH.


In some embodiments, R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, C3-6cycloalkyl, —(CH2)0-2CO2H, —(CH2)0-2CO2C1-4alkyl, —(CH2)1-4CO(N-proline), —(CH2)0-2C(O)NRa(CH2)1-4CO2H, —OH, —(CH2)1-2OH, —(CH2)0-2OC1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-2OC(O)C1-4alkyl, —(CH2)0-2SO3H, —(CH2)0-2SO2NH2, —(CH2)0-2NRaSO2C0-2alkyl, —(CH2)0-2OP(OH)2, or —(CR2)0-2OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl.


In some embodiments, each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, C3-6cycloalkyl, —(CH2)1-4CO2H, —(CH2)1-4CO(N-proline), —(CH2)1-4C(O)NRa(CH2)1-4CO2H, —OH, —(CH2)1-4OH, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)1-4SO3H, —(CH2)1-4SO2NH2, —(CH2)1-4NRaSO2C1-4alkyl, —(CH2)1-4OP(OH)2, or —(CR2)1-4OP(O)(H)OH.


In some embodiments, each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, —CO2H, —CO2C1-4alkyl, —CO(N-proline), —CH2CO2H, —OH, —OC1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —OC(O)C1-4alkyl, —SO3H, —SO2NH2, —NRaSO2C1-4alkyl, —OP(OH)2, or —OP(O)(H)OH.


In some embodiments, R2 is




embedded image


In some embodiments, m is 1, 2, 3, or 4. In some embodiments, m is 1 or 2.


In some embodiments, each R6 is independently halogen, ═CH2, ═O, —C1-4alkyl, —C1-4haloalkyl, C3-6cycloalkyl, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CO2C1-4haloalkyl, —(CH2)0-4CONH2, —(CH2)0-4CONHC1-4alkyl, —(CH2)0-4CON(C1-4alkyl)2, —(CH2)0-4CO(N-proline), —(CH2)0-4CO(N-pyrrolidine-3-carboxylic acid), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa—CH(C1-4alkyl)-CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-4OC(O)C1-4alkyl, —NH2, —(CH2)1-4NH2, —(CH2)0-4NHC1-4alkyl, —(CH2)0-4NRaC(O)C1-4alkyl, —(CH2)0-4NRaC(O)Ph, —(CH2)0-4NRaCO(CH2)1-4OH, —(CH2)0-4N(C1-4alkyl)2, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4SO2NRaC1-4alkyl, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4NRaSO2Ph, —(CH2)0-4NRaSO2(CH2)1-4CO2H, —(CH2)0-4OP(OH)2, —(CH2)0-4OP(OH)(OC1-4alkyl), or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-6alkyl.


In some embodiments, each R6 is independently halogen, ═CH2, ═O, —C1-4alkyl, —C1-4haloalkyl, C3-6cycloalkyl, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CO2C1-4haloalkyl, —(CH2)0-4CO(N-proline), —(CH2)0-4CO(N-pyrrolidine-3-carboxylic acid), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa—CH(C1-4alkyl)-CO2H, —OH, —(CH2)1-4OH, —OC1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-4NRaC(O)C1-4alkyl, —(CH2)0-4NRaC(O)Ph, —(CH2)0-4NRaCO(CH2)1-4OH, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4NRaSO2Ph, —(CH2)0-4NRaSO2(CH2)1-4CO2H, —(CH2)0-4OP(OH)2, or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl.


In some embodiments, each R6 is independently halogen, ═CH2, ═O, —C1-4alkyl, —C1-4haloalkyl, —CO2H, —CO2C1-4alkyl, —CO2C1-4haloalkyl, —CO(N-proline), —CO(N-pyrrolidine-3-carboxylic acid), —C(O)NRa(CH2)1-4CO2H, —C(O)NRa—CH(C1-4alkyl)-CO2H, —OH, —OC1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —NRaC(O)C1-4alkyl, —NRaC(O)Ph, —NRaCO(CH2)1-4OH, —SO3H, —SO2NH2, —NRaSO2C1-4alkyl, —NRaSO2Ph, —NRaSO2(CH2)1-4CO2H, —OP(OH)2, or —OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl.


In some embodiments, each R6 is independently fluoro, ═O, ═CH2, methyl, cyclohexyl, morpholinyl, phenyl, —CF3, —OMe, —OtBu, —CO2H, —CO2C1-4alkyl, —CO2C1-4haloalkyl, —(CH2)1-4O(CH2)1-5CO2H, —(CH2)1-4O(CH2)1-5CO2C1-4alkyl, or —OH.


In some embodiments, Ring B1 is a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen and sulfur, wherein a carbon atom of the 5- to 6-membered heteroaryl ring is directly bonded to the carbon at the 2-position of the quinolone ring;


In some embodiments, Ring B1 is imidazole or pyrazole. In some embodiments, the imidazole or pyrazole are unsubstituted.


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, m is 1, 2, 3, or 4.


In some embodiments, Ring B2 is phenyl.


In some embodiments, Ring B2 is a 4- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur.


In some embodiments, Ring B2 is a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.


In some embodiments, R2 is




embedded image


wherein X is absent (i.e., a bond in the ring), —CR2—, —NR—, —O—, —S—, or —SO2—; and m is 1, 2, 3, or 4. In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, X is —CR2—, —NR—, or —O—. In some embodiments, q is 1 or 2. In some embodiments, q is 1. In some embodiments, m is 1 or 2.


In some embodiments, each R6 is independently halogen, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CONH2, —(CH2)0-4CONHC1-4alkyl, —(CH2)0-4CON(C1-4alkyl)2, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —NH2, —(CH2)1-4NH2, —(CH2)0-4NHC1-4alkyl, —(CH2)0-4N(C1-4alkyl)2, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4SO2NHC1-4alkyl, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, —(CH2)0-4OP(OH)(OC1-4alkyl), or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl.


In some embodiments, each R6 is independently halogen, —(CH2)0-4CO2H, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —OH, —(CH2)1-4OH, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl.


In some embodiments, each R6 is independently halogen, —CO2H, —CO(N-proline), —C(O)NRa(CH2)1-4CO2H, —OH, —SO3H, —SO2NH2, —NRaSO2C1-4alkyl, —OP(OH)2, or —OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl.


In some embodiments, R6 is not RB.


In some embodiments, m is at least 1, and at least one R6 includes a terminal —CO2H or —CO2C1-4alkyl group.


In some embodiments, at least one R6 is —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-4CO2H or —(CH2)0-4CO2C1-4alkyl.


In some embodiments, the present disclosure provides a pharmaceutical composition comprising a compound of formula I* or a pharmaceutically acceptable salt thereof.


In some embodiments, the present disclosure provides a method of antagonizing cyclic GMP-AMP synthase (cGAS) in a patient in need thereof, comprising administering an effective amount of a compound of formula I* or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.


In some embodiments, the present disclosure provides a method of treating an inflammatory, allergic, or autoimmune disease in a patient in need thereof, comprising administering an effective amount of a compound of formula I* or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.


In some embodiments, the present disclosure provides a compound of formula I or formula I*,




embedded image


wherein R1 is thereby forming a compound of formula I-a-1:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of R2, R3, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-a-1, wherein R is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, the present disclosure provides a compound of formula I-a-1, wherein R2 is RB, wherein RB is an optionally substituted C1-6 aliphatic, preferably a substituted C1-6 aliphatic, such as —(CR2)1-4NR(CR2)0-6CO2R, —(CR2)1-4O(CR2)0-6CO2R, —(CR2)1-4CO2R, —(CR2)1-4OH, —(CR2)1-4NH2, —(CR2)1-4C(O)NH2, —(CR2)1-4C(O)NHC1-4alkyl, or —(CR2)1-4C(O)N(C1-4alkyl)2, particularly —(CR2)1-4NR(CR2)0-6CO2R or —(CR2)1-4CO2R. In some embodiments, R2 is —(CH2)1-4NRa(CH2)0-4CO2Ra, such as —(CH2)1-4NRa(CH2)0-4CO2H, wherein Ra, independently for each occurrence, is H or C1-4alkyl. In other embodiments, R2 is —(CH2)1-4N(SO2C1-4alkyl)(CH2)0-4CO2Ra, such as is —(CH2)1-4N(SO2C1-4alkyl)(CH2)0-4CO2H, wherein Ra, independently for each occurrence, is H or C1-4alkyl. In other embodiments, R2 is —(CR2)1-4CO2R, such as —(CR2)1-4CO2H, including —(CH2)1-4CO2H. In certain embodiments, R2 includes a terminal —CO2H group. For example, in certain instances, R2 includes a terminal —(CH2)0-4CO2H group.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


thereby forming a compound of formula I-a-2:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of R2, R3, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-a-2, wherein R2 is RB, wherein RB is an optionally substituted C1-6 aliphatic, preferably a substituted C1-6 aliphatic, such as —(CR2)1-4NR(CR2)0-6CO2R, —(CR2)1-4O(CR2)0-6CO2R, —(CR2)1-4CO2R, —(CR2)1-4OH, —(CR2)1-4NH2, —(CR2)1-4C(O)NH2, —(CR2)1-4C(O)NHC1-4alkyl, or —(CR2)1-4C(O)N(C1-4alkyl)2, particularly —(CR2)1-4NR(CR2)0-6CO2R or —(CR2)1-4CO2R. In some embodiments, R2 is —(CH2)1-4NRa(CH2)0-4CO2Ra, such as —(CH2)1-4NRa(CH2)0-4CO2H, wherein Ra, independently for each occurrence, is H or C1-4alkyl. In other embodiments, R2 is —(CH2)1-4N(SO2C1-4alkyl)(CH2)0-4CO2R, such as is —(CH2)1-4N(SO2C1-4alkyl)(CH2)0-4CO2H, wherein Ra, independently for each occurrence, is H or C1-4alkyl. In other embodiments, R2 is —(CR2)1-4CO2R, such as —(CR2)1-4CO2H, including —(CH2)1-4CO2H. In certain embodiments, R2 includes a terminal —CO2H group. For example, in certain instances, R2 includes a terminal —(CH2)0-4CO2H group.


In certain embodiments, the present disclosure provides a compound of formula I-a-2, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is —NR2 thereby forming a compound of formula I-a-3:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of R, R2, R3, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-a-3, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


thereby forming a compound of formula I-a-4:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • Ring A is an optionally substituted 4- to 7-membered saturated or partially unsaturated heterocyclic ring or heteroaryl ring, having 0 to 3 heteroatoms (in addition to the nitrogen already depicted in Ring A) independently selected from nitrogen, oxygen, phosphorus, and sulfur;
    • each of R2, R3, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-a-4, wherein R2 is RB, wherein RB is an optionally substituted C1-6 aliphatic, preferably a substituted C1-6 aliphatic, such as —(CR2)1-4NR(CR2)0-6CO2R, —(CR2)1-4O(CR2)0-6CO2R, —(CR2)1-4CO2R, —(CR2)1-4OH, —(CR2)1-4NH2, —(CR2)1-4C(O)NH2, —(CR2)1-4C(O)NHC1-4alkyl, or —(CR2)1-4C(O)N(C1-4alkyl)2, particularly —(CR2)1-4NR(CR2)0-6CO2R or —(CR2)1-4CO2R. In some embodiments, R2 is —(CH2)1-4NRa(CH2)0-4CO2Ra, such as —(CH2)1-4NRa(CH2)0-4CO2H, wherein Ra, independently for each occurrence, is H or C1-4alkyl. In other embodiments, R2 is —(CH2)1-4N(SO2C1-4alkyl)(CH2)0-4CO2Ra, such as is —(CH2)1-4N(SO2C1-4alkyl)(CH2)0-4CO2H, wherein Ra, independently for each occurrence, is H or C1-4alkyl. In other embodiments, R2 is —(CR2)1-4CO2R, such as —(CR2)1-4CO2H, including —(CH2)1-4CO2H. In certain embodiments, R2 includes a terminal —CO2H group. For example, in certain instances, R2 includes a terminal —(CH2)0-4CO2H group.


In certain embodiments, the present disclosure provides a compound of formula I-a-4, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


thereby forming a compound of formula I-a-5:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • R4 is hydrogen or an optionally substituted C1-6 aliphatic; and
    • each of R, R2, R, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-a-5, wherein R and R4 in




embedded image


independently are hydrogen or lower alkyl, particularly methyl.


In certain embodiments, the present disclosure provides a compound of formula I-a-5, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R is chloro at the 7- and 8-positions with respect to the quinoline ring.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R2 is —NR2 thereby forming a compound of formula I-b-1:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of R, R1, R3, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-b-1, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R2 is —NRR5 thereby forming a compound of formula I-b-2:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • R5 is —(CR2)2-4OR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —(CR2)0-6C(O)NR(CR2)0-6CO2R, —(CR2)0-6C(O)NR(CR2)0-6CONR2, —(CR2)0-4NRC(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or RB; and
    • each of R, R1, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-b-2, wherein R5 is —(CR2)2-4OR, —(CR2)1-4CO2R, —(CR2)1-4CONR2, —(CR2)0-6C(O)NR(CR2)0-6CO2R, —(CR2)0-6C(O)NR(CR2)0-6CONR2, —(CR2)0-4NRC(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or RB.


In some embodiments, the present disclosure provides a compound of formula I-b-2, wherein R5 is not RB.


In some embodiments, the present disclosure provides a compound of formula I-b-2, wherein R in —NRR5 is Ra, which is H or C1-6alkyl.


In certain embodiments, the present disclosure provides a compound of formula I-b-2, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, the present disclosure provides a compound of formula I-b-2, wherein:

    • R1 is an optionally substituted 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur (particularly imidazole or pyrazole, including 1-imidazole or 4-pyrazole);
    • R in —NRR5 is H or lower alkyl, particularly methyl;
    • R5 is —(CR2)0-6OR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —(CR2)0-6C(O)NR(CR2)0-6CO2R, —(CR2)0-6C(O)NR(CR2)0-6CONR2, —(CR2)0-4NRC(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, or —(CR2)0-6OP(O)(H)OR; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-b-2, wherein:

    • R1 is an optionally substituted imidazole or pyrazole, including 1-imidazole or 4-pyrazole, particularly unsubstituted 1-imidazole or 4-pyrazole;
    • R in —NRR5 is H or lower alkyl, particularly methyl;
    • R5 is —(CH2)2-4OH, —(CH2)2-4O(CH2)1-5CO2H, —(CH2)2-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-3CH(CH2OH)2, —(CH2)2-4OC1-4alkyl, —(CH2)1-4CO2H, —(CH2)1-4CO2C1-4alkyl, —(CH2)1-4CONH2, —(CH2)1-4CONHC1-4alkyl, —(CH2)1-4CON(C1-4alkyl)2, —(CH2)1-4C(O)NRa(CH2)1-4CO2H, —(CH2)1-4C(O)NRa(CH2)1-4CONH2, —(CH2)1-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)1-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —(CH2)0-4NRaC(O)(CH2)1-5CO2H, —(CH2)0-4NRaC(O)(CH2)1-5CO2C1-4alkyl, —(CH2)2-4SO3H, —(CH2)2-4SO3C1-4alkyl, —(CH2)2-4SO2NH2, —(CH2)2-4SO2NHC1-4alkyl, —(CH2)2-4SO2N(C1-4alkyl)2, —(CH2)2-4OSO2NH2, —(CH2)2-4NRaSO2C1-4alkyl, —(CH2)2-4NRaSO3H, —(CH2)1-4OP(OH)2, —(CH2)1-4P(O)(OH)2, —(CH2)1-4P(O)(OH)(OC1-4alkyl), or —(CH2)2-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-b-2, wherein:

    • R1 is an optionally substituted imidazole or pyrazole, including 1-imidazole or 4-pyrazole, particularly unsubstituted 1-imidazole or 4-pyrazole;
    • R in —NRR5 is H or lower alkyl, particularly methyl;
    • R5 is —(CH2)2-4OH, —(CH2)2-4O(CH2)1-5CO2H, —(CH2)2-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-3CH(CH2OH)2, —(CH2)1-4CO2H, —(CH2)1-4CONH2, —(CH2)1-4C(O)NRa(CH2)1-4CO2H, —(CH2)1-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4NRaC(O)(CH2)1-5CO2H, —(CH2)0-4NRaC(O)(CH2)1-5CO2C1-4alkyl, —(CH2)2-4SO3H, —(CH2)2-4SO2NH2, —(CH2)2-4SO2NRaC1-4alkyl, —(CH2)2-4NRaSO2C1-4alkyl, —(CH2)1-4P(O)(OH)2, or —(CH2)2-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-b-2, R5 includes a terminal —CO2H group. For example, in certain instances, R5 includes a terminal —(CH2)0-4CO2H group.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R2 is




embedded image


thereby forming a compound of formula I-b-3:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • Ring B1 is phenyl, preferably substituted phenyl; a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur; or a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur;
    • each R6 is independently halogen, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4; and
    • each of R, R1, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-b-3, wherein Ring B1 is phenyl, particularly substituted phenyl. In other embodiments, Ring B1 is not phenyl. For example, Ring B1 may be a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur. In some embodiments, Ring B1 is a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, the present disclosure provides a compound of formula I-b-3, wherein R6 is not RB.


In certain embodiments, the present disclosure provides a compound of formula I-b-3, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-b-3, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R2 is




embedded image


thereby forming a compound of formula I-b-4:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • each R6 is independently halogen, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4; and
    • each of R, R1, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-b-4, wherein R6 is not RB.


In certain embodiments, the present disclosure provides a compound of formula I-b-4, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-b-4, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R2 is




embedded image


thereby forming a compound of formula I-b-5:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • each R6 is independently halogen, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • X is absent (i.e., a bond in the ring), —CR2—, —NR—, —O—, —S—, or —SO2—; particularly —CR2—, —NR—, —O—, —S—, or —SO2—; more particularly —CR2—, —NR—, or —O—;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4;
    • p is 0, 1, or 2, particularly 1; and
    • each of R, R1, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-b-5, wherein R6 is not RB.


In certain embodiments, the present disclosure provides a compound of formula I-b-5, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, the present disclosure provides a compound of formula I-b-5, wherein:

    • R1 is an optionally substituted 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur (particularly imidazole or pyrazole, including 1-imidazole or 4-pyrazole);
    • each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, ═CR2, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or —B(OR)2;
    • m is 1, 2, 3, or 4, particularly 1 or 2;
    • p is 0 or 1, particularly 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-b-5, wherein:

    • R1 is an optionally substituted imidazole or pyrazole, including 1-imidazole or 4-pyrazole, particularly unsubstituted 1-imidazole or 4-pyrazole;
    • each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, ═CH2, ═O, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CONH2, —(CH2)0-4CONHC1-4alkyl, —(CH2)0-4CON(C1-4alkyl)2, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —NH2, —(CH2)1-4NH2, —(CH2)0-4NHC1-4alkyl, —(CH2)0-4N(C1-4alkyl)2, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4SO2NHC1-4alkyl, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, —(CH2)0-4OP(OH)(OC1-4alkyl), or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • X is —CH2—, —NH—, or —O—;
    • m is 1, 2, 3, or 4, particularly 1 or 2;
    • p is 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-b-5, wherein:

    • R1 is an optionally substituted imidazole or pyrazole, including 1-imidazole or 4-pyrazole, particularly unsubstituted 1-imidazole or 4-pyrazole;
    • each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, ═CH2, —(CH2)0-4CO2H, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —OH, —(CH2)1-4OH, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, or —(CR2)0-4OP(O)(H)OH;
    • X is —CH2—, —NH—, or —O—;
    • m is 1 or 2;
    • p is 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-b-5, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R2 is




embedded image


thereby forming a compound of formula I-b-6:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • each R6 is independently halogen, ═O, ═CR2, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4; and
    • each of R, R1, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-b-6, wherein R6 is not RB.


In certain embodiments, the present disclosure provides a compound of formula I-b-6, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, the present disclosure provides a compound of formula I-b-6, wherein:

    • R1 is an optionally substituted 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur (particularly imidazole or pyrazole, including 1-imidazole or 4-pyrazole);
    • each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, ═CR2, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or —B(OR)2;
    • m is 1, 2, 3, or 4, particularly 1 or 2; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-b-6, wherein:

    • R1 is an optionally substituted imidazole or pyrazole, including 1-imidazole or 4-pyrazole, particularly unsubstituted 1-imidazole or 4-pyrazole;
    • each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, ═CH2, ═O, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CO2C1-4haloalkyl, —(CH2)0-4CONH2, —(CH2)0-4CONHC1-4alkyl, —(CH2)0-4CON(C1-4alkyl)2, —(CH2)0-4CO(N-proline), —(CH2)0-4CO(N-pyrrolidine-3-carboxylic acid), —(CH2)0-4(N-pyrazole)-(CH2)0-4CO2H, —(CH2)0-4CO(N-pyrazole)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4(N-pyrrole)-(CH2)0-4CO2H, —(CH2)0-4CO(N-pyrrole)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4—O(phenyl)-(CH2)0-4CO2H, —(CH2)0-4—O(phenyl)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4—O(cycloalkane)-(CH2)0-4CO2H, —(CH2)0-4—O(cycloalkane)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa—CH(C1-4alkyl)-CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)1-4O(CH2)1-4CHFCO2H, —(CH2)1-4O(CH2)1-4CHFCO2C1-4alkyl, —(CH2)1-4OCH(CH3)(CH2)1-4CO2H, —(CH2)1-4OCH(CH3)(CH2)1-4CO2C1-4alkyl, —(CH2)1-4O(CH2)1-4CH(CH3)CO2H, —(CH2)1-4O(CH2)1-4CH(CH3)CO2C1-4alkyl, —CHOH(CH2)1-4O(CH2)1-5CO2H, —CHOH(CH2)1-4O(CH2)1-5CO2C1-4alkyl, —CHCF3(CH2)1-4O(CH2)1-5CO2H, —CHCF3(CH2)1-4O—(CH2)1-5CO2C1-4alkyl, —(CH2)1-4O(CH2)1-5CONRa2, —(CH2)1-4O(CH2)1-5CONRaOH, —(CH2)1-4O(CH2)1-5CONRaCN, —(CH2)1-4O(CH2)1-5P(O)OH2, —NH2, —(CH2)0-4NRa(CH2)1-4CO2H, —(CH2)0-4NRa(CH2)1-4CO2C1-4alkyl, —(CH2)1-4NH2, —(CH2)0-4NHC1-4alkyl, —(CH2)0-4NRaC(O)C1-4alkyl, —(CH2)0-4NRaC(O)Ph, —(CH2)0-4NRaCO(CH2)1-4OH, —(CH2)0-4N(C1-4alkyl)2, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4SO2NRaC1-4alkyl, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4NRaSO2Ph, —(CH2)0-4NRaSO2(CH2)1-4CO2H, —(CH2)0-4OP(OH)2, —(CH2)0-4OP(OH)(OC1-4alkyl), or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • m is 1, 2, 3, or 4, particularly 1 or 2; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-b-6, wherein:

    • R1 is an optionally substituted imidazole or pyrazole, including 1-imidazole or 4-pyrazole, particularly unsubstituted 1-imidazole or 4-pyrazole;
    • each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, ═CH2, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CO2C1-4haloalkyl, —(CH2)0-4CO(N-proline), —(CH2)0-4CO(N-pyrrolidine-3-carboxylic acid), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa—CH(C1-4alkyl)-CO2H, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-4NRaC(O)C1-4alkyl, —(CH2)0-4NRaC(O)Ph, —(CH2)0-4NRaCO(CH2)1-4OH, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4NRaSO2Ph, —(CH2)0-4NRaSO2(CH2)1-4CO2H, —(CH2)0-4OP(OH)2, or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • m is 1 or 2; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-b-6, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R2 is




embedded image


thereby forming a compound of formula I-b-7:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • Ring B2 is phenyl; a 4- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur; or a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur;
    • each R6 is independently halogen, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4;
    • q is 0, 1, or 2; and
    • each of R, R1, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-b-7, wherein Ring B2 is phenyl, particularly substituted phenyl. In other embodiments, Ring B2 is not phenyl. For example, Ring B2 may be a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur. In some embodiments, Ring B2 is a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, the present disclosure provides a compound of formula I-b-7, wherein R6 is not RB.


In some embodiments, the present disclosure provides a compound of formula I-b-7, wherein R in




embedded image


is Ra, which is H or C1-6alkyl.


In certain embodiments, the present disclosure provides a compound of formula I-b-7, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-b-7, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R2 is




embedded image


thereby forming a compound of formula I-b-8 or formula I-b-8*, respectively:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • each R6 is independently hydrogen (only on N), halogen (not on N), —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • X is absent (i.e., a bond in the ring), —CR2—, —NR—, —O—, —S—, or —SO2—, particularly —CR2—, —NR—, or —O;
    • m is 1, 2, 3, or 4;
    • q is 0, 1, or 2; and
    • each of R, R1, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-b-8 or formula I-b-8*, wherein R6 is not RB.


In some embodiments, the present disclosure provides a compound of formula I-b-8 or formula I-(b-8*, wherein R in




embedded image


or in is Ra, which is H or C1-6alkyl.


In certain embodiments, the present disclosure provides a compound of formula I-b-8 or formula I-b-8*, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, the present disclosure provides a compound of formula I-b-8 or formula I-b-8*, wherein:

    • R1 is an optionally substituted 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur (particularly imidazole or pyrazole, including 1-imidazole or 4-pyrazole);
    • each R6 is independently halogen, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or —B(OR)2;
    • m is 1, 2, 3, or 4, particularly 1 or 2;
    • q is 1 or 2; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-b-8 or formula I-b-8*, wherein:

    • R1 is an optionally substituted imidazole or pyrazole, including 1-imidazole or 4-pyrazole, particularly unsubstituted 1-imidazole or 4-pyrazole;
    • each R6 is independently halogen, ═O, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CONH2, —(CH2)0-4CONHC1-4alkyl, —(CH2)0-4CON(C1-4alkyl)2, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —NH2, —(CH2)1-4NH2, —(CH2)0-4NHC1-4alkyl, —(CH2)0-4N(C1-4alkyl)2, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4SO2NHC1-4alkyl, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, —(CH2)0-4OP(OH)(OC1-4alkyl), or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • m is 1, 2, 3, or 4, particularly 1 or 2;
    • q is 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-b-8 or formula I-b-8*, wherein:

    • R1 is an optionally substituted imidazole or pyrazole, including 1-imidazole or 4-pyrazole, particularly unsubstituted 1-imidazole or 4-pyrazole;
    • each R6 is independently halogen, —(CH2)0-4CO2H, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —OH, —(CH2)1-4OH, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • m is 1 or 2;
    • q is 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-b-8 or I-b-8*, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R2 is




embedded image


thereby forming a compound of formula I-b-9:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of R, R1, R3, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-b-9, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, the present disclosure provides a compound of formula I-b-9, wherein:

    • R1 is




embedded image




    •  wherein R and R4 independently are hydrogen or an optionally substituted C1-6 aliphatic; and

    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.





In particular embodiments, the present disclosure provides a compound of formula I-b-9, wherein:

    • R1 is




embedded image




    •  wherein R and R4 independently are hydrogen or lower alkyl, particularly methyl; and

    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R is chloro at the 7- and 8-positions with respect to the quinoline ring.





In further particular embodiments, the present disclosure provides a compound of formula I-b-9, wherein:

    • R1 is




embedded image




    •  wherein R is hydrogen and R4 is hydrogen or lower alkyl, particularly methyl; and

    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R is chloro at the 7- and 8-positions with respect to the quinoline ring.





In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein at least one R3 is halogen, such as chloro at the 8-position, thereby forming a compound of formula I-c-1:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of R1, R2, and R is as defined above and described in embodiments herein, and n is 0, 1, 2, or 3, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein n is 2 and R is chloro at the 7- and 8-positions, thereby forming a compound of formula I-c-2:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of R1 and R2 is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is —NR2, thereby forming a compound of formula I-d-1:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of R, R3, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-d-1, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is —NRR5, thereby forming a compound of formula I-d-2:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • R5 is —(CR2)2-4OR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —(CR2)0-6C(O)NR(CR2)0-6CO2R, —(CR2)0-6C(O)NR(CR2)0-6CONR2, —(CR2)0-4NRaC(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or RB; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-d-2, wherein R5 is —(CR2)2-4OR, —(CR2)1-4CO2R, —(CR2)1-4CONR2, —(CR2)0-6C(O)NR(CR2)0-6CO2R, —(CR2)0-6C(O)NR(CR2)0-6CONR2, —(CR2)0-4NRaC(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or RB.


In some embodiments, the present disclosure provides a compound of formula I-d-2, wherein R5 is not RB.


In some embodiments, the present disclosure provides a compound of formula I-d-2, wherein R in —NRR5 is Ra, which is H or C1-6alkyl.


In certain embodiments, the present disclosure provides a compound of formula I-d-2, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-d-2, wherein:

    • R in —NRR5 is H or lower alkyl, particularly methyl;
    • R5 is —(CH2)2-4OH, —(CH2)2-4O(CH2)1-5CO2H, —(CH2)2-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-3CH(CH2OH)2, —(CH2)2-4OC1-4alkyl, —(CH2)1-4CO2H, —(CH2)1-4CO2C1-4alkyl, —(CH2)1-4CONH2, —(CH2)1-4CONHC1-4alkyl, —(CH2)1-4CON(C1-4alkyl)2, —(CH2)1-4C(O)NRa(CH2)1-4CO2H, —(CH2)1-4C(O)NRa(CH2)1-4CONH2, —(CH2)1-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)1-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —(CH2)0-4NRaC(O)(CH2)1-5CO2H, —(CH2)0-4NRaC(O)(CH2)1-5CO2C1-4alkyl, —(CH2)2-4SO3H, —(CH2)2-4SO3C1-4alkyl, —(CH2)2-4SO2NH2, —(CH2)2-4SO2NHC1-4alkyl, —(CH2)2-4SO2N(C1-4alkyl)2, —(CH2)2-4OSO2NH2, —(CH2)2-4NRaSO2C1-4alkyl, —(CH2)2-4NRaSO3H, —(CH2)1-4OP(OH)2, —(CH2)1-4P(O)(OH)2, —(CH2)1-4P(O)(OH)(OC1-4alkyl), or —(CH2)2-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-d-2, wherein:

    • R in —NRR5 is H or lower alkyl, particularly methyl;
    • R5 is —(CH2)2-4OH, —(CH2)2-4O(CH2)1-5CO2H, —(CH2)2-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-3CH(CH2OH)2, —(CH2)1-4CO2H, —(CH2)1-4CONH2, —(CH2)1-4C(O)NRa(CH2)1-4CO2H, —(CH2)1-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4NRaC(O)(CH2)1-5CO2H, —(CH2)0-4NRaC(O)(CH2)1-5CO2C1-4alkyl, —(CH2)2-4SO3H, —(CH2)2-4SO2NH2, —(CH2)2-4SO2NRaC1-4alkyl, —(CH2)2-4NRaSO2C1-4alkyl, —(CH2)1-4P(O)(OH)2, or —(CH2)2-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-d-2, R5 includes a terminal —CO2H group. For example, in certain instances, R5 includes a terminal —(CH2)0-4CO2H group.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-d-3:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • Ring B1 is phenyl, preferably substituted phenyl; a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur; or a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur;
    • each R6 is independently halogen, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-d-3, wherein Ring B1 is phenyl, particularly substituted phenyl. In other embodiments, Ring B1 is not phenyl. For example, Ring B1 may be a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur. In some embodiments, Ring B1 is a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, the present disclosure provides a compound of formula I-d-3, wherein R6 is not RB.


In certain embodiments, the present disclosure provides a compound of formula I-d-3, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-d-3, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-d-4:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • each R6 is independently halogen, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-d-4, wherein R6 is not RB.


In certain embodiments, the present disclosure provides a compound of formula I-d-4, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-d-4, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-d-5:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • each R6 is independently halogen, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • X is —CR2—, —NR—, —O—, —S—, or —SO2—, particularly —CR2—, —NR—, or —O;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4;
    • p is 0, 1, or 2, particularly 1; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-d-5, wherein R6 is not RB.


In certain embodiments, the present disclosure provides a compound of formula I-d-5, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-d-5, wherein:

    • each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, ═CH2, ═O, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)1-4O(CH2)1-4CHFCO2H, —(CH2)1-4O(CH2)1-4CHFCO2C1-4alkyl, —(CH2)1-4OCH(CH3)(CH2)1-4CO2H, —(CH2)1-4OCH(CH3)(CH2)1-4CO2C1-4alkyl, —(CH2)1-4O(CH2)1-4CH(CH3)CO2H, —(CH2)1-4O(CH2)1-4CH(CH3)CO2C1-4alkyl, —CHOH(CH2)1-4O(CH2)1-5CO2H, —CHOH(CH2)1-4O(CH2)1-5CO2C1-4alkyl, —CHCF3(CH2)1-4O(CH2)1-5CO2H, —CHCF3(CH2)1-4O—(CH2)1-5CO2C1-4alkyl, —(CH2)1-4O(CH2)1-5CONRa2, —(CH2)1-4O(CH2)1-5CONRaOH, —(CH2)1-4O(CH2)1-5CONRaCN, —(CH2)1-4O(CH2)1-5P(O)OH2, —(CH2)0-4CONH2, —(CH2)0-4CONHC1-4alkyl, —(CH2)0-4CON(C1-4alkyl)2, —(CH2)0-4CO(N-proline), —(CH2)0-4(N-pyrazole)-(CH2)0-4CO2H, —(CH2)0-4CO(N-pyrazole)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4(N-pyrrole)-(CH2)0-4CO2H, —(CH2)0-4CO(N-pyrrole)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4—O(phenyl)-(CH2)0-4CO2H, —(CH2)0-4—O(phenyl)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4—O(cycloalkane)-(CH2)0-4CO2H, —(CH2)0-4—O(cycloalkane)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —NH2, —(CH2)0-4NRa(CH2)1-4CO2H, —(CH2)0-4NRa(CH2)1-4CO2C1-4alkyl, —(CH2)0-4N[(CH2)1- 4CO2C1-4alkyl]2, —(CH2)1-4NH2, —(CH2)0-4NHC1-4alkyl, —(CH2)0-4N(C1-4alkyl)2, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4SO2NHC1-4alkyl, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, —(CH2)0-4OP(OH)(OC1-4alkyl), or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • X is —CH2—, —NH—, or —O—;
    • m is 1, 2, 3, or 4, particularly 1 or 2;
    • p is 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-b-5, wherein:

    • each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, ═CH2, —(CH2)0-4CO2H, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)1-4O(CH2)0-5CO2C1-4alkyl, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, or —(CR2)0-4OP(O)(H)OH;
    • X is —CH2—, —NH—, or —O—;
    • m is 1 or 2;
    • p is 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-b-5, wherein each R6 is independently fluoro, -methyl, -methoxy, —(CH2)1-5CO2H, —(CH2)1-5CO2C1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, or —OH; and m is 1, 2, or 3, particularly 1 or 2.


In certain embodiments, for the above disclosed compounds of formula I-d-5, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-d-6:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • each R6 is independently halogen, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-d-6, wherein R6 is not RB.


In certain embodiments, the present disclosure provides a compound of formula I-d-6, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, the present disclosure provides a compound of formula I-d-6, wherein:

    • each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, —C3-6cycloalkyl, ═CR2, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or —B(OR)2;
    • m is 1, 2, 3, or 4, particularly 1 or 2; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-d-6, wherein:

    • each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, —C3-6cycloalkyl, ═CH2, ═O, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CO2C1-4haloalkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)1-4O(CH2)1-4CHFCO2H, —(CH2)1-4O(CH2)1-4CHFCO2C1-4alkyl, —(CH2)1-4OCH(CH3)(CH2)1-4CO2H, —(CH2)1-4OCH(CH3)(CH2)1-4CO2C1-4alkyl, —(CH2)1-4O(CH2)1-4CH(CH3)CO2H, —(CH2)1-4O(CH2)1-4CH(CH3)CO2C1-4alkyl, —CHOH(CH2)1-4O(CH2)1-5CO2H, —CHOH(CH2)1-4O(CH2)1-5CO2C1-4alkyl, —CHCF3(CH2)1-4O(CH2)1-5CO2H, —CHCF3(CH2)1-4O—(CH2)1-5CO2C1-4alkyl, —(CH2)1-4O(CH2)1-5CONRa2, —(CH2)1-4O(CH2)1-5CONRaOH, —(CH2)1-4O(CH2)1-5CONRaCN, —(CH2)1-4O(CH2)1-5P(O)OH2, —(CH2)0-4CONH2, —(CH2)0-4CONHC1-4alkyl, —(CH2)0-4CON(C1-4alkyl)2, —(CH2)0-4CO(N-proline), —(CH2)0-4CO(N-pyrrolidine-3-carboxylic acid), —(CH2)0-4(N-pyrazole)-(CH2)0-4CO2H, —(CH2)0-4CO(N-pyrazole)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4(N-pyrrole)-(CH2)0-4CO2H, —(CH2)0-4CO(N-pyrrole)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4—O(phenyl)-(CH2)0-4CO2H, —(CH2)0-4—O(phenyl)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4-O(cycloalkane)-(CH2)0-4CO2H, —(CH2)0-4-O(cycloalkane)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa—CH(C1-4alkyl)-CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —(CH2)0-4OC(O)C1-4alkyl, —NH2, —(CH2)0-4NRa(CH2)1-4CO2H, —(CH2)0-4NRa(CH2)1-4CO2C1-4alkyl, —(CH2)1-4NH2, —(CH2)0-4NHC1-4alkyl, —(CH2)0-4NRaC(O)C1-4alkyl, —(CH2)0-4NRaC(O)Ph, —(CH2)0-4NRaCO(CH2)1-4OH, —(CH2)0-4N(C1-4alkyl)2, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4SO2NRaC1-4alkyl, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4NRaSO2Ph, —(CH2)0-4NRaSO2(CH2)1-4CO2H, —(CH2)0-4OP(OH)2, —(CH2)0-4OP(OH)(OC1-4alkyl), or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • m is 1, 2, 3, or 4, particularly 1 or 2; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-d-6, wherein:

    • each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, —C3-6cycloalkyl, ═CH2, —(CH2)0-4CO2H, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-4CO(N-proline), —(CH2)0-4CO(N-pyrrolidine-3-carboxylic acid), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa—CH(C1-4alkyl)-CO2H, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —(CH2)0-4OC(O)C1-4alkyl, —(CH2)0-4NRaC(O)C1-4alkyl, —(CH2)0-4NRaC(O)Ph, —(CH2)0-4NRaCO(CH2)1-4OH, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4NRaSO2Ph, —(CH2)0-4NRaSO2(CH2)1-4CO2H, —(CH2)0-4OP(OH)2, or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • m is 1 or 2; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-d-6, wherein each R6 is independently fluoro, -methyl, cyclohexyl, -methoxy, —OtBu, —CF3, —(CH2)1-5CO2H, —(CH2)1-5CO2C1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, or —OH; and m is 1, 2, or 3, particularly 1 or 2.


In certain embodiments, for the above disclosed compounds of formula I-d-6, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-d-7:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • Ring B2 is phenyl; a 4- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur; or a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur;
    • each R6 is independently halogen, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4;
    • q is 0, 1, or 2; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-d-7, wherein Ring B2 is phenyl, particularly substituted phenyl. In other embodiments, Ring B2 is not phenyl. For example, Ring B2 may be a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur. In some embodiments, Ring B2 is a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, the present disclosure provides a compound of formula I-d-7, wherein R6 is not RB.


In some embodiments, the present disclosure provides a compound of formula I-d-7, wherein R in




embedded image


is Ra, which is H or C1-6alkyl.


In certain embodiments, the present disclosure provides a compound of formula I-d-7, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-d-7, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-d-8 or formula I-d-8*, respectively:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • each R6 is independently hydrogen (only on N), halogen (not on N), —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • X is —CR2—, —NR—, —O—, —S—, or —SO2—, particularly —CR2—, —NR—, or —O;
    • m is 1, 2, 3, or 4;
    • q is 0, 1, or 2; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-d-8 or formula I-d-8*, wherein R6 is not RB.


In some embodiments, the present disclosure provides a compound of formula I-d-8 or formula I-d-8*, wherein R in




embedded image


is Ra, which is H or C1-6alkyl.


In certain embodiments, the present disclosure provides a compound of formula I-d-8 or formula I-d-8*, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, the present disclosure provides a compound of formula I-d-8 or formula I-d-8*, wherein:

    • each R6 is independently halogen, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or —B(OR)2;
    • m is 1, 2, 3, or 4, particularly 1 or 2;
    • q is 1 or 2; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-d-8 or formula I-d-8*, wherein:

    • each R6 is independently halogen, ═O, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CONH2, —(CH2)0-4CONHC1-4alkyl, —(CH2)0-4CON(C1-4alkyl)2, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —NH2, —(CH2)1-4NH2, —(CH2)0-4NHC1-4alkyl, —(CH2)0-4N(C1-4alkyl)2, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4SO2NHC1-4alkyl, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, —(CH2)0-4OP(OH)(OC1-4alkyl), or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • m is 1, 2, 3, or 4, particularly 1 or 2;
    • q is 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-d-8 or formula I-d-8*, wherein:

    • each R6 is independently halogen, —(CH2)0-4CO2H, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —OH, —(CH2)1-4OH, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • m is 1 or 2;
    • q is 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-d-8 or I-d-8*, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-d-9:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of R, R1, R3, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-d-9, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-d-10:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • Ring C is pyrrolyl or a 4- to 10-membered saturated or partially unsaturated monocyclic, bicyclic, bridged bicyclic, or spirocyclic heterocyclic ring having 0 to 3 heteroatoms (in addition to the nitrogen already depicted in Ring C) independently selected from nitrogen, oxygen, and sulfur;
    • each R6 is independently halogen, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 1, 2, 3, or 4; and
    • each of R, RB, R3, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-d-10, wherein Ring C is azetidinyl, pyrrolyl, 2,3-dihydro-1H-pyrrolyl, pyrrolidinyl, piperidinyl, morpholinyl, thiazolidinyl, indolinyl, isoindolinyl, octahydroindolyl, azepanyl, an oxazepane, an azabicyclohexane, an azabicycloheptane, an azabicyclooctane, an azabicyclononane, an azaspiroheptane, or an octahydrocyclicpentapyrrole.


In particular embodiments, the present disclosure provides a compound of formula I-d-10, wherein Ring C is pyrrolyl, 2-azabicyclo[3.1.0]hexanyl, 3-azabicyclo[3.1.0]hexane, 5-azaspiro[2.4]heptanyl, or octahydrocyclopenta[b]pyrrolyl.


In particular embodiments, the present disclosure provides a compound of formula I-d-10, wherein Ring C is 3-hydroxyproline, C1-6alkyl 3-hydroxyproline ester, or C1-6haloalkyl 3-hydroxyproline ester.


In some embodiments, the present disclosure provides a compound of formula I-d-10, wherein:

    • each R6 is independently fluoro, —CN, methyl, —C3-6cycloalkyl, —CF3, —CO2H, —NH2, —OH, —OC1-4alkyl, ═CH2, =0, tetrazolyl, imidazoyl, thiophenyl, 1,2,4-oxadiazol-3(2H)-onyl, morpholinyl, phenyl, —(CH2)1-4OC1-4alkyl, —(CH2)1-4O(CH2)1-5CO2H, —(CH2)1-4O(CH2)1-5CO2C1-4alkyl, —(CH2)1-4O(CH2)1-4CHFCO2H, —(CH2)1-4O(CH2)1-4CHFCO2C1-4alkyl, —(CH2)1-4OCH(CH3)(CH2)1-4CO2H, —(CH2)1-4OCH(CH3)(CH2)1-4CO2C1-4alkyl, —(CH2)1-4O(CH2)1-4CH(CH3)CO2H, —(CH2)1-4O(CH2)1-4CH(CH3)CO2C1-4alkyl, —CHOH(CH2)1-4O(CH2)1-5CO2H, —CHOH—(CH2)1-4O(CH2)1-5CO2C1-4alkyl, —CHCF3(CH2)1-4O(CH2)1-5CO2H, —CHCF3(CH2)1-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-4O(CH2)1-5CONRa2, —(CH2)1-4O(CH2)1-5CONRaOH, —(CH2)1-4O(CH2)1-5CONRaCN, —(CH2)1-4O(CH2)1-5P(O)OH2, —CH(OH)CF3, —COH(CF3)2, —(CH2)1-5CO2H, —(CH2)1-5CO2C1-4alkyl, —(CH2)0-4CHCHCO2H, —(CH2)0-4CHCHCO2C1-4alkyl, —CO2C1-4alkyl, —CO2C1-4haloalkyl, —OCOC1-4alkyl, —O(CH2)1-4CO2H, —O(CH2)1-4CO2C1-4alkyl, —OCONRa(CH2)0-4CO2H, —OCONRa(CH2)0-4CO2C1-4alkyl, —OCO—(N-morpholine), —CONH2, —CONHOH, —CONHOC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CO2C1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4(pyrrolid-2-one), —(CH2)0-4C(O)NRa(CH2)1-4(piperidin-2-one), —(CH2)0-4C(O)NRa(CH2)1-4C(OH)(CF3)2, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4CONRaSO2C1-4haloalkyl, —(CH2)0-4CO(N-pyrrolidine)-(CH2)0-4CO2H, —(CH2)0-4CO(N-pyrrolidine)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4(N-pyrazole)-(CH2)0-4CO2H, —(CH2)0-4CO(N-pyrazole)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4(N-pyrrole)-(CH2)0-4CO2H, —(CH2)0-4CO(N-pyrrole)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4—O-(phenyl)-(CH2)0-4CO2H, —(CH2)0-4—O-(phenyl)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4—O(cycloalkane)-(CH2)0-4CO2H, —(CH2)0-4—O(cycloalkane)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CO(N-pyrrolidine)-(CH2)0-4SO2NH2, —(CH2)0-4CO(N-pyrrolidine)-(tetrazole), —(CH2)0-4CO(N-piperdine)-(CH2)0-4CO2H, —(CH2)0-4CO(N-piperdine)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CO(N-morpholine)-(CH2)0-4CO2H, —CONRa(bicyclo[1.1.1]pentane)-(CH2)0-4CO2H, —NH2, —(CH2)0-4NRa(CH2)1-4CO2H, —(CH2)0-4NRa(CH2)1-4CO2C1-4alkyl, —(CH2)0-4N[(CH2)1-4CO2C1-4alkyl]2, —(CH2)0-4NRaCO(isoxazole)-OH, —(CH2)0-4NRaSO2(benzene)-CO2H, —(CH2)0-4NRaSO2(CH2)1-4CO2H, —(CH2)0-4NRaSO2(CH2)1-4CO2C1-4alkyl, —(CH2)0-4NRaSO2(CH2)1-4(imidazolidine-2,4-dione), —(CH2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl; and
    • m is 1, 2, 3, or 4.


In particular embodiments, the present disclosure provides a compound of formula I-d-10, wherein each R6 is independently fluoro, -methyl, cyclohexyl, —CF3, —CO2H, —CO2C1-4alkyl, —CO2C1-4haloalkyl, or —OH; and m is 1, 2, or 3, particularly 1 or 2.


In particular embodiments, the present disclosure provides a compound of formula I-d-10, wherein each R6 is independently fluoro, -methyl, cyclohexyl, —CF3, -methoxy, —(CH2)1-5CO2H, —(CH2)1-5CO2C1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, or —OH; and m is 1, 2, or 3, particularly 1 or 2.


In certain embodiments, the present disclosure provides a compound of formula I-d-10, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


R2 is



embedded image


and R3 is chloro at the 7- and 8-positions thereby forming a compound of formula I-d-11:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • each R6 is independently halogen, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-4OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 1, 2, 3, or 4; and
    • each of R and RB is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-d-11, wherein:

    • each R6 is independently fluoro, —CN, methyl, ═O, ═CH2, —C3-6cycloalkyl, —CF3, —CO2H, —NH2, —OH, —OC1-4alkyl, tetrazolyl, imidazoyl, thiophenyl, 1,2,4-oxadiazol-3(2H)-onyl, morpholinyl, phenyl, —(CH2)1-4OC1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)1-4O(CH2)1-4CHFCO2H, —(CH2)1-4O(CH2)1-4CHFCO2C1-4alkyl, —(CH2)1-4OCH(CH3)(CH2)1-4CO2H, —(CH2)1-4OCH(CH3)(CH2)1-4CO2C1-4alkyl, —(CH2)1-4O(CH2)1-4CH(CH3)CO2H, —(CH2)1-4O(CH2)1-4CH(CH3)CO2C1-4alkyl, —CHOH(CH2)1-4O(CH2)1-5CO2H, —CHOH(CH2)1-4O(CH2)1-5CO2C1-4alkyl, —CHCF3(CH2)1-4O(CH2)1-5CO2H, —CHCF3(CH2)1-4O—(CH2)1-5CO2C1-4alkyl, —(CH2)1-4O(CH2)1-5CONRa2, —(CH2)1-4O(CH2)1-5CONRaOH, —(CH2)1-4O(CH2)1-5CONRaCN, —(CH2)1-4O(CH2)1-5P(O)OH2, —CH(OH)CF3, —COH(CF3)2, —(CH2)1-5CO2H, —(CH2)1-5CO2C1-4alkyl, —(CH2)0-4CHCHCO2H, —(CH2)0-4CHCHCO2C1-4alkyl, —CO2C1-4alkyl, —CO2C1-4haloalkyl, —OCOC1-4alkyl, —O(CH2)1-4CO2H, —O(CH2)1-4CO2C1-4alkyl, —OCONRa(CH2)0-4CO2H, —OCONRa(CH2)0-4CO2C1-4alkyl, —OCO—(N-morpholine), —CONH2, —CONHOH, —CONHOC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CO2C1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4(pyrrolid-2-one), —(CH2)0-4C(O)NRa(CH2)1-4(piperidin-2-one), —(CH2)0-4C(O)NRa(CH2)1-4C(OH)(CF3)2, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4CONRaSO2C1-4haloalkyl, —(CH2)0-4CO(N-pyrrolidine)-(CH2)0-4CO2H, —(CH2)0-4CO(N-pyrrolidine)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4(N-pyrazole)-(CH2)0-4CO2H, —(CH2)0-4CO(N-pyrazole)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4(N-pyrrole)-(CH2)0-4CO2H, —(CH2)0-4CO(N-pyrrole)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4—O(phenyl)-(CH2)0-4CO2H, —(CH2)0-4—O(phenyl)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4—O(cycloalkane)-(CH2)0-4CO2H, —(CH2)0-4—O(cycloalkane)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CO(N-pyrrolidine)-(CH2)0-4SO2NH2, —(CH2)0-4CO(N-pyrrolidine)-(tetrazole), —(CH2)0-4CO(N-piperdine)-(CH2)0-4CO2H, —(CH2)0-4CO—(N-piperdine)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CO(N-morpholine)-(CH2)0-4CO2H, —CONRa(bicyclo[1.1.1]pentane)-(CH2)0-4CO2H, —NH2, —(CH2)0-4NRa(CH2)1-4CO2H, —(CH2)0-4NRa(CH2)1-4CO2C1-4alkyl, —(CH2)0-4N[(CH2)1-4CO2C1-4alkyl]2, —(CH2)0-4NRaCO(isoxazole)OH, —(CH2)0-4NRaSO2(phenyl)-CO2H, —(CH2)0-4NRaSO2(CH2)1-4CO2H, —(CH2)0-4NRaSO2(CH2)1-4CO2C1-4alkyl, —(CH2)0-4NRaSO2(CH2)1-4(imidazolidine-2,4-dione), —(CH2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl; and
    • m is 1, 2, 3, or 4.


In particular embodiments, the present disclosure provides a compound of formula I-d-11, wherein each R6 is independently fluoro, ═O, ═CH2, -methyl, cyclohexyl, —CF3, —CO2H, —CO2C1-4alkyl, —CO2C1-4haloalkyl, or —OH; m is 1, 2, or 3, particularly 1 or 2.


In particular embodiments, the present disclosure provides a compound of formula I-d-11, wherein each R6 is independently fluoro, ═O, ═CH2, -methyl, cyclohexyl, —CF3, -methoxy, —OtBu, —(CH2)1-5CO2H, —(CH2)1-5CO2C1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)1-4O(CH2)1-5CONRa2, or —OH; and m is 1, 2, or 3, particularly 1 or 2.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


R2 is —NRR5, and R3 is chloro at the 7- and 8-positions thereby forming a compound of formula I-d-12:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • R5 is —(CR2)2-4OR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —(CR2)0-6C(O)NR(CR2)0-6CO2R, —(CR2)0-6C(O)NR(CR2)0-6CONR2, —(CR2)0-6NRCOR, —(CR2)0-4NRaC(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or RB; and
    • each of R and RB is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-d-12, wherein R5 is not RB.


In particular embodiments, the present disclosure provides a compound of formula I-d-12, wherein R5 is —(CH2)2-4OH, —(CH2)2-4O(CH2)1-5CO2H, —(CH2)2-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-3CH(CH2OH)2, —(CH2)2-4OC1-4alkyl, —(CH2)1-4CO2H, —(CH2)1-4CO2C1-4alkyl, —(CH2)1-4CONH2, —(CH2)1-4CONHC1-4alkyl, —(CH2)1-4CON(C1-4alkyl)2, —(CH2)1-4C(O)NRa(CH2)1-4CO2H, —(CH2)1-4C(O)NRa(CHMe)CO2H, —(CH2)1-4C(O)NRa(CH2)1-4CONH2, —(CH2)1-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)1-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —(CH2)0-4NRaC(O)(CH2)1-5CO2H, —(CH2)0-4NRaC(O)(CH2)1-5CO2C1-4alkyl, —(CH2)2-4SO3H, —(CH2)2-4SO3C1-4alkyl, —(CH2)2-4SO2NH2, —(CH2)1-4C(O)NRa(CH2)2- 4SO2NH2, —(CH2)1-4CO(N-pyrrolidine)-SO2NH2, —(CH2)2-4SO2NHC1-4alkyl, —(CH2)2-4SO2N(C1-4alkyl)2, —(CH2)2-4OSO2NH2, —(CH2)2-4NRa(CH2)2-4CO2H, —(CH2)2-4NRaCO(CH2)2-4CO2H, —(CH2)2-4NRaCO(cyclopentane)-CO2H, —(CH2)2-4NRaCO(benzene)-OH, —(CH2)2-4NRaCO(benzene)-CO2H, —(CH2)2-4NRaSO3H, —(CH2)2-4NRaSO2C1-4alkyl, —(CH2)2-4NRaSO2(CH2)2-4OH, —(CH2)2-4NRaSO2(CH2)2-4OC1-4alkyl, —(CH2)2-4NRaSO2(CH2)2-4CO2H, —(CH2)2-4NRaSO2(benzene)-CO2H, —(CH2)2-4NRaSO2(benzene)-CO2C1-4alkyl, —(CH2)2-4SO2NRa(CH2)2-4OH, —(CH2)2-4(N-saccharin), —(CH2)2-4SO2NRa(CH2)2-4CO2H, —(CH2)2-4SO2NRa(cyclopentane)-CO2H, —(CH2)2-4NRa(cyclohexane)-CO2C1-4alkyl, —(CH2)2-4SO2(N-pyrrolidine)-CO2H, —(CH2)2-4SO2(N-piperdine)-CO2H, —(CH2)1-4OP(OH)2, —(CH2)1-4P(O)(OH)2, —(CH2)1-4P(O)(OH)(OC1-4alkyl), or —(CH2)2-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H, C1-4alkyl, or —SO2C1-4alkyl.


In some embodiments, the present disclosure provides a compound of formula I-d-12, wherein R in —NRR5 is Ra, which is H or C1-6alkyl, particularly methyl.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein




embedded image


thereby forming a compound of formula




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • Ring C is pyrrolyl or a 4- to 10-membered saturated or partially unsaturated monocyclic, bicyclic, bridged bicyclic, or spirocyclic heterocyclic ring having 0 to 3 heteroatoms (in addition to the nitrogen already depicted in Ring C) independently selected from nitrogen, oxygen, and sulfur;
    • each R6 is independently halogen, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, or 3; and
    • each of R, RB, R3, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-d-13, wherein Ring C is azetidinyl, pyrrolyl, 2,3-dihydro-1H-pyrrolyl, pyrrolidinyl, piperidinyl, morpholinyl, thiazolidinyl, indolinyl, isoindolinyl, octahydroindolyl, azepanyl, an oxazepane, an azabicyclohexane, an azabicycloheptane, an azabicyclooctane, an azabicyclononane, an azaspiroheptane, or an octahydrocyclicpentapyrrole.


In particular embodiments, the present disclosure provides a compound of formula I-d-13, wherein each R is independently fluoro, -methyl, ═O, ═CH2, -cyclohexyl, phenyl, morpholinyl, —OMe, —OtBu, —CF3, —CO2H, —CO2C1-4alkyl, —CO2C1-4haloalkyl, or —OH; and m is 0 or 1, or 2, particularly 0 or 1.


In certain embodiments, the present disclosure provides a compound of formula I-d-13, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and at least one R3 is halogen, such as chloro at the 8-position, thereby forming a compound of formula I-e-1:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of R2 and R3 is as defined above and described in embodiments herein, and n is 0, 1, 2, or 3, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


n is 2, and R3 is chloro at the 7- and 8-positions, thereby forming a compound of formula I-e-2:




embedded image


or a pharmaceutically acceptable salt thereof, wherein R2 is defined above and described in embodiments herein.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is —NR2, thereby forming a compound of formula I-f-1:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of R, R3, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-f-1, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R is chloro at the 7- and 8-positions with respect to the quinoline ring.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is —NR2, thereby forming a compound of formula I-f-2:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • R5 is —(CR2)2-4OR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —(CR2)0-6C(O)NR(CR2)0-6CO2R, —(CR2)0-6C(O)NR(CR2)0-6CONR2, —(CR2)0-4NRC(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or RB; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-f-2, wherein R5 is —(CR2)2-4OR, —(CR2)1-4CO2R, —(CR2)1-4CONR2, —(CR2)0-6C(O)NR(CR2)0-6CO2R, —(CR2)0-6C(O)NR(CR2)0-6CONR2, —(CR2)0-4NRC(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or RB.


In some embodiments, the present disclosure provides a compound of formula I-f-2, wherein R5 is not RB.


In some embodiments, the present disclosure provides a compound of formula I-f-2, wherein R in —NRR5 is Ra, which is H or C1-6alkyl.


In certain embodiments, the present disclosure provides a compound of formula I-f-2, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-f-2, wherein:

    • R in —NRR5 is H or lower alkyl, particularly methyl;
    • R5 is —(CH2)2-4OH, —(CH2)2-4O(CH2)1-5CO2H, —(CH2)2-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-3CH(CH2OH)2, —(CH2)2-4OC1-4alkyl, —(CH2)1-4CO2H, —(CH2)1-4CO2C1-4alkyl, —(CH2)1-4CONH2, —(CH2)1-4CONHC1-4alkyl, —(CH2)1-4CON(C1-4alkyl)2, —(CH2)1-4C(O)NRa(CH2)1-4CO2H, —(CH2)1-4C(O)NRa(CH2)1-4CONH2, —(CH2)1-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)1-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —(CH2)0-4NRaC(O)(CH2)1-5CO2H, —(CH2)0-4NRaC(O)(CH2)1-5CO2C1-4alkyl, —(CH2)2-4SO3H, —(CH2)2-4SO3C1-4alkyl, —(CH2)2-4SO2NH2, —(CH2)2-4SO2NHC1-4alkyl, —(CH2)2-4SO2N(C1-4alkyl)2, —(CH2)2-4OSO2NH2, —(CH2)2-4NRaSO2C1-4alkyl, —(CH2)2-4NRaSO3H, —(CH2)1-4OP(OH)2, —(CH2)1-4P(O)(OH)2, —(CH2)1-4P(O)(OH)(OC1-4alkyl), or —(CH2)2-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-f-2, wherein:

    • R in —NRR5 is H or lower alkyl, particularly methyl;
    • R5 is —(CH2)2-4OH, —(CH2)2-4O(CH2)1-5CO2H, —(CH2)2-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-3CH(CH2OH)2, —(CH2)1-4CO2H, —(CH2)1-4CONH2, —(CH2)1-4C(O)NRa(CH2)1-4CO2H, —(CH2)1-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4NRaC(O)(CH2)1-5CO2H, —(CH2)0-4NRaC(O)(CH2)1-5CO2C1-4alkyl, —(CH2)2-4SO3H, —(CH2)2-4SO2NH2, —(CH2)2-4SO2NRaC1-4alkyl, —(CH2)2-4NRaSO2C1-4alkyl, —(CH2)1-4P(O)(OH)2, or —(CH2)2-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-f-2, R5 includes a terminal —CO2H group. For example, in certain instances, R5 includes a terminal —(CH2)0-4CO2H group.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-f-3:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • Ring B1 is phenyl; a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur; or a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur;
    • each R6 is independently halogen, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-f-3, wherein Ring B1 is phenyl, particularly substituted phenyl. In other embodiments, Ring B1 is not phenyl. For example, Ring B1 may be a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur. In some embodiments, Ring B1 is a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, the present disclosure provides a compound of formula I-f-3, wherein R6 is not RB.


In certain embodiments, the present disclosure provides a compound of formula I-f-3, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-f-3, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-f-4:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • each R6 is independently halogen, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-f-4, wherein R6 is not RB.


In certain embodiments, the present disclosure provides a compound of formula I-f-4, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-f-4, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-f-5:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • each R6 is independently halogen, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • X is —CR2—, —NR—, —O—, —S—, or —SO2—, particularly —CR2—, —NR—, or —O;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4;
    • p is 0, 1, or 2, particularly 1; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-f-5, wherein R6 is not RB.


In certain embodiments, the present disclosure provides a compound of formula I-f-5, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-f-5, wherein:

    • each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, —C3-6cycloalkyl, ═CH2, ═O, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-4CONH2, —(CH2)0-4CONHC1-4alkyl, —(CH2)0-4CON(C1-4alkyl)2, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —NH2, —(CH2)1-4NH2, —(CH2)0-4NHC1-4alkyl, —(CH2)0-4N(C1-4alkyl)2, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4SO2NHC1-4alkyl, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, —(CH2)0-4OP(OH)(OC1-4alkyl), or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • X is —CH2—, —NH—, or —O—;
    • m is 1, 2, 3, or 4, particularly 1 or 2;
    • p is 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-f-5, wherein:

    • each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, —C3-6cycloalkyl, ═CH2, —(CH2)0-4CO2H, —(CH2)1-4O(CH2)1-5CO2H, —(CH2)1-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —OH, —(CH2)1-4OH, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, or —(CR2)0-4OP(O)(H)OH;
    • X is —CH2—, —NH—, or —O—;
    • m is 1 or 2;
    • p is 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-f-5, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-f-6:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • each R6 is independently halogen, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-f-6, wherein R6 is not RB.


In certain embodiments, the present disclosure provides a compound of formula I-f-6, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, the present disclosure provides a compound of formula I-f-6, wherein:

    • each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, —C3-6cycloalkyl, ═CR2, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or —B(OR)2;
    • m is 1, 2, 3, or 4, particularly 1 or 2; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-f-6, wherein:

    • each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, —C3-6cycloalkyl, ═CH2, ═O, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-4CONH2, —(CH2)0-4CONHC1-4alkyl, —(CH2)0-4CON(C1-4alkyl)2, —(CH2)0-4CO(N-proline), —(CH2)0-4CO(N-pyrrolidine-3-carboxylic acid), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa—CH(C1-4alkyl)-CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —NH2, —(CH2)1-4NH2, —(CH2)0-4NHC1-4alkyl, —(CH2)0-4NRaC(O)C1-4alkyl, —(CH2)0-4NRaC(O)Ph, —(CH2)0-4NRaCO(CH2)1-4OH, —(CH2)0-4N(C1-4alkyl)2, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4SO2NRaC1-4alkyl, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4NRaSO2Ph, —(CH2)0-4NRaSO2(CH2)1-4CO2H, —(CH2)0-4OP(OH)2, —(CH2)0-4OP(OH)(OC1-4alkyl), or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • m is 1, 2, 3, or 4, particularly 1 or 2; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-f-6, wherein:

    • each R6 is independently halogen, —C1-4alkyl, —C1-4haloalkyl, —C3-6cycloalkyl, ═CH2, —(CH2)0-4CO2H, —(CH2)1-4O(CH2)1-5CO2H, —(CH2)1-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-4CO(N-proline), —(CH2)0-4CO(N-pyrrolidine-3-carboxylic acid), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa—CH(C1-4alkyl)-CO2H, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —(CH2)0-4NRaC(O)C1-4alkyl, —(CH2)0-4NRaC(O)Ph, —(CH2)0-4NRaCO(CH2)1-4OH, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4NRaSO2Ph, —(CH2)0-4NRaSO2(CH2)1-4CO2H, —(CH2)0-4OP(OH)2, or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • m is 1 or 2; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-f-6, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-f-7:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • Ring B2 is phenyl; a 4- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur; or a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur;
    • each R6 is independently halogen, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4;
    • q is 0, 1, or 2; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-f-7, wherein Ring B2 is phenyl, particularly substituted phenyl. In other embodiments, Ring B2 is not phenyl. For example, Ring B2 may be a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur. In some embodiments, Ring B2 is a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, the present disclosure provides a compound of formula I-f-7, wherein R6 is not RB.


In some embodiments, the present disclosure provides a compound of formula I-f-7, wherein R in




embedded image


is Ra, which is H or C1-6alkyl.


In certain embodiments, the present disclosure provides a compound of formula I-f-7, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-f-7, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-f-8 or formula I-f-8*, respectively:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • each R6 is independently hydrogen (only on N), halogen (not on N), —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • X is —CR2—, —NR—, —O—, —S—, or —SO2—, particularly —CR2—, —NR—, or —O;
    • m is 1, 2, 3, or 4;
    • q is 0, 1, or 2; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-f-8 or formula I-f-8*, wherein R6 is not RB.


In some embodiments, the present disclosure provides a compound of formula I-f-8 or formula I-f-8*, wherein R in




embedded image


is Ra, which is H or C1-6alkyl.


In certain embodiments, the present disclosure provides a compound of formula I-f-8 or formula I-f-8*, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, the present disclosure provides a compound of formula I-f-8 or formula I-f-8*, wherein:

    • each R6 is independently halogen, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or —B(OR)2;
    • m is 1, 2, 3, or 4, particularly 1 or 2;
    • q is 1 or 2; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-f-8 or formula I-f-8*, wherein:

    • each R6 is independently halogen, ═O, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CONH2, —(CH2)0-4CONHC1-4alkyl, —(CH2)0-4CON(C1-4alkyl)2, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —NH2, —(CH2)1-4NH2, —(CH2)0-4NHC1-4alkyl, —(CH2)0-4N(C1-4alkyl)2, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4SO2NHC1-4alkyl, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, —(CH2)0-4OP(OH)(OC1-4alkyl), or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • m is 1, 2, 3, or 4, particularly 1 or 2;
    • q is 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-f-8 or formula I-f-8*, wherein:

    • each R6 is independently halogen, —(CH2)0-4CO2H, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —OH, —(CH2)1-4OH, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • m is 1 or 2;
    • q is 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-f-8 or I-f-8*, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-f-9:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of R, R1, R3, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-f-9, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and at least one R3 is halogen, such as chloro at the 8-position, thereby forming a compound of formula I-g-1:




embedded image


or a pharmaceutically acceptable salt thereof, wherein each of R2 and R3 is as defined above and described in embodiments herein, and n is 0, 1, 2, or 3, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-g-1, wherein R2 is RB, wherein RB is an optionally substituted C1-6 aliphatic, preferably a substituted C1-6 aliphatic, such as —(CR2)1-4NR(CR2)0-6CO2R, —(CR2)1-4O(CR2)0-4CO2R, —(CR2)1-4CO2R, —(CR2)1-4OH, —(CR2)1-4NH2, —(CR2)1-4C(O)NH2, —(CR2)1-4C(O)NHC1-4alkyl, or —(CR2)1-4C(O)N(C1-4alkyl)2, particularly —(CR2)1-4NR(CR2)0-4CO2R or —(CR2)1-4CO2R. In some embodiments, R2 is —(CH2)1-4NRa(CH2)0-4CO2Ra, such as —(CH2)1-4NRa(CH2)0-4CO2H, wherein Ra, independently for each occurrence, is H or C1-4alkyl. In other embodiments, R2 is —(CH2)1-4N(SO2C1-4alkyl)(CH2)0-4CO2Ra, such as is —(CH2)1-4N(SO2C1-4alkyl)(CH2)0-4CO2H, wherein Ra, independently for each occurrence, is H or C1-4alkyl. In other embodiments, R2 is —(CR2)1-4CO2R, such as —(CR2)1-4CO2H, including —(CH2)1-4CO2H. In certain embodiments, R2 includes a terminal —CO2H group. For example, in certain instances, R2 includes a terminal —(CH2)0-4CO2H group.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


n is 2, and R3 is chloro at the 7- and 8-positions, thereby forming a compound




embedded image


or a pharmaceutically acceptable salt thereof, wherein R2 is defined above and described in embodiments herein.


In certain embodiments, the present disclosure provides a compound of formula I-g-2, wherein R2 is RB, wherein R is an optionally substituted C1-6 aliphatic, preferably a substituted C1-6 aliphatic, such as —(CR2)1-4NR(CR2)0-6CO2R, —(CR2)1-4O(CR2)0-6CO2R, —(CR2)1-4CO2R, —(CR2)1-4OH, —(CR2)1-4NH2, —(CR2)1-4C(O)NH2, —(CR2)1-4C(O)NHC1-4alkyl, or —(CR2)1-4C(O)N(C1-4alkyl)2, particularly —(CR2)1-4NR(CR2)0-6CO2R or —(CR2)1-4CO2R. In some embodiments, R2 is —(CH2)1-4NRa(CH2)0-4CO2Ra, such as —(CH2)1-4NRa(CH2)0-4CO2H, wherein Ra, independently for each occurrence, is H or C1-4alkyl. In other embodiments, R2 is —(CH2)1-4N(SO2C1-4alkyl)(CH2)0-4CO2Ra, such as is —(CH2)1-4N(SO2C1-4alkyl)(CH2)0-4CO2H, wherein Ra, independently for each occurrence, is H or C1-4alkyl. In other embodiments, R2 is —(CR2)1-4CO2R, such as —(CR2)1-4CO2H, including —(CH2)1-4CO2H. In certain embodiments, R2 includes a terminal —CO2H group. For example, in certain instances, R2 includes a terminal —(CH2)0-4CO2H group.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is —NR2, thereby forming a compound of formula I-h-1:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • R4 is hydrogen or an optionally substituted C1-6 aliphatic; and
    • each of R, R3, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-h-1, wherein R and R4 in




embedded image


independently are hydrogen or lower alkyl, particularly methyl.


In certain embodiments, the present disclosure provides a compound of formula I-h-1, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is —NRR5, thereby forming a compound of formula I-h-2:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • R4 is hydrogen or an optionally substituted C1-6 aliphatic;
    • R5 is —(CR2)2-4OR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —(CR2)0-6C(O)NR(CR2)0-6CO2R, —(CR2)0-6C(O)NR(CR2)0-6CONR2, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or RB; and
    • each of R, R3, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-h-2, wherein R5 is —(CR2)2-4OR, —(CR2)1-4CO2R, —(CR2)1-4CONR2, —(CR2)0-6C(O)NR(CR2)0-6CO2R, —(CR2)0-6C(O)NR(CR2)0-6CONR2, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or RB.


In some embodiments, the present disclosure provides a compound of formula I-h-2, wherein R5 is not RB.


In some embodiments, the present disclosure provides a compound of formula I-h-2, wherein R in —NRR5 is Ra, which is H or C1-6alkyl.


In some embodiments, the present disclosure provides a compound of formula I-h-2, wherein R and R4 in




embedded image


independently are hydrogen or lower alkyl, particularly methyl.


In certain embodiments, the present disclosure provides a compound of formula I-h-2, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-h-2, wherein:

    • R in —NRR5 is H or lower alkyl, particularly methyl;
    • R5 is —(CH2)2-4OH, —(CH2)0-3CH(CH2OH)2, —(CH2)2-4OC1-4alkyl, —(CH2)1-4CO2H, —(CH2)1-4CO2C1-4alkyl, —(CH2)1-4CONH2, —(CH2)1-4CONHC1-4alkyl, —(CH2)1-4CON(C1-4alkyl)2, —(CH2)1-4C(O)NRa(CH2)1-4CO2H, —(CH2)1-4C(O)NRa(CH2)1-4CONH2, —(CH2)1-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)1-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —(CH2)2-4SO3H, —(CH2)2-4SO3C1-4alkyl, —(CH2)2-4SO2NH2, —(CH2)2-4SO2NHC1-4alkyl, —(CH2)2-4SO2N(C1-4alkyl)2, —(CH2)2-4OSO2NH2, —(CH2)2-4NRaSO2C1-4alkyl, —(CH2)2-4NRaSO3H, —(CH2)1-4OP(OH)2, —(CH2)1-4P(O)(OH)2, —(CH2)1-4P(O)(OH)(OC1-4alkyl), or —(CH2)2-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-h-2, wherein:

    • R in —NRR5 is H or lower alkyl, particularly methyl;
    • R5 is —(CH2)2-4OH, —(CH2)0-3CH(CH2OH)2, —(CH2)1-4CO2H, —(CH2)1-4CONH2, —(CH2)1-4C(O)NRa(CH2)1-4CO2H, —(CH2)1-4C(O)NRa(CH2)1-4CONH2, —(CH2)2-4SO3H, —(CH2)2-4SO2NH2, —(CH2)2-4SO2NRaC1-4alkyl, —(CH2)2-4NRaSO2C1-4alkyl, —(CH2)1-4P(O)(OH)2, or —(CH2)2-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-h-2, R5 includes a terminal —CO2H group. For example, in certain instances, R5 includes a terminal —(CH2)0-4CO2H group.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-h-3:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • Ring B1 is phenyl; a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur; or a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur;
    • R4 is hydrogen or an optionally substituted C1-6 aliphatic;
    • each R6 is independently halogen, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-h-3, wherein Ring B1 is phenyl, particularly substituted phenyl. In other embodiments, Ring B1 is not phenyl. For example, Ring B1 may be a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur. In some embodiments, Ring B1 is a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, the present disclosure provides a compound of formula I-h-3, wherein R6 is not RB.


In some embodiments, the present disclosure provides a compound of formula I-h-3, wherein R and R4 in




embedded image


independently are hydrogen or lower alkyl, particularly methyl.


In certain embodiments, the present disclosure provides a compound of formula I-h-3, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-h-3, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-h-4:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • R4 is hydrogen or an optionally substituted C1-6 aliphatic;
    • each R6 is independently halogen, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-h-4, wherein R6 is not RB.


In some embodiments, the present disclosure provides a compound of formula I-h-4, wherein R and R4 in




embedded image


independently are hydrogen or lower alkyl, particularly methyl.


In certain embodiments, the present disclosure provides a compound of formula I-h-4, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-h-5:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • R4 is hydrogen or an optionally substituted C1-6 aliphatic;
    • each R6 is independently halogen, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • X is —CR2—, —NR—, —O—, —S—, or —SO2—, particularly —CR2—, —NR—, or —O;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4;
    • p is 0, 1, or 2, particularly 1; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-h-5, wherein R6 is not RB.


In some embodiments, the present disclosure provides a compound of formula I-h-5, wherein R and R4 in




embedded image


independently are hydrogen or lower alkyl, particularly methyl.


In certain embodiments, the present disclosure provides a compound of formula I-h-5, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-h-5, wherein:

    • each R6 is independently halogen, ═O, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CONH2, —(CH2)0-4CONHC1-4alkyl, —(CH2)0-4CON(C1-4alkyl)2, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —NH2, —(CH2)1-4NH2, —(CH2)0-4NHC1-4alkyl, —(CH2)0-4N(C1-4alkyl)2, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4SO2NHC1-4alkyl, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, —(CH2)0-4OP(OH)(OC1-4alkyl), or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • X is —CH2—, —NH—, or —O—;
    • m is 1, 2, 3, or 4, particularly 1 or 2;
    • p is 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-h-5, wherein:

    • each R6 is independently halogen, —(CH2)0-4CO2H, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —OH, —(CH2)1-4OH, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, or —(CR2)0-4OP(O)(H)OH;
    • X is —CH2—, —NH—, or —O—;
    • m is 1 or 2;
    • p is 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-h-5, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-h-6:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • R4 is hydrogen or an optionally substituted C1-6 aliphatic;
    • each R6 is independently halogen, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-h-6, wherein R6 is not RB.


In some embodiments, the present disclosure provides a compound of formula I-h-6, wherein R and R4 in




embedded image


independently are hydrogen or lower alkyl, particularly methyl.


In certain embodiments, the present disclosure provides a compound of formula I-h-6, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, the present disclosure provides a compound of formula I-h-6, wherein:

    • each R6 is independently halogen, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or —B(OR)2;
    • m is 1, 2, 3, or 4, particularly 1 or 2; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-h-6, wherein:

    • each R6 is independently halogen, ═O, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CONH2, —(CH2)0-4CONHC1-4alkyl, —(CH2)0-4CON(C1-4alkyl)2, —(CH2)0-4CO(N-proline), —(CH2)0-4CO(N-pyrrolidine-3-carboxylic acid), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa—CH(C1-4alkyl)-CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —NH2, —(CH2)1-4NH2, —(CH2)0-4NHC1-4alkyl, —(CH2)0-4NRaC(O)C1-4alkyl, —(CH2)0-4NRaC(O)Ph, —(CH2)0-4NRaCO(CH2)1-4OH, —(CH2)0-4N(C1-4alkyl)2, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4SO2NRaC1-4alkyl, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4NRaSO2Ph, —(CH2)0-4NRaSO2(CH2)1-4CO2H, —(CH2)0-4OP(OH)2, —(CH2)0-4OP(OH)(OC1-4alkyl), or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • m is 1, 2, 3, or 4, particularly 1 or 2; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-h-6, wherein:

    • each R6 is independently halogen, —(CH2)0-4CO2H, —(CH2)0-4CO(N-proline), —(CH2)0-4CO(N-pyrrolidine-3-carboxylic acid), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa—CH(C1-4alkyl)-CO2H, —OH, —(CH2)1-4OH, —(CH2)0-4NRaC(O)C1-4alkyl, —(CH2)0-4NRaC(O)Ph, —(CH2)0-4NRaCO(CH2)1-4OH, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4NRaSO2Ph, —(CH2)0-4NRaSO2(CH2)1-4CO2H, —(CH2)0-4OP(OH)2, or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • m is 1 or 2; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-h-6, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • Ring B2 is phenyl; a 4- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur; or a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur;
    • R4 is hydrogen or an optionally substituted C1-6 aliphatic;
    • each R6 is independently halogen, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • m is 0, 1, 2, 3, or 4, particularly 1, 2, 3, or 4;
    • q is 0, 1, or 2; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In certain embodiments, the present disclosure provides a compound of formula I-h-7, wherein Ring B2 is phenyl, particularly substituted phenyl. In other embodiments, Ring B2 is not phenyl. For example, Ring B2 may be a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur. In some embodiments, Ring B2 is a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, the present disclosure provides a compound of formula I-h-7, wherein R6 is not RB.


In some embodiments, the present disclosure provides a compound of formula I-h-7, wherein R in




embedded image


is Ra, which is H or C1-6alkyl.


In some embodiments, the present disclosure provides a compound of formula I-h-7, wherein R and R4 in




embedded image


independently are hydrogen or lower alkyl, particularly methyl.


In certain embodiments, the present disclosure provides a compound of formula I-h-7, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-h-7, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-h-8 or formula I-h-8*, respectively:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • R4 is hydrogen or an optionally substituted C1-6 aliphatic;
    • each R6 is independently hydrogen (only on N), halogen (not on N), —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB;
    • X is —CR2—, —NR—, —O—, —S—, or —SO2—, particularly —CR2—, —NR—, or —O;
    • m is 1, 2, 3, or 4;
    • q is 0, 1, or 2; and
    • each of R, R3, RB, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-h-8 or formula I-h-8*, wherein R6 is not RB.


In some embodiments, the present disclosure provides a compound of formula I-h-8 or formula I-h-8*, wherein R4 in




embedded image


is Ra, which is H or C1-6alkyl.


In some embodiments, the present disclosure provides a compound of formula I-h-8 or formula I-h-8*, wherein R and R4 in




embedded image


independently are hydrogen or lower alkyl, particularly methyl.


In certain embodiments, the present disclosure provides a compound of formula I-h-8 or formula I-h-8*, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, the present disclosure provides a compound of formula I-h-8 or formula I-h-8*, wherein:

    • each R6 is independently halogen, ═O, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or —B(OR)2;
    • m is 1, 2, 3, or 4, particularly 1 or 2;
    • q is 1 or 2; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In particular embodiments, the present disclosure provides a compound of formula I-h-8 or formula I-h-8*, wherein:

    • each R6 is independently halogen, ═O, —(CH2)0-4CO2H, —(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CONH2, —(CH2)0-4CONHC1-4alkyl, —(CH2)0-4CON(C1-4alkyl)2, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —OH, —(CH2)1-4OH, —(CH2)0-4OC1-4alkyl, —NH2, —(CH2)1-4NH2, —(CH2)0-4NHC1-4alkyl, —(CH2)0-4N(C1-4alkyl)2, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4SO2NHC1-4alkyl, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, —(CH2)0-4OP(OH)(OC1-4alkyl), or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • m is 1, 2, 3, or 4, particularly 1 or 2;
    • q is 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In further particular embodiments, the present disclosure provides a compound of formula I-h-8 or formula I-h-8*, wherein:

    • each R6 is independently halogen, —(CH2)0-4CO2H, —(CH2)0-4CO(N-proline), —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —OH, —(CH2)1-4OH, —(CH2)0-4SO3H, —(CH2)0-4SO2NH2, —(CH2)0-4NRaSO2C1-4alkyl, —(CH2)0-4OP(OH)2, or —(CR2)0-4OP(O)(H)OH, wherein Ra, independently for each occurrence, is H or C1-4alkyl;
    • m is 1 or 2;
    • q is 1; and
    • each R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In certain embodiments, for the above disclosed compounds of formula I-h-8 or I-h-8*, m is 1 or 2, and at least one R6 includes a terminal —CO2H group. For example, in certain instances, at least one R6 is —(CH2)0-4CO2H.


In some embodiments, the present disclosure provides a compound of formula I or formula I*, wherein R1 is




embedded image


and R2 is



embedded image


thereby forming a compound of formula I-h-9:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • R4 is hydrogen or an optionally substituted C1-6 aliphatic; and
    • each of R, R, and n is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-h-9, wherein R and R4 in




embedded image


independently are hydrogen or lower alkyl, particularly methyl.


In certain embodiments, the present disclosure provides a compound of formula I-h-9, wherein R3 is independently halogen, such as chloro, including dichloro, including wherein n is 2 and R3 is chloro at the 7- and 8-positions with respect to the quinoline ring.


In some embodiments, the present disclosure provides a compound of formula I, wherein R1 is




embedded image


and at least one R3 is halogen, such as chloro at the 8-position, thereby forming a compound of formula I-i-1:




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • R4 is hydrogen or an optionally substituted C1-6 aliphatic; and
    • each of R, R2, and R3 is as defined above and described in embodiments herein, and n is 0, 1, 2, or 3, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-i-1, wherein R and R4 in




embedded image


independently are hydrogen or lower alkyl, particularly methyl.


In some embodiments, the present disclosure provides a compound of formula I, wherein R1 is




embedded image


n is 2, and R3 is chloro at the 7- and 8-positions, thereby forming a compound of formula




embedded image


or a pharmaceutically acceptable salt thereof, wherein:

    • R4 is hydrogen or an optionally substituted C1-6 aliphatic; and
    • each of R and R2 is as defined above and described in embodiments herein, both singly and in combination.


In some embodiments, the present disclosure provides a compound of formula I-i-2, wherein R and R4 in




embedded image


independently are hydrogen or lower alkyl, particularly methyl.


As defined above and described herein, R1 is halogen, —OR, —NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —NRSO2R, —SR, —SO2R, —SO2NR2, —S(O)R, or RA, particularly —NR2 or RA.


In some embodiments, R1 is halogen. In some embodiments, R1 is —OR. In some embodiments, R1 is —NR2. In some embodiments, R1 is —NRC(O)OR. In some embodiments, R1 is —NRC(O)R. In some embodiments, R1 is —NRC(O)NR2. In some embodiments, R1 is —NRSO2R. In some embodiments, R1 is —SR. In some embodiments, R1 is —SO2R. In some embodiments, R1 is —SO2NR2. In some embodiments, R1 is —S(O)R. In some embodiments, R1 is RA.


In some embodiments R1 is hydrogen. In some embodiments, R1 is chloro. In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is




embedded image


In some embodiments, R1 is selected from those groups depicted in Table 1.


As defined above and described herein, each R is independently hydrogen or an optionally substituted group selected from C1-6 aliphatic; benzyl; phenyl; a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carbocyclic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur; and a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur, or two R groups on the same nitrogen are optionally taken together with their intervening atoms to form an optionally substituted 4- to 7-membered saturated, partially unsaturated, or heteroaryl ring having 0 to 3 heteroatoms, in addition to the nitrogen, independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, R is hydrogen. In some embodiments, R is an optionally substituted C1-6 aliphatic. In some embodiments, R is an optionally substituted benzyl. In some embodiments, R is an optionally substituted phenyl. In some embodiments, R is an optionally substituted 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carbocyclic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur. In some embodiments, R is an optionally substituted 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, 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- to 7-membered saturated, partially unsaturated, or heteroaryl ring having 0 to 3 heteroatoms, in addition to the nitrogen, independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, two R groups on the same carbon atom (e.g., —(CR2)—, etc.) are optionally taken together with their intervening atoms to form an optionally substituted 4- to 7-membered saturated or partially unsaturated ring having 0 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, two R groups on different atoms (e.g., —P(O)(OR)2, —B(OR)2, etc.) are optionally taken together with their intervening atoms to form an optionally substituted 4- to 7-membered saturated or partially unsaturated ring having 0 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, R is hydrogen. In some embodiments, R is methyl. In some embodiments, R is ethyl. In some embodiments, R is isopropyl. In some embodiments, R is phenyl. In some embodiments, R is benzyl. In some embodiments, R is —(CH2)1-5CO2H. In some embodiments, R is —(CH2)1-5CO2C1-4alkyl.


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


As defined above and described herein, R2 is halogen, —OR, —NR2, —NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —NRSO2R, —SR, —SO2R, —SO2NR2, —S(O)R, or RB, particularly —NR2 or RB;


In some embodiments, R2 is halogen. In some embodiments, R2 is —OR. In some embodiments, R2 is —NR2. In some embodiments, R2 is —NRC(O)OR. In some embodiments, R2 is —NRC(O)R. In some embodiments, R2 is —NRC(O)NR2. In some embodiments, R2 is —NRSO2R, —SR. In some embodiments, R2 is —SO2R. In some embodiments, R2 is —SO2NR2. In some embodiments, R2 is —S(O)R. In some embodiments, R2 is RB.


In some embodiments, R2 is hydrogen. In some embodiments, R2 is chloro. In some embodiments, R2 is methyl. In some embodiments, R2 is —CF3. In some embodiments, R2 is phenyl. In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In one embodiments R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some


embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some is embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is selected from those groups depicted in Table 1.


As defined above and described herein, each R3 is independently halogen, —OR, —NR2, —SR, or RC, particularly halogen, such as chloro.


In some embodiments, R3 is halogen. In some embodiments, R3 is —OR. In some embodiments, R3 is —NR2. In some embodiments, R3 is —SR. In some embodiments, R3 is RC.


In some embodiments, R3 is fluoro. In some embodiments, R3 is chloro. In some embodiments, R3 is bromo. In some embodiments, R3 is —OH. In some embodiments, R3 is —OMe. In some embodiments, R3 is —OEt. In some embodiments, R3 is methyl. In some embodiments, R3 is isopropyl. In some embodiments, R3 is tert-butyl. In some embodiments, R3 is —CF3. In some embodiments, R3 is phenyl. In some embodiments, R3 is




embedded image


In some embodiments, R3 is selected from those groups depicted in Table 1.


As defined above and described herein, R4 is hydrogen or an optionally substituted C1-6 aliphatic.


In some embodiments, R4 is hydrogen. In some embodiments, R4 is an optionally substituted C1-6 aliphatic. In some embodiments, R4 is methyl.


In some embodiments, R4 is selected from those groups depicted in Table 1.


As defined above and described herein, R5 is —(CR2)0-6OR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —(CR2)0-6C(O)NR(CR2)0-6CO2R, —(CR2)0-6C(O)NR(CR2)0-6CONR2, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, or RB


In some embodiments, R5 is —(CR2)0-6OR. In some embodiments, R5 is —(CR2)0-6CO2R. In some embodiments, R5 is —(CR2)0-6CONR2. In some embodiments, R5 is —(CR2)0-6C(O)NR(CR2)0-6CO2R. In some embodiments, R5 is —(CR2)0-6C(O)NR(CR2)0-6CONR2. In some embodiments, R5 is —(CR2)0-6SO3R. In some embodiments, R5 is —(CR2)0-6SO2NR2. In some embodiments, R5 is —(CR2)0-6OSO2NR2. In some embodiments, R5 is —(CR2)0-6NRSO2R. In some embodiments, R5 is —(CR2)0-6NRSO2OR. In some embodiments, R5 is —(CR2)0-6OP(OR)2. In some embodiments, R5 is —(CR2)0-6OP(O)(OR)2. In some embodiments, R5 is —(CR2)0-6P(O)(OR)2. In some embodiments, R5 is —(CR2)0-6OP(O)(H)OR. In some embodiments, R5 is RB.


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R2 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is




embedded image


In some embodiments, R5 is selected from those groups depicted in Table 1.


As defined above and described herein, each R6 is independently halogen, —COR, —(CR2)0-6CO2R, —(CR2)0-6CONR2, —OR, —(CR2)1-4OR, —NR2, —(CR2)1-4NR2, NRC(O)OR, —NRC(O)R, —NRC(O)NR2, —SR, —SO2R, —S(O)R, —(CR2)0-6SO3R, —(CR2)0-6SO2NR2, —(CR2)0-6OSO2NR2, —(CR2)0-6NRSO2R, —(CR2)0-6NRSO2OR, —(CR2)0-6OP(OR)2, —(CR2)0-6OP(O)(OR)2, —(CR2)0-6P(O)(OR)2, —(CR2)0-6OP(O)(H)OR, —B(OR)2, or RB.


In some embodiments, R6 is halogen. In some embodiments, R6 is —COR. In some embodiments, R6 is —(CR2)0-6CO2R. In some embodiments, R6 is —(CR2)0-6CONR2. In some embodiments, R6 is —OR. In some embodiments, R6 is —(CR2)1-4OR. In some embodiments, R6 is —NR2. In some embodiments, R6 is —(CR2)1-4NR2. In some embodiments, R6 is —NRC(O)OR. In some embodiments, R6 is —NRC(O)R. In some embodiments, R6 is —NRC(O)NR2. In some embodiments, R6 is —SR. In some embodiments, R6 is —SO2R. In some embodiments, R6 is —S(O)R. In some embodiments, R6 is —(CR2)0-6SO3R. In some embodiments, R6 is —(CR2)0-6SO2NR2. In some embodiments, R6 is —(CR2)0-6OSO2NR2. In some embodiments, R6 is —(CR2)0-6NRSO2R. In some embodiments, R6 is —(CR2)0-6NRSO2OR. In some embodiments, R6 is —(CR2)0-6OP(OR)2. In some embodiments, R6 is —(CR2)0-6OP(O)(OR)2. In some embodiments, R6 is —(CR2)0-6P(O)(OR)2. In some embodiments, R6 is —(CR2)0-6OP(O)(H)OR. In some embodiments, R6 is —B(OR)2. In some embodiments, R6 is RB.


In some embodiments, R6 is fluoro. In some embodiments, R6 is chloro. In some embodiments, R6 is bromo. In some embodiments, R6 is —OH. In some embodiments, R6 is —OMe. In some embodiments, R6 is —OEt. In some embodiments, R6 is —OtBu. In some embodiments, R6 is —OBn. In some embodiments, R6 is —OCH2CH2OH. In some embodiments, R6 is —OCH2CO2H. In some embodiments, R6 is —OCONHCH2CO2H. In some embodiments, R6 is —CF3. In some embodiments, R6 is —OCF3. In some embodiments, R6 is —CH(OH)CF3. In some embodiments, R6 is —C(OH)(CF3)2. In some embodiments, R6 is cyano. In some embodiments, R6 is methyl. In some embodiments, R6 is ═CH2. In some embodiments, R6 is ethyl. In some embodiments, R6 is —CH2OH. In some embodiments, R6 is —CH2OMe. In some embodiments, R6 is acetyl. In some embodiments, R6 is —OAc. In some embodiments, R6 is —NH2. In some embodiments, R6 is —NHBoc. In some embodiments, R6 is —CH2NH2. In some embodiments, R6 is —CH2NHBoc. In some embodiments, R6 is —CH2CH2OH. In some embodiments, R6 is tert-butyl. In some embodiments, R6 is —C3-6cycloalkyl. In some embodiments, R6 is cyclohexyl. In some embodiments, R6 is phenyl. In some embodiments, R6 is N-morpholinyl. In some embodiments, R6 is —CO2H. In some embodiments, R6 is —CO2Me. In some embodiments, R6 is —CO2Et. In some embodiments, R6 is —CO2iPr. In some embodiments, R6 is —CO2nBu. In some embodiments, R6 is —CO2isoBu. In some embodiments, R6 is —CO2tBu. In some embodiments, R6 is —CO2CH2CF3. In some embodiments, R6 is —CO2CH(CF3)2. In some embodiments, R6 is —CONH2. In some embodiments, R6 is —CONHOH. In some embodiments, R6 is —CONHOMe. In some embodiments, R6 is —CONHMe. In some embodiments, R6 is —CONHEt. In some embodiments, R6 is —CONMe2. In some embodiments, R6 is —COCH2OH. In some embodiments, R6 is —COCH2OAc. In some embodiments, R6 is —CONHCH2CH2OH. In some embodiments, R6 is —CONHCH2CONH2. In some embodiments, R6 is —CONHCH2CH2CONH2. In some embodiments, R6 is —CONHCH2CO2Me. In some embodiments, R6 is —CONHCH2CH2CO2H. In some embodiments, R6 is —NH2. In some embodiments, R6 is —NHAc. In some embodiments, R6 is —CH2CO2H. In some embodiments, R6 is —CH2CO2Me. In some embodiments, R6 is —CH2CO2tBu. In some embodiments, R6 is —CH2CONH2. In some embodiments, R6 is —CH2NHSO2Me. In some embodiments, R6 is —NHSO2Me. In some embodiments, R6 is —NHSO2Ph. In some embodiments, R6 is —NHCONH2. In some embodiments, R6 is —NHCOPh. In some embodiments, R6 is —NHCOCH2OH. In some embodiments, R6 is —NHCOCH2OAc. In some embodiments, R6 is —NHSO2CH2CH2CO2H. In some embodiments, R6 is —CH2NHCOCH2OH. In some embodiments, R6 is —CH2NHCOCH2CO2H. In some embodiments, R6 is —SO3H. In some embodiments, R6 is —SO2Me. In some embodiments, R6 is —SO2NH2. In some embodiments, R6 is —SO2NHBoc. In some embodiments, R6 is —CH2SO2NH2. In some embodiments, R6 is —OSO2NH2. In some embodiments, R6 is —OCONHCH2CO2Me. In some embodiments, R6 is —B(OH)2. In some embodiments R6 is —CH2OSO2NH2. In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is —OP(OH)2. In some embodiments, R6 is —CH2OP(OH)2. In some embodiments, R6 is —CH2OP(O)(H)OH. In some embodiments, R6 is —OP(O)(OH)2. In some embodiments, R6 is —CH2OP(O)(OH)2. In some embodiments, R6 is —CH2P(O)(OH)2. In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


In some embodiments, R6 is




embedded image


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


As defined above and described herein, RA, RB, and RC, independently, are an optionally substituted group selected from C1-6 aliphatic; phenyl, including substituted phenyl; a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur; and a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, RA, RB, or RC is an optionally substituted C1-6 aliphatic. In some embodiments, RA, RB, or RC is an optionally substituted phenyl. In some embodiments, RA, RB, or RC is an optionally substituted 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur. In some embodiments, RA, RB, or RC is an optionally substituted 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, RA, RB, or RC, is selected from those groups depicted in Table 1.


As defined above and described herein, Ring A is an optionally substituted 4- to 7-membered saturated, partially unsaturated, or heteroaryl ring having 0 to 3 heteroatoms, in addition to the nitrogen, independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, Ring A is an optionally substituted 4- to 7-membered saturated, partially unsaturated, or heteroaryl ring having 0 to 3 heteroatoms, in addition to the nitrogen, independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


In some embodiments, Ring A is




embedded image


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


As defined above and described herein, Ring B1 is phenyl, particularly substituted phenyl; a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur; or a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, Ring B1 is phenyl, particularly substituted phenyl. In some embodiments, Ring B1 is a 4- to 10-membered saturated or partially unsaturated monocyclic or bicyclic carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur. In some embodiments, Ring B1 is a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is




embedded image


In some embodiments, Ring B1 is selected from those groups depicted in Table 1.


As defined above and described herein, Ring B2 is phenyl, including substituted phenyl; a 4- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur; or a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, Ring B2 is phenyl. In some embodiments, Ring B2 is a 4- to 7-membered saturated or partially unsaturated carboxylic or heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur. In some embodiments, Ring B2 is a 5- to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, phosphorus, and sulfur.


In some embodiments, Ring B2 is




embedded image


In some embodiments, Ring B2 is




embedded image


In some embodiments, Ring B2 is




embedded image


In some embodiments, Ring B2 is selected from those groups depicted in Table 1.


As defined above and described herein, Ring C is pyrrolidinyl or a 4- to 10-membered saturated or partially unsaturated monocyclic, bicyclic, bridged bicyclic, or spirocyclic heterocyclic ring having 0 to 3 heteroatoms (in addition to the nitrogen already depicted in Ring C) independently selected from nitrogen, oxygen, and sulfur.


In some embodiments, Ring C is pyrrolidinyl. In some embodiments, Ring C is a 4- to 10-membered saturated or partially unsaturated monocyclic, bicyclic, bridged bicyclic, or spirocyclic heterocyclic ring having 0 to 3 heteroatoms (in addition to the nitrogen already depicted in Ring C) independently selected from nitrogen, oxygen, and sulfur.


In some embodiments, Ring C is azetidinyl. In some embodiments, Ring C is pyrrolyl. In some embodiments, Ring C is 2,3-dihydro-1H-pyrrolyl. In some embodiments, Ring C is morpholinyl. In some embodiments, Ring C is thiazolidinyl. In some embodiments, Ring C is indolinyl. In some embodiments, Ring C is isoindolinyl. In some embodiments, Ring C is octahydroindolyl. In some embodiments, Ring C is azepanyl. In some embodiments, Ring C is oxazepanyl. In some embodiments, Ring C is an azabicyclohexane. In some embodiments, Ring C is an azabicycloheptane. In some embodiments, Ring C is an azabicyclooctane. In some embodiments, Ring C is an azabicyclononane. In some embodiments, Ring C is an azaspiroheptane. In some embodiments, Ring C is octahydrocyclicpentapyrrole.


In particular embodiments, Ring C is pyrrolidinyl, 2-azabicyclo[3.1.0]hexanyl, 3-azabicyclo[3.1.0]hexane, 5-azaspiro[2.4]heptanyl, or octahydrocyclopenta[b]pyrrolyl.


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is




embedded image


In some embodiments, Ring C is selected from those groups depicted in Table 1.


As defined above and described herein, n is 1, 2, 3, or 4.


In some embodiments, n is 1. In some embodiments, n is 2. In some embodiments, n is 3. In some embodiments, n is 4.


In some embodiments, n is selected from those groups depicted in Table 1.


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


In some embodiments, m is 0. In some embodiments, m is 1, 2, 3, or 4. In some embodiments, m is 1. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, m is 4.


In some embodiments, m is selected from those groups depicted in Table 1.


As defined above and described herein, p is 0, 1, or 2.


In some embodiments, p is 0. In some embodiments, p is 1. In some embodiments, p is 2.


In some embodiments, p is selected from those groups depicted in Table 1.


As defined above and described herein, q is 0, 1, or 2.


In some embodiments, q is 0. In some embodiments, q is 1. In some embodiments, q is 2.


In some embodiments, q is selected from those groups depicted in Table 1.


Exemplary compounds of the present disclosure 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







I-45


embedded image







I-46


embedded image







I-47


embedded image







I-48


embedded image







I-49


embedded image







I-50


embedded image







I-51


embedded image







I-52


embedded image







I-53


embedded image







I-54


embedded image







I-55


embedded image







I-56


embedded image







I-57


embedded image







I-58


embedded image







I-59


embedded image







I-60


embedded image







I-61


embedded image







I-62


embedded image







I-63


embedded image







I-64


embedded image







I-65


embedded image







I-66


embedded image







I-67


embedded image







I-68


embedded image







I-69


embedded image







I-70


embedded image







I-71


embedded image







I-72


embedded image







I-73


embedded image







I-74


embedded image







I-75


embedded image







I-76


embedded image







I-77


embedded image







I-78


embedded image







I-79


embedded image







I-80


embedded image







I-81


embedded image







I-82


embedded image







I-83


embedded image







I-84


embedded image







I-85


embedded image







I-86


embedded image







I-87


embedded image







I-88


embedded image







I-89


embedded image







I-90


embedded image







I-91


embedded image







I-92


embedded image







I-93


embedded image







1-94


embedded image







I-95


embedded image







I-96


embedded image







I-97


embedded image







I-98


embedded image







I-99


embedded image







I-100


embedded image







I-101


embedded image







I-102


embedded image







I-103


embedded image







I-104


embedded image







I-105


embedded image







I-106


embedded image







I-107


embedded image







I-108


embedded image







I-109


embedded image







I-110


embedded image







I-111


embedded image







I-112


embedded image







I-113


embedded image







I-114


embedded image







I-115


embedded image







I-116


embedded image







I-117


embedded image







I-118


embedded image







I-119


embedded image







I-120


embedded image







I-121


embedded image







I-122


embedded image







I-123


embedded image







I-124


embedded image







I-125


embedded image







I-126


embedded image







I-127


embedded image







I-128


embedded image







I-130


embedded image







I-131


embedded image







I-132


embedded image







I-133


embedded image







I-134


embedded image







I-135


embedded image







I-136


embedded image







I-137


embedded image







I-138


embedded image







I-139


embedded image







I-140


embedded image







I-141


embedded image







I-142


embedded image







I-143


embedded image







I-144


embedded image







I-145


embedded image







I-147


embedded image







I-148


embedded image







I-149


embedded image







I-150


embedded image







I-151


embedded image







I-152


embedded image







I-153


embedded image







I-154


embedded image







I-155


embedded image







I-156


embedded image







I-157


embedded image







I-158


embedded image







I-159


embedded image







I-160


embedded image







I-161


embedded image







I-162


embedded image







I-163


embedded image







I-164


embedded image







I-165


embedded image







I-166


embedded image







I-167


embedded image







I-168


embedded image







I-169


embedded image







I-170


embedded image







I-171


embedded image







I-172


embedded image







I-173


embedded image







I-174


embedded image







I-175


embedded image







I-176


embedded image







I-177


embedded image







I-178


embedded image







I-179


embedded image







I-180


embedded image







I-181


embedded image







I-182


embedded image







I-183


embedded image







I-184


embedded image







I-185


embedded image







I-186


embedded image







I-187


embedded image







I-188


embedded image







I-189


embedded image







I-190


embedded image







I-191


embedded image







I-192


embedded image







I-193


embedded image







I-194


embedded image







I-195


embedded image







I-196


embedded image







I-197


embedded image







I-198


embedded image







I-199


embedded image







I-200


embedded image







I-201


embedded image







I-202


embedded image







I-203


embedded image







I-204


embedded image







I-205


embedded image







I-206


embedded image







I-207


embedded image







I-208


embedded image







I-209


embedded image







I-210


embedded image







I-211


embedded image







I-212


embedded image







I-213


embedded image







I-214


embedded image







I-215


embedded image







I-216


embedded image







I-217


embedded image







I-218


embedded image







I-219


embedded image







I-220


embedded image







I-221


embedded image







I-222


embedded image







I-223


embedded image







I-224


embedded image







I-225


embedded image







I-226


embedded image







I-227


embedded image







I-228


embedded image







I-229


embedded image







I-230


embedded image







I-231


embedded image







I-232


embedded image







I-234


embedded image







I-235


embedded image







I-236


embedded image







I-237


embedded image







I-238


embedded image







I-239


embedded image







I-240


embedded image







I-241


embedded image







I-242


embedded image







I-243


embedded image







I-244


embedded image







I-245


embedded image







I-246


embedded image







I-247


embedded image







I-248


embedded image







I-249


embedded image







I-250


embedded image







I-251


embedded image







I-252


embedded image







I-253


embedded image







I-254


embedded image







I-255


embedded image







I-256


embedded image







I-257


embedded image







I-258


embedded image







I-259


embedded image







I-260


embedded image







I-261


embedded image







I-262


embedded image







I-263


embedded image







I-264


embedded image







I-265


embedded image







I-266


embedded image







I-267


embedded image







I-268


embedded image







I-269


embedded image







I-270


embedded image







I-271


embedded image







I-272


embedded image







I-273


embedded image







I-274


embedded image







I-275


embedded image







I-276


embedded image







I-277


embedded image







I-278


embedded image







I-279


embedded image







I-280


embedded image







I-281


embedded image







I-282


embedded image







I-283


embedded image







I-284


embedded image







I-285


embedded image







I-286


embedded image







I-287


embedded image







I-288


embedded image







I-289


embedded image







I-290


embedded image







I-291


embedded image







I-292


embedded image







I-293


embedded image







I-294


embedded image







I-295


embedded image







I-296


embedded image







I-297


embedded image







I-298


embedded image







I-299


embedded image







I-300


embedded image







I-301


embedded image







I-302


embedded image







I-303


embedded image







I-304


embedded image







I-305


embedded image







I-306


embedded image







I-307


embedded image







I-308


embedded image







I-309


embedded image







I-310


embedded image







I-311


embedded image







I-312


embedded image







I-313


embedded image







I-314


embedded image







I-315


embedded image







I-316


embedded image







I-317


embedded image







I-318


embedded image







I-319


embedded image







I-320


embedded image







I-321


embedded image







I-322


embedded image







I-323


embedded image







I-324


embedded image







I-325


embedded image







I-326


embedded image







I-327


embedded image







I-328


embedded image







I-329


embedded image







I-330


embedded image







I-331


embedded image







I-332


embedded image







I-333


embedded image







I-334


embedded image







I-335


embedded image







I-336


embedded image







I-337


embedded image







I-338


embedded image







I-339


embedded image







I-340


embedded image







I-341


embedded image







I-342


embedded image







I-343


embedded image







I-344


embedded image







I-345


embedded image







I-346


embedded image







I-347


embedded image







I-348


embedded image







I-349


embedded image







I-350


embedded image







I-351


embedded image







I-352


embedded image







I-353


embedded image







I-354


embedded image







I-355


embedded image







I-356


embedded image







I-357


embedded image







I-358


embedded image







I-359


embedded image







I-360


embedded image







I-361


embedded image







I-362


embedded image







I-363


embedded image







I-364


embedded image







I-365


embedded image







I-366


embedded image







I-367


embedded image







I-368


embedded image







I-369


embedded image







I-370


embedded image







I-371


embedded image







I-372


embedded image







I-373


embedded image







I-374


embedded image







I-375


embedded image







I-376


embedded image







I-377


embedded image







I-378


embedded image







I-379


embedded image







I-380


embedded image







I-381


embedded image







I-382


embedded image







I-383


embedded image







I-384


embedded image







I-385


embedded image







I-386


embedded image







I-387


embedded image







I-388


embedded image







I-389


embedded image







I-390


embedded image







I-391


embedded image







I-392


embedded image







I-393


embedded image







I-394


embedded image







I-395


embedded image







I-396


embedded image







I-397


embedded image







I-398


embedded image







I-399


embedded image







I-400


embedded image







I-401


embedded image







I-402


embedded image







I-403


embedded image







I-404


embedded image







I-405


embedded image







I-406


embedded image







I-407


embedded image







I-408


embedded image







I-409


embedded image







I-410


embedded image







I-411


embedded image







I-412


embedded image







I-413


embedded image







I-414


embedded image







I-415


embedded image







I-416


embedded image







I-417


embedded image







I-418


embedded image







I-419


embedded image







I-420


embedded image







I-421


embedded image







I-422


embedded image







I-423


embedded image







I-424


embedded image







I-425


embedded image







I-426


embedded image







I-427


embedded image







I-428


embedded image







I-429


embedded image







I-430


embedded image







I-431


embedded image







I-432


embedded image







I-433


embedded image







I-434


embedded image







I-435


embedded image







I-436


embedded image







I-437


embedded image







I-438


embedded image







I-439


embedded image







I-443


embedded image







I-444


embedded image







I-445


embedded image







I-446


embedded image







I-447


embedded image







I-448


embedded image







I-449


embedded image







I-450


embedded image







I-451


embedded image







I-452


embedded image







I-453


embedded image







I-454


embedded image







I-455


embedded image







I-456


embedded image







I-457


embedded image







I-458


embedded image







I-459


embedded image







I-460


embedded image







I-461


embedded image







I-462


embedded image







I-463


embedded image







I-464


embedded image







I-465


embedded image







I-466


embedded image







I-467


embedded image







I-468


embedded image







I-469


embedded image







I-470


embedded image







I-471


embedded image







I-472


embedded image







I-473


embedded image







I-474


embedded image







I-475


embedded image







I-476


embedded image







I-477


embedded image







I-478


embedded image







I-479


embedded image







I-480


embedded image







I-481


embedded image







I-482


embedded image







I-483


embedded image







I-484


embedded image







I-485


embedded image







I-486


embedded image







I-487


embedded image







I-488


embedded image







I-489


embedded image







I-490


embedded image







I-491


embedded image







I-492


embedded image







I-493


embedded image







I-494


embedded image







I-495


embedded image







I-496


embedded image







I-497


embedded image







I-498


embedded image







I-499


embedded image







I-500


embedded image







I-501


embedded image







I-502


embedded image







I-503


embedded image







I-504


embedded image







I-505


embedded image







I-506


embedded image







I-507


embedded image







I-508


embedded image







I-509


embedded image







I-510


embedded image







I-511


embedded image







I-512


embedded image







I-513


embedded image







I-514


embedded image







I-515


embedded image







I-516


embedded image







I-517


embedded image







I-518


embedded image







I-519


embedded image







I-520


embedded image







I-521


embedded image







I-522


embedded image







I-524


embedded image







I-525


embedded image







I-526


embedded image







I-527


embedded image







I-528


embedded image







I-529


embedded image







I-530


embedded image







I-531


embedded image







I-532


embedded image







I-533


embedded image







I-534


embedded image







I-535


embedded image







I-536


embedded image







I-537


embedded image







I-538


embedded image







I-539


embedded image







I-540


embedded image







I-541


embedded image







I-542


embedded image







I-543


embedded image







I-544


embedded image







I-545


embedded image







I-546


embedded image







I-547


embedded image







I-548


embedded image







I-549


embedded image







I-550


embedded image







I-551


embedded image







I-552


embedded image







I-553


embedded image







I-554


embedded image







I-555


embedded image







I-556


embedded image







I-557


embedded image







I-558


embedded image







I-559


embedded image







I-560


embedded image







I-561


embedded image







I-562


embedded image







I-563


embedded image







I-564


embedded image







I-565


embedded image







I-566


embedded image







I-567


embedded image







I-568


embedded image







I-569


embedded image







I-570


embedded image







I-571


embedded image







I-572


embedded image







I-573


embedded image







I-574


embedded image







I-575


embedded image







I-576


embedded image







I-577


embedded image







I-578


embedded image







I-579


embedded image







I-580


embedded image







I-581


embedded image







I-582


embedded image







I-583


embedded image







I-584


embedded image







I-585


embedded image







I-586


embedded image







I-587


embedded image







I-588


embedded image







I-589


embedded image







I-590


embedded image







I-591


embedded image







I-592


embedded image







I-593


embedded image







I-594


embedded image







I-595


embedded image







I-596


embedded image







I-597


embedded image







I-598


embedded image







I-599


embedded image







I-600


embedded image







I-601


embedded image







I-602


embedded image







I-603


embedded image







I-604


embedded image







I-605


embedded image







I-606


embedded image







I-607


embedded image







I-608                         I-609


embedded image







I-610


embedded image







I-611


embedded image







I-612


embedded image







I-613


embedded image







I-614


embedded image







I-615


embedded image







I-616


embedded image







I-617


embedded image







I-618


embedded image







I-619


embedded image







I-620


embedded image







I-621


embedded image







I-622


embedded image







I-623


embedded image







I-624


embedded image







I-625


embedded image







I-626


embedded image







I-627


embedded image







I-628


embedded image







I-629


embedded image







I-630


embedded image







I-631


embedded image







I-632


embedded image







I-633


embedded image







I-634


embedded image







I-635


embedded image







I-636


embedded image







I-637


embedded image







I-638


embedded image







I-639


embedded image







I-640


embedded image







I-641


embedded image







I-642


embedded image







I-643


embedded image







I-644


embedded image







I-645


embedded image







I-646


embedded image







I-647


embedded image







I-648


embedded image







I-649


embedded image







I-650


embedded image







I-651


embedded image







I-652


embedded image







I-653


embedded image







I-654


embedded image







I-655


embedded image







I-656


embedded image







I-657


embedded image







I-658


embedded image







I-659


embedded image







I-660


embedded image







I-661


embedded image







I-662


embedded image







I-663


embedded image







I-664


embedded image







I-665


embedded image







I-666


embedded image







I-667


embedded image







I-668


embedded image







I-669


embedded image







I-670


embedded image







I-671


embedded image







I-672


embedded image







I-673


embedded image







I-674


embedded image







I-675


embedded image







I-676


embedded image







I-677


embedded image







I-678


embedded image







I-679


embedded image







I-680


embedded image







I-681                       I-682


embedded image







I-683


embedded image







I-684


embedded image







I-685


embedded image







I-686


embedded image







I-687


embedded image







I-688


embedded image







I-689


embedded image







I-690


embedded image







I-691


embedded image







I-692


embedded image







I-693


embedded image







I-694


embedded image







I-695


embedded image







I-696


embedded image







I-697


embedded image







I-698


embedded image







I-699                           I-700


embedded image







I-701


embedded image







I-702


embedded image







I-703


embedded image







I-704


embedded image







I-705


embedded image







I-706


embedded image







I-707


embedded image







I-708


embedded image







I-709


embedded image







I-710


embedded image







I-711


embedded image







I-712


embedded image







I-713


embedded image







I-714


embedded image







I-715


embedded image







I-716


embedded image







I-717


embedded image







I-718


embedded image







I-719


embedded image







I-720


embedded image







I-721


embedded image







I-722


embedded image







I-723


embedded image







I-724


embedded image







I-725


embedded image







I-726


embedded image







I-727


embedded image







I-728


embedded image







I-729


embedded image







I-730


embedded image







I-731


embedded image







I-732


embedded image







I-733


embedded image







I-734


embedded image







I-735


embedded image







I-736


embedded image







I-737


embedded image







I-738


embedded image







I-739


embedded image







I-740


embedded image







I-741


embedded image







I-742


embedded image







I-743


embedded image







I-744


embedded image







I-745


embedded image







I-746


embedded image







I-747


embedded image







I-748


embedded image







I-749


embedded image







I-750


embedded image







I-751


embedded image







I-752


embedded image







I-753


embedded image







I-754


embedded image







I-755


embedded image







I-756


embedded image







I-757


embedded image







I-758


embedded image







I-759


embedded image







I-760


embedded image







I-761


embedded image







I-762


embedded image







I-763


embedded image







I-764


embedded image







I-765


embedded image







I-766


embedded image







I-767


embedded image







I-768


embedded image







I-769


embedded image







I-770


embedded image







I-771


embedded image







I-772


embedded image







I-773


embedded image







I-774


embedded image







I-775


embedded image







I-776


embedded image







I-778


embedded image







I-779


embedded image







I-780


embedded image







I-781


embedded image







I-782


embedded image







I-783


embedded image







I-784


embedded image







I-785


embedded image







I-786


embedded image







I-787


embedded image







I-788


embedded image







I-789


embedded image







I-790


embedded image







I-791


embedded image







I-792


embedded image







I-793


embedded image







I-794


embedded image







I-795


embedded image







I-796


embedded image







I-797


embedded image







I-798


embedded image







I-799


embedded image







I-800


embedded image







I-801


embedded image







I-802


embedded image







I-803


embedded image







I-804


embedded image







I-805


embedded image







I-806


embedded image







I-807


embedded image







I-808


embedded image







I-809


embedded image







I-810


embedded image







I-811


embedded image







I-812


embedded image







I-813


embedded image







I-814


embedded image







I-815


embedded image







I-816


embedded image







I-817


embedded image







I-818


embedded image







I-819


embedded image







I-820


embedded image







I-821


embedded image







I-822


embedded image







I-823


embedded image







I-824


embedded image







I-825


embedded image







I-826


embedded image







I-827


embedded image







I-828


embedded image







I-829


embedded image







I-830


embedded image







I-831


embedded image







I-832


embedded image







I-833


embedded image







I-834


embedded image







I-835


embedded image







I-836


embedded image







I-837


embedded image







I-838


embedded image







I-839


embedded image







I-840


embedded image







I-841


embedded image







I-842


embedded image







I-843


embedded image







I-844


embedded image







I-845


embedded image







I-846


embedded image







I-847


embedded image







I-848


embedded image







I-849


embedded image







I-850


embedded image







I-851


embedded image







I-852


embedded image







I-853


embedded image







I-854


embedded image







I-855


embedded image







I-856


embedded image







I-857


embedded image







I-858


embedded image







I-859


embedded image







I-860


embedded image







I-861


embedded image







I-862


embedded image







I-863


embedded image







I-864


embedded image







I-865


embedded image







I-866


embedded image







I-867


embedded image







I-868


embedded image







I-869


embedded image







I-870


embedded image







I-871


embedded image







I-872


embedded image







I-873


embedded image







I-874


embedded image







I-875


embedded image







I-876


embedded image







I-877


embedded image







I-878


embedded image







I-879


embedded image







I-880


embedded image







I-881


embedded image







I-882


embedded image







I-883


embedded image







I-884


embedded image







I-885


embedded image







I-886


embedded image







I-887


embedded image







I-888


embedded image







I-889


embedded image







I-890


embedded image







I-891


embedded image











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


3.3. Pharmaceutical Compositions of the Present Compounds

While it is possible that, for use in therapy, a provided compound may be administered as the raw chemical, it is possible to present the provided compound as the active ingredient in a pharmaceutical composition. Such compositions can be prepared in a manner well known in the pharmaceutical art and comprise at least one active compound. Accordingly in one embodiment, the disclosure further provides pharmaceutical compositions comprising a provided compound or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients. The excipient(s) must be acceptable in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipient thereof. In accordance with another aspect of the disclosure there is also provided a process for the preparation of a pharmaceutical composition including a provided compound or pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable excipients. The pharmaceutical composition can be for use in the treatment and/or prophylaxis of any of the conditions described herein.


Generally, a provided compound is administered in a pharmaceutically effective amount. The amount of the compound actually administered will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like. Pharmaceutical compositions may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose. The term “unit dosage forms” refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient, vehicle or carrier. Typical unit dosage forms include prefilled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions.


Preferred unit dosage compositions are those containing a daily dose or sub-dose, or an appropriate fraction thereof, of an active ingredient. Such unit doses may therefore be administered once or more than once a day. Such pharmaceutical compositions may be prepared by any of the methods well known in the pharmacy art.


Pharmaceutical compositions may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, inhaled, intranasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route. Such compositions may be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s).


Pharmaceutical compositions adapted for oral administration may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil liquid emulsions.


For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert excipient such as ethanol, glycerol, water and the like. Powders are prepared by reducing the compound to a suitable fine size and mixing with a similarly prepared pharmaceutical excipient such as an edible carbohydrate, as, for example, starch or mannitol. Flavoring, preservative, dispersing and coloring agent can also be present.


Capsules are made by preparing a powder mixture, as described above, and filling formed gelatin sheaths. Excipients including glidants and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate or solid polyethylene glycol can be added to the powder mixture before the filling operation. A disintegrating or solubilizing agent such as agar-agar, calcium carbonate or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested.


Moreover, when desired or necessary, excipients including suitable binders, glidants, lubricants, sweetening agents, flavors, disintegrating agents and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like. Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant and pressing into tablets. A powder mixture is prepared by mixing a provided compound, suitably comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymethylcellulose, an alginate, gelatin, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/or an absorption agent such as bentonite, kaolin or dicalcium phosphate. The powder mixture can be granulated by wetting with a binder such as syrup, starch paste, acadia mucilage or solutions of cellulosic or polymeric materials and forcing through a screen. As an alternative to granulating, the powder mixture can be run through the tablet machine and the result is imperfectly formed slugs broken into granules. The granules can be lubricated to prevent sticking to the tablet forming dies by means of the addition of stearic acid, a stearate salt, talc or mineral oil. The lubricated mixture is then compressed into tablets. A provided compound can also be combined with a free flowing inert carrier and compressed into tablets directly without going through the granulating or slugging steps. A clear or opaque protective coating consisting of a sealing coat of shellac, a coating of sugar or polymeric material and a polish coating of wax can be provided. Dyestuffs can be added to these coatings to distinguish different unit dosages.


Oral fluids such as solution, suspensions, syrups and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of the compound. Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic alcoholic vehicle. Suspensions can be formulated by dispersing the compound in a non-toxic vehicle. Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and polyoxy ethylene sorbitol ethers, preservatives, flavor additive such as peppermint oil or natural sweeteners or saccharin or other artificial sweeteners, and the like can also be added.


Where appropriate, dosage unit compositions for oral administration can be microencapsulated. The composition can also be prepared to prolong or sustain the release as for example by coating or embedding particulate material in polymers, wax or the like.


The compounds of the disclosure may also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines. Pharmaceutical compositions adapted for transdermal administration may be presented as discrete patches intended to remain in intimate contact with the epidermis of the recipient for a prolonged period of time.


Pharmaceutical compositions adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils.


For treatments of the eye or other external tissues, for example mouth and skin, the compositions are preferably applied as a topical ointment or cream. When formulated in an ointment, the active ingredient may be employed with either a paraffinic or a water-miscible ointment base. Alternatively, the active ingredient may be formulated in a cream with an oil-in-water cream base or a water-in-oil base.


Pharmaceutical compositions adapted for topical administrations to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent.


Pharmaceutical compositions adapted for topical administration in the mouth include lozenges, pastilles and mouth washes.


Pharmaceutical compositions adapted for rectal administration may be presented as suppositories or as enemas.


Dosage forms for nasal or inhaled administration may conveniently be formulated as aerosols, solutions, suspension drops, gels or dry powders.


Compositions for intranasal administration include aqueous compositions administered to the nose by drops or by pressurized pump. Suitable compositions contain water as the diluent or carrier for this purpose. Compositions for administration to the lung or nose may contain one or more excipients, for example one or more suspending agents, one or more preservatives, one or more surfactants, one or more tonicity adjusting agents, one or more co-solvents, and may include components to control the pH of the composition, for example a buffer system. Further, the compositions may contain other excipients such as antioxidants, for example sodium metabisulphite, and taste-masking agents. Compositions may also be administered to the nose or other regions of the respiratory tract by nebulization. Intranasal compositions may permit a provided compound or pharmaceutically acceptable salt thereof to be delivered to all areas of the nasal cavities (the target tissue) and further, may permit the provided compound or pharmaceutically acceptable salt thereof to remain in contact with the target tissue for longer periods of time. A suitable dosing regimen for intranasal compositions would be for the patient to inhale slowly through the nose subsequent to the nasal cavity being cleared. During inhalation, the composition would be administered to one nostril while the other is manually compressed. This procedure would then be repeated for the other nostril. Typically, one or two sprays per nostril would be administered by the above procedure one, two, or three times each day, ideally once daily. Of particular interest are intranasal compositions suitable for once-daily administration.


The suspending agent(s), if included, will typically be present in an amount of from 0.1 to 5% (w/w), such as from 1.5% to 2.4% (w/w), based on the total weight of the composition. Examples of pharmaceutically acceptable suspending agents include, but are not limited to, Avicef (microcrystalline cellulose and carboxymethylcellulose sodium), carboxymethylcellulose sodium, veegum, tragacanth, bentonite, methylcellulose, xanthan gum, carbopol and polyethylene glycols.


Compositions for administration to the lung or nose may contain one or more excipients may be protected from microbial or fungal contamination and growth by inclusion of one or more preservatives. Examples of pharmaceutically acceptable antimicrobial agents or preservatives include, but are not limited to, quaternary ammonium compounds (for example benzalkonium chloride, benzethonium chloride, cetrimide, cetylpyridinium chloride, lauralkonium chloride and myristyl picolinium chloride), mercurial agents (for example phenylmercuric nitrate, phenylmercuric acetate and thimerosal), alcoholic agents (for example chlorobutanol, phenylethyl alcohol and benzyl alcohol), antibacterial esters (for example esters of p-hydroxybenzoic acid), chelating agents such as disodium edetate (EDTA) and other antimicrobial agents such as chlorhexidine, chlorocresol, sorbic acid and its salts (such as potassium sorbate) and polymyxin. Examples of pharmaceutically acceptable antifungal agents or preservatives include, but are not limited to, sodium benzoate, sorbic acid, sodium propionate, methylparaben, ethylparaben, propylparaben, and butylparaben. The preservative(s), if included, may be present in an amount of from 0.001 to 1% (w/w), such as from 0.015% to 0.5% (w/w) based on the total weight of the composition. Compositions (for example wherein at least one compound is in suspension) may include one or more surfactants which functions to facilitate dissolution of the medicament particles in the aqueous phase of the composition. For example, the amount of surfactant used is an amount which will not cause foaming during mixing. Examples of pharmaceutically acceptable surfactants include fatty alcohols, esters, and ethers, such as polyoxyethylene (20) sorbitan monooleate (Polysorbate 80), macrogol ethers, and poloxamers. The surfactant may be present in an amount of between about 0.01 to 10% (w/w), such as from 0.01 to 0.75% (w/w), for example about 0.5% (w/w), based on the total weight of the composition.


One or more tonicity-adjusting agent(s) may be included to achieve tonicity with body fluids (e.g., fluids of the nasal cavity) resulting in reduced levels of irritancy. Examples of pharmaceutically acceptable tonicity-adjusting agents include, but are not limited to, sodium chloride, dextrose, xylitol, calcium chloride, glucose, glycerine, and sorbitol. A tonicity-adjusting agent, if present, may be included in an amount of from 0.1 to 10% (w/w), such as from 4.5 to 5.5% (w/w), for example about 5.0% (w/w), based on the total weight of the composition.


The compositions of the disclosure may be buffered by the addition of suitable buffering agents such as sodium citrate, citric acid, trometamol, phosphates such as disodium phosphate (e.g., dodecahydrate, heptahydrate, dihydrate and anhydrous forms), or sodium phosphate and mixtures thereof.


A buffering agent, if present, may be included in an amount of from 0.1 to 5% (w/w), for example 1 to 3% (w/w) based on the total weight of the composition.


Examples of taste-masking agents include sucralose, sucrose, saccharin or a salt thereof, fructose, dextrose, glycerol, corn syrup, aspartame, acesulfame-K, xylitol, sorbitol, erythritol, ammonium glycyrrhizinate, thaumatin, neotame, mannitol, menthol, eucalyptus oil, camphor, a natural flavoring agent, an artificial flavoring agent, and combinations thereof.


One or more co-solvent may be included to aid solubility of the medicament compound(s) and/or other excipients. Examples of pharmaceutically acceptable co-solvents include, but are not limited to, propylene glycol, dipropylene glycol, ethylene glycol, glycerol, ethanol, polyethylene glycols (for example PEG300 or PEG400), and methanol. In one embodiment, the co-solvent is propylene glycol.


Co-solvent(s), if present, may be included in an amount of from 0.05 to 30% (w/w), such as from 1 to 25% (w/w), for example from 1 to 10% (w/w) based on the total weight of the composition.


Compositions for inhaled administration include aqueous, organic or aqueous/organic mixtures, dry powder or crystalline compositions administered to the respiratory tract by pressurized pump or inhaler, for example, reservoir dry powder inhalers, unit-dose dry powder inhalers, pre-metered multi-dose dry powder inhalers, nasal inhalers or pressurized aerosol inhalers, nebulizers or insufflators. Suitable compositions contain water as the diluent or carrier for this purpose and may be provided with conventional excipients such as buffering agents, tonicity modifying agents and the like. Aqueous compositions may also be administered to the nose and other regions of the respiratory tract by nebulization. Such compositions may be aqueous solutions or suspensions or aerosols delivered from pressurized packs, such as a metered dose inhaler, with the use of a suitable liquefied propellant.


Compositions for administration topically to the nose (for example, for the treatment of rhinitis) or to the lung, include pressurized aerosol compositions and aqueous compositions delivered to the nasal cavities by pressurized pump. Compositions which are non-pressurized and are suitable for administration topically to the nasal cavity are of particular interest. Suitable compositions contain water as the diluent or carrier for this purpose. Aqueous compositions for administration to the lung or nose may be provided with conventional excipients such as buffering agents, tonicity-modifying agents and the like. Aqueous compositions may also be administered to the nose by nebulization.


A fluid dispenser may typically be used to deliver a fluid composition to the nasal cavities. The fluid composition may be aqueous or non-aqueous, but typically aqueous. Such a fluid dispenser may have a dispensing nozzle or dispensing orifice through which a metered dose of the fluid composition is dispensed upon the application of a user-applied force to a pump mechanism of the fluid dispenser. Such fluid dispensers are generally provided with a reservoir of multiple metered doses of the fluid composition, the doses being dispensable upon sequential pump actuations. The dispensing nozzle or orifice may be configured for insertion into the nostrils of the user for spray dispensing of the fluid composition into the nasal cavity.


Dry powder compositions for topical delivery to the lung by inhalation may, for example, be presented in capsules and cartridges of for example gelatin, or blisters of for example laminated aluminum foil, for use in an inhaler or insufflator. Powder blend compositions generally contain a powder mix for inhalation of a provided compound or pharmaceutically acceptable salt thereof and a suitable powder base (carrier/diluent/excipient substance) such as mono-, di-, or polysaccharides (e.g., lactose or starch). Dry powder compositions may also include, in addition to the drug and carrier, a further excipient (e.g., a ternary agent such as a sugar ester for example cellobiose octaacetate, calcium stearate, or magnesium stearate.


Pharmaceutical compositions adapted for parental administration include aqueous and nonaqueous sterile injection solutions which may contain antioxidants, buffers, bacteriostats and solutes which render the composition isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The compositions may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (e.g., lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules, and tablets.


It should be understood that in addition to the ingredients particularly mentioned above, the compositions may include other agents conventional in the art having regard to the type of formulation in question, for example, those suitable for oral administration may include flavoring agents.


3.4. Uses of the Present Pharmaceutical Compositions and Compounds in Therapy

A therapeutically effective amount of the agent will depend upon a number of factors including, for example, the age and weight of the subject, the precise condition requiring treatment and its severity, the nature of the formulation, and the route of administration, and will ultimately be at the discretion of the attendant physician or veterinarian. In particular, the subject to be treated is a mammal, particularly a human.


The agent may be administered in a daily dose. This amount may be given in a single dose per day or more usually in a number (e.g., two, three, four, five, or six) of sub-doses per day such that the total daily dose is the same.


Suitably, the amount of the compound of the present disclosure administered may be an amount selected from 0.01 mg to 10 g per day (calculated as the free or unsalted compound).


In some embodiments, a provided compound or a pharmaceutically acceptable salt thereof may be employed alone or in combination with other therapeutic agents. A provided compound or a pharmaceutically acceptable salt thereof and the other pharmaceutically active agent(s) may be administered together or separately and, when administered separately, administration may occur simultaneously or sequentially, in any order, by any convenient route in separate or combined pharmaceutical compositions. The amounts of a provided compound or a pharmaceutically acceptable salt thereof and the other pharmaceutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect. A provided compound or a pharmaceutically acceptable salt thereof and further therapeutic agent(s) may be employed in combination by administration simultaneously in a unitary pharmaceutical composition including both compounds. Alternatively, the combination may be administered separately in separate pharmaceutical compositions, each including one of the compounds in a sequential manner wherein, for example, the compound of the present disclosure is administered first and the other second and vice versa. Such sequential administration may be close in time (e.g. simultaneously) or remote in time. Furthermore, it does not matter if the compounds are administered in the same dosage form, e.g., one compound may be administered topically and the other compound may be administered orally. Suitably, both compounds are administered orally.


The combinations may be presented as a combination kit. By the term “combination kit” “or kit of parts” as used herein is meant the pharmaceutical composition or compositions that are used to administer the combination according to the present disclosure. When both compounds are administered simultaneously, the combination kit can contain both compounds in a single pharmaceutical composition, such as a tablet, or in separate pharmaceutical compositions. When the compounds are not administered simultaneously, the combination kit will contain each compound in separate pharmaceutical compositions either in a single package or in separate pharmaceutical compositions in separate packages. The combination kit can also be provided by instruction, such as dosage and administration instructions. Such dosage and administration instructions can be of the kind that are provided to a doctor, for example by a drug product label, or they can be of the kind that are provided by a doctor, such as instructions to a patient.


When the combination is administered separately in a sequential manner wherein one is administered first and the other second or vice versa, such sequential administration may be close in time or remote in time. For example, administration of the other agent several minutes to several dozen minutes after the administration of the first agent, and administration of the other agent several hours to several days after the administration of the first agent are included, wherein the lapse of time is not limited. For example, one agent may be administered once a day, and the other agent may be administered 2 or 3 times a day, or one agent may be administered once a week, and the other agent may be administered once a day and the like. It will be clear to a person skilled in the art that, where appropriate, the other therapeutic ingredients(s) may be used in the form of salts, for example as alkali metal or amine salts or as acid addition salts, or prodrugs, or as esters, for example lower alkyl esters, or as solvates, for example hydrates, to optimize the activity and/or stability and/or physical characteristics, such as solubility, of the therapeutic ingredient. It will be clear also that, where appropriate, the therapeutic ingredients may be used in optically pure form.


When combined in the same composition it will be appreciated that the two compounds must be stable and compatible with each other and the other components of the composition and may be formulated for administration. When formulated separately they may be provided in any convenient composition, conveniently, in such a manner as known for such compounds in the art.


When a provided compound or a pharmaceutically acceptable salt thereof is used in combination with a second therapeutic agent active against the same disease, condition, or disorder, the dose of each compound may differ from that when the compound is used alone. Appropriate doses will be readily appreciated by those skilled in the art.


In one embodiment, the mammal in the methods and uses of the present disclosure is a human. The provided compounds or pharmaceutically acceptable salts thereof are useful in the treatment of diseases and conditions in which modulation of cGAS is beneficial. As modulators of the immune response, a provided compound or a pharmaceutically acceptable salts thereof may also be useful, as stand-alone, in combination or as adjuvants, in the treatment of diseases and conditions in which modulation of cGAS is beneficial.


In one embodiment, the disease or condition is an inflammatory, allergic, or autoimmune diseases such as systemic lupus erythematosus, psoriasis, insulin-dependent diabetes mellitus (IDDM), scleroderma, Aicardi Goutibres syndrome, dermatomyositis, inflammatory bowel diseases, multiple sclerosis, rheumatoid arthritis, and Sjogren's syndrome (SS).


In another embodiment, the disease or condition is an infectious disease such as bacterial, viral or parasitic disease in which modulation of cGAS activity is beneficial.


In another embodiment, the disease or condition is a senescence- or age-related disease, including a neurodegenerative disease such as Alzheimer's or Parkinson disease, cardiovascular diseases such as atherosclerosis or myocardial infarction, liver or renal diseases, cancer, or premature aging.


Inflammation represents a group of vascular, cellular, and neurological responses to trauma. Inflammation can be characterized as the movement of inflammatory cells such as monocytes, neutrophils, and granulocytes into the tissues. This is usually associated with reduced endothelial barrier function and edema into the tissues. Inflammation can be classified as either acute or chronic. Acute inflammation is the initial response of the body to harmful stimuli and is achieved by the increased movement of plasma and leukocytes from the blood into the injured tissues. A cascade of biochemical event propagates and matures the inflammatory response, involving the local vascular system, the immune system, and various cells within the injured tissue. Prolonged inflammation, known as chronic inflammation, leads to a progressive shift in the type of cells which are present at the site of inflammation and is characterized by simultaneous destruction and healing of the tissue from the inflammatory process.


When occurring as part of an immune response to infection or as an acute response to trauma, inflammation can be beneficial and is normally self-limiting. However, inflammation can be detrimental under various conditions. This includes the production of excessive inflammation in response to infectious agents, which can lead to significant organ damage and death (e.g., in the setting of sepsis). Moreover, chronic inflammation is generally deleterious and is at the root of numerous chronic diseases, causing severe and irreversible damage to tissues. In such settings, the immune response is often directed against self-tissues (autoimmunity), although chronic responses to foreign entities can also lead to bystander damage to self-tissues. The aim of anti-inflammatory therapy is therefore to reduce this inflammation, to inhibit autoimmunity when present and to allow for the physiological process or healing and tissue repair to progress.


In some embodiments, a provided compound or a pharmaceutically acceptable salt thereof may be used to treat inflammation of any tissue and organs of the body, including musculoskeletal inflammation, vascular inflammation, neural inflammation, digestive system inflammation, ocular inflammation, inflammation of the reproductive system, and other inflammation, as exemplified below.


Musculoskeletal inflammation refers to any inflammatory condition of the musculoskeletal system, particularly those conditions affecting skeletal joints, including joints of the hand, wrist, elbow, shoulder, jaw, spine, neck, hip, knew, ankle, and foot, and conditions affecting tissues connecting muscles to bones such as tendons. Examples of musculoskeletal inflammation which may be treated with compounds of the present disclosure include arthritis (including, for example, osteoarthritis, rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, acute and chronic infectious arthritis, arthritis associated with gout and pseudogout, and juvenile idiopathic arthritis), tendonitis, synovitis, tenosynovitis, bursitis, fibrositis (fibromyalgia), epicondylitis, myositis, and osteitis (including, for example, Paget's disease, osteitis pubis, and osteitis fibrosa cystic). Ocular inflammation refers to inflammation of any structure of the eye, including the eye lids. Examples of ocular inflammation which may be treated with the compounds of the present disclosure include blepharitis, blepharochalasis, conjunctivitis, dacryoadenitis, keratitis, keratoconjunctivitis sicca (dry eye), scleritis, trichiasis, and uveitis. Examples of inflammation of the nervous system which may be treated with the compounds of the present disclosure include encephalitis, Guillain-Barre syndrome, meningitis, neuromyotonia, narcolepsy, multiple sclerosis, myelitis, and schizophrenia.


Examples of inflammation of the vasculature or lymphatic system which may be treated with a provided compound or a pharmaceutically acceptable salt thereof include arthrosclerosis, arthritis, phlebitis, vasculitis, and lymphangitis.


Examples of inflammatory conditions of the digestive system which may be treated with a provided compound or a pharmaceutically acceptable salt thereof include cholangitis, cholecystitis, enteritis, enterocolitis, gastritis, gastroenteritis, inflammatory bowel disease (such as Crohn's disease and ulcerative colitis), ileitis, and proctitis.


Examples of inflammatory conditions of the reproductive system which may be treated with a provided compound or a pharmaceutically acceptable salt thereof include cervicitis, chorioamnionitis, endometritis, epididymitis, omphalitis, oophoritis, orchitis, salpingitis, tubo-ovarian abscess, urethritis, vaginitis, vulvitis, and vulvodynia.


The agents may be used to treat autoimmune conditions having an inflammatory component. Such conditions include systemic lupus erythematosus, acute disseminated alopecia universalise, Behcet's disease, Chagas' disease, chronic fatigue syndrome, dysautonomia, encephalomyelitis, ankylosing spondylitis, aplastic anemia, hidradenitis suppurativa, autoimmune hepatitis, autoimmune oophoritis, celiac disease, Crohn's disease, diabetes mellitus type 1, giant cell arteritis, goodpasture's syndrome. Grave's disease, Guillain-Barre syndrome, Hashimoto's disease, Henoch-Schonlein purpura, Kawasaki's disease, microscopic colitis, microscopic polyarteritis, mixed connective tissue disease, multiple sclerosis, myasthenia gravis, opsoclonus myoclonus syndrome, optic neuritis, ord's thyroiditis, pemphigus, polyarteritis nodosa, polymyalgia, rheumatoid arthritis, Reiter's syndrome, Sjogren's syndrome, Aicardi Goutibres syndrome, temporal arteritis, Wegener's granulomatosis, warm autoimmune haemolytic anemia, interstitial cystitis, lyme disease, morphea, psoriasis, sarcoidosis, scleroderma, ulcerative colitis, and vitiligo.


In some embodiments, a provided compound or a pharmaceutically acceptable salt thereof may be used to treat T-cell mediated hypersensitivity diseases having an inflammatory component. Such conditions include contact hypersensitivity, contact dermatitis (including that due to poison ivy), uticaria, skin allergies, respiratory allergies (hayfever, allergic rhinitis) and gluten-sensitive enteropathy (Celliac disease).


Other inflammatory conditions which may be treated with a provided compound or a pharmaceutically acceptable salt thereof include, for example, appendicitis, dermatitis, dermatomyositis, endocarditis, fibrositis, gingivitis, glossitis, hepatitis, hidradenitis suppurativa, iritis, laryngitis, mastitis, myocarditis, nephritis, otitis, pancreatitis, parotitis, percarditis, peritonoitis, pharyngitis, pleuritis, pneumonitis, prostatistis, pyelonephritis, and stomatisi, transplant rejection (involving organs such as kidney, liver, heart, lung, pancreas (e.g., islet cells), bone marrow, cornea, small bowel, skin allografts, skin homografts, and heart valve xenografts, serum sickness, and graft vs host disease), acute pancreatitis, chronic pancreatitis, acute respiratory distress syndrome. Sexary's syndrome, congenital adrenal hyperplasis, nonsuppurative thyroiditis, hypercalcemia associated with cancer, pemphigus, bullous dermatitis herpetiformis, severe erythema multiforme, exfoliative dermatitis, seborrheic dermatitis, seasonal or perennial allergic rhinitis, bronchial asthma, contact dermatitis, atopic dermatitis, drug hypersensitivity reactions, allergic conjunctivitis, keratitis, herpes zoster ophthalmicus, iritis and oiridocyclitis, chorioretinitis, optic neuritis, symptomatic sarcoidosis, fulminating or disseminated pulmonary tuberculosis chemotherapy, idiopathic thrombocytopenic purpura in adults, secondary thrombocytopenia in adults, acquired (autoimmune) haemolytic anemia, leukemia and lymphomas in adults, acute leukemia of childhood, regional enteritis, autoimmune vasculitis, multiple sclerosis, chronic obstructive pulmonary disease, solid organ transplant rejection, sepsis. Preferred treatments include treatment of transplant rejection, rheumatoid arthritis, psoriatic arthritis, multiple sclerosis. Type 1 diabetes, asthma, inflammatory bowel disease, systemic lupus erythematosis, psoriasis, chronic pulmonary disease, and inflammation accompanying infectious conditions (e.g., sepsis).


In some embodiments, the disclosure provides a provided compound or a pharmaceutically acceptable salt thereof for use in the treatment of an inflammatory, allergic, or autoimmune disease.


In some embodiments, the disclosure provides a method of treating an inflammatory, allergic, or autoimmune disease comprising: administering to a patient in need thereof a therapeutically effective amount of a provided compound or a pharmaceutically acceptable salt thereof.


In some embodiments, the disclosure provides the use of a provided compound or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of an inflammatory, allergic, or autoimmune disease.


In some embodiments, a provided compound or a pharmaceutically acceptable salts thereof may be used in combination with one or more other agents in the prevention or treatment of an allergic inflammatory autoimmune disease, wherein such other agents can include: antigen immunotherapy agents; antihistamines; steroids, NSAIDs; bronchodilators (e.g. beta 2 agonists, adrenergic agonists, anticholinergic agents, theophylline); methotrexate; leukotriene modulators; monoclonal antibody agents such as anti-lgE, anti-TNF, anti-IL-5, anti-IL-6, anti-IL-12, anti-IL-1 and similar agents; receptor therapies agents such as entanercept; and antigen non-specific immunotherapeutic agents such interferon or other cytokines/chemokines, cytokine/chemokine receptor modulators, cytokine agonists or antagonists, and TLR antagonist.


In some embodiments, the present disclosure provides a pharmaceutical composition comprising a provided compound or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent useful in the treatment of an allergic, inflammatory, or autoimmune disease.


In some embodiments, the present disclosure provides a pharmaceutical composition comprising a provided compound or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent useful in the treatment of an allergic, inflammatory, or autoimmune disease for use in therapy.


In some embodiments, the present disclosure provides a pharmaceutical composition comprising a provided compound or pharmaceutically acceptable salt thereof, and at least one further therapeutic agent useful in the treatment of an allergic inflammatory or autoimmune disease, for use in the treatment of allergic, inflammatory, or autoimmune disease.


In some embodiments, the present disclosure provides the use of a pharmaceutical composition comprising a provided compound or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent useful in the treatment of an allergic, inflammatory, or autoimmune disease in the manufacture of a medicament for the treatment of an allergic, inflammatory or autoimmune disease.


In some embodiments, the present disclosure provides a method of treating an allergic, inflammatory or autoimmune disease comprising: administering to a patient in need thereof a therapeutically effective amount of a pharmaceutical composition comprising a provided compound or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent useful in the treatment of an allergic, inflammatory, or autoimmune disease.


In some embodiments, the present disclosure provides a pharmaceutical composition comprising a provided compound or a pharmaceutically acceptable salt thereof, at least one further therapeutic agent useful in the treatment of an allergic, inflammatory, or autoimmune disease, and one or more of pharmaceutically acceptable excipients.


In some embodiments, the present disclosure provides a provided compound or a pharmaceutically acceptable salt thereof, for use in the treatment of an infectious disease.


In some embodiments, the present disclosure provides a method of treating an infectious disease comprising administering to a patient in need thereof a therapeutically effective amount of a provided compound or a pharmaceutically acceptable salt thereof.


In some embodiments, the present disclosure provides the use of a provided compound or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of an infectious disease. In one embodiment, a compound of the present disclosure may be employed with other therapeutic methods of treating infectious disease. In particular, bacterial and parasite infections, such as Mycobacterium tuberculosis and malaria, respectively, which exploit the type-I interferon pathway for their advantage, may be treated with a cGAS inhibitor.


In some embodiments, a provided compound or a pharmaceutically acceptable salts thereof may be used in combination with one or more agents useful in the prevention or treatment of bacterial and viral infections. Examples of such agents include: polymerase inhibitors; replication inhibitors such as acyclovir, famciclovir, ganciclovir, cidofovir and lamivudine; protease inhibitors such as the HIV protease inhibitors saquinavir, ritonavir, indinavir, nelfinavir, amprenavir, fosamprenavir, brecanavir, atazanavir, tipranavir, palinavir, lasinavir, and the HCV protease inhibitors BILN2061, VX-950, SCH503034; nucleoside and nucleotide reverse transcriptase inhibitors such as zidovudine, didanosine, lamivudine, zalcitabine, abacavir, stavidine, adefovir, adefovir dipivoxil, fozivudine, todoxil, emtricitabine, alovudine, amdoxovir, and elvucitabine; non-nucleoside reverse transcriptase inhibitors (including an agent having antioxidation activity such as immunocal or oltipraz) such as nevirapine, delavirdine, efavirenz, loviride, immunocal, oltipraz, capravirine, TMC-278, TMC-125, and etravirine; entry inhibitors such as enfuvirtide (T-20), T-1249, PRO-542, PRO-140, TNX-355, BMS-806, 5-Helix and similar agents; integrase inhibitors such as L-870 and 180; budding inhibitors such as PA-344 and PA-457; chemokine receptor inhibitors such as vicriviroc (Sch-C), Sch-D, TAK779, maraviroc (UK-427,857), and TAK449; neuraminidase inhibitors such as CS-8958, zanamivir, oseltamivir, and peramivir; ion channel blockers such as amantadine or rimantadine; interfering RNA and antisense oligonucleotides and such as ISIS-14803; and antiviral agents of undetermined mechanism of action, such as ribavirin.


In some embodiments, a provided compound or a pharmaceutically acceptable salts thereof may also be used in combination with one or more other agents which may be useful in the prevention or treatment of viral infections such as immune therapies (e.g. interferon or other cytokines/chemokines, cytokine/chemokine receptor modulators, cytokine agonists or antagonists and similar agents); therapeutic vaccines; antifibrotic agents; and antiinflammatory agents such as corticosteroids or NSAIDs (non-steroidal antiinflammatory agents).


In some embodiments, the present disclosure provides a pharmaceutical composition comprising a provided compound or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent useful in the treatment of an infectious disease.


In some embodiments, the present disclosure provides a pharmaceutical composition comprising a provided compound or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent useful in the treatment of an infectious disease for use in therapy.


In some embodiments, the present disclosure provides a pharmaceutical composition comprising a provided compound or pharmaceutically acceptable salt thereof, and at least one further therapeutic agent useful in the treatment of an infectious disease, for use in the treatment of an infectious disease.


In some embodiments, the present disclosure provides the use of a pharmaceutical composition comprising a provided compound or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent useful in the treatment of an infectious disease in the manufacture of a medicament for the treatment of an infectious disease.


In some embodiments, the present disclosure provides a method of treating an infectious disease comprising administering to a patient in need thereof, a therapeutically effective amount of a pharmaceutical composition comprising a provided compound or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent useful in the treatment of an infectious disease.


In some embodiments, the present disclosure provides a pharmaceutical composition comprising a provided compound or a pharmaceutically acceptable salt thereof, at least one further therapeutic agent useful in the treatment of infectious disease, and one or more of pharmaceutically acceptable excipients.


In some embodiments, the disclosure provides a provided compound or a pharmaceutically acceptable salt thereof for use in the treatment of a senescence- or age-related disease.


In some embodiments, the disclosure provides a method of treating a senescence- or age-related disease comprising: administering to a patient in need thereof a therapeutically effective amount of a provided compound or a pharmaceutically acceptable salt thereof.


In some embodiments, the disclosure provides the use of a provided compound or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of a senescence- or age-related disease.


In some embodiments, the present disclosure provides a pharmaceutical composition comprising a provided compound or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent useful in the treatment of a senescence- or age-related disease.


In some embodiments, the present disclosure provides a pharmaceutical composition comprising a provided compound or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent useful in the treatment of a senescence- or age-related disease for use in therapy.


In some embodiments, the present disclosure provides a pharmaceutical composition comprising a provided compound or pharmaceutically acceptable salt thereof, and at least one further therapeutic agent useful in the treatment of a senescence- or age-related disease, for use in the treatment of a senescence- or age-related disease.


In some embodiments, the present disclosure provides the use of a pharmaceutical composition comprising a provided compound or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent useful in the treatment of a senescence- or age-related disease in the manufacture of a medicament for the treatment of a senescence- or age-related disease.


In some embodiments, the present disclosure provides a method of treating a senescence- or age-related disease comprising administering to a patient in need thereof, a therapeutically effective amount of a pharmaceutical composition comprising a provided compound or a pharmaceutically acceptable salt thereof, and at least one further therapeutic agent useful in the treatment of a senescence- or age-related disease.


In some embodiments, the present disclosure provides a pharmaceutical composition comprising a provided compound or a pharmaceutically acceptable salt thereof, at least one further therapeutic agent useful in the treatment of a senescence- or age-related disease, and one or more of pharmaceutically acceptable excipients.


The provided compounds may be prepared by methods known in the art of organic synthesis as set forth in the schemes below and/or the specific Examples described below. In all of the methods, it is well understood that protecting groups for sensitive or reactive groups may be employed where necessary in accordance with general principles of chemistry. Protecting groups are manipulated according to standard methods of organic synthesis (T. W. Green and P. G. M. Wuts (1999) Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons). These groups are removed at a convenient stage of the compound synthesis using methods that are readily apparent to those skilled in the art. The selection of processes as well as the reaction conditions and order of their execution shall be consistent with the preparation of the provided compounds.


The following list provides definitions of certain abbreviations as used herein. It will be appreciated that the list is not exhaustive, but the meaning of those abbreviations not herein below defined will be readily apparent to those skilled in the art: AIBN is 2,2′-azobisisobutyronitrile; ATP is adenosine 5′-triphosphate; BPO is benzoyl peroxide; n-BuLi is n-butyllithium; BzCl is benzoyl chloride; CDI is 1,1′-carbonyldiimidazole; cGAS is cyclic GMP-AMP synthase; CO is carbon monooxide; Cu(OAc)2 is copper(II) acetate; CuCN is copper(I) cynide; CuI is copper(I) iodide; DAST is (diethylamino)sulfur trifluoride; DBU is 1,8-diazabicyclo[5.4.0]undec-7-ene; DCE is dichloroethane; DCM is dichloromethane; DDQ is 2,3-dichloro-5,6-dicyano-p-benzoquinone; DHP is 3,4-dihydro-2H-pyran; DIAD is diisopropyl azodicarboxylate; DIBAL-H is diisobutylaluminum hydride; DIPA is diisopropylamine; DIPEA is N,N-diisopropylethylamine; DMAP is 4-(dimethylamino)pyridine; DMB is 2,4-dimethoxybenzyl; DMF is N,N-dimethylformamide; DMP is Dess-Martin periodinane; DMSO is dimethyl sulfoxide; EA is ethyl acetate; EtMgBr is ethylmagnesium bromide; Et2O is diethyl ether; EtOH is ethanol; GTP is guanosine triphosphate; HCl is hydrochloric acid; HMTA is hexamethylenetetramine; HOAc is acetic acid; HPLC is high performance liquid chromatography; LAH is lithium aluminum hydride; mCPBA is 3-chloroperbenzoic acid; MeCN is acetonitrile; Mel is iodomethane; MeOH is methanol; MeMgBr is methylmagnesium bromide; MOMCl is chloromethyl methyl ether; MOM is methoxymethoxy; MS is mass spectrometer or mass spectrum; MsCl is methanesulfonyl chloride; MTBE is methyl tert-butyl ether; NaH is sodium hydride; NaOH is sodium hydroxide; NBS is N-bromosuccinimide; NMM is N-methylmorpholine; NMR is nuclear magnetic resonance; Pd(dba)2 is bis(dibenzylideneacetone)palladium(O); Pd(dppf)Cl2 is [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II); Pd(OAc)2 is palladium(II) acetate; Pd(PPh3)2Cl2 is bis(triphenylphosphine)palladium(II) dichloride; Pd(PPh3)4 is tetrakis(triphenylphosphine) palladium(O); Pd/C is palladium on carbon; PDC is pridinium dichromate; PE is petroleum ether; PMB is 4-methoxybenzyl; PPh3 is triphenylphosphine; prep-HPLC is preparative high performance liquid chromatography; prep-TLC is preparative thin-layer chromatography; Py is pyridine; TBAF is tetra-n-butylammonium fluoride; TBSCl is tert-butyldimethylsilyl chloride; TEA is triethylamine; TFA is trifluoroacetic acid; THF is tetrahydrofuran; THP is tetrahydropyranyl; TLC is thin-layer chromatography; TSA is p-toluenesulfonic acid monohydrate, and TsCl is p-toluenesulfonyl chloride.


4. EXAMPLES

The following Examples provide syntheses of the provided compounds and their in vitro activity.


Intermediates
Preparation of 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (Intermediate 1)



embedded image


Step 1: Preparation of ethyl 3-((2,3-dichlorophenyl)amino)-3-oxopropanoate. 2,3-dichloroaniline (10.33 g, 64 mmol, 1.0 eq) was mixed with diethyl malonate 1-2 (24 mL, 24.3 g, 158 mmol) and heated at 180° C. for 16 hours until product formation ceased (confirmed by LC-MS). The resulting oil mixture was used in the next step. MS (ES): [M+1]+ 276.


Step 2: Preparation of 3-((2,3-dichlorophenyl)amino)-3-oxopropanoic acid. The mixture of the first step was diluted with MeOH (80 mL) and water (80 mL) and chilled with an ice-water bath. To the above solution was added a solution of NaOH (12 g, ˜2 eq to the amount of diethyl malonate) in 50 mL water slowly. After stirring 30 to 60 min, solids precipitated. Water was added to help stirring. After stirring for 3 to 4 hours at room temperature, starting material was consumed (confirmed by LC-MS), the reaction mixture was acidified with conc. HCl to pH 1 to form a precipitate. After filtration, rinsing with water, and drying under vacuum, the titled compound (10.57 g) was used in the next step without further purification. MS (ES): [M+1]+ 248.


Step 3: Preparation of 7,8-dichloroquinoline-2,4(1H,3H)-dione. The product of step 2 (10.57 g) was suspended and stirred with PPA (55 g) at 140° C. for 3 hours until all solids were dissolved. After starting material was consumed (confirmed by LC-MS), the reaction mixture was quenched with ice. The formed solids were collected by filtration and rinsed with water. After drying under vacuum, the titled compound (17 g) was used in the next step without further purification. MS (ES): [M+1]+ 230


Step 4: Preparation of 2,4,7,8-tetrachloroquinoline (1.6). The product of step 3 was suspended in POCl3 (45 g) and heated at 130° C. until all solids were dissolved. After starting material was consumed (confirmed by LC-MS), excess POCl3 was removed by under vacuum. The residue was treated with ice-water (exothermic) to afford solids and the suspended solids were stirred for 4 hours. Filtration, rinsing with water, and drying under vacuum afforded the crude product as a brown solid (9.25 g) that was suspended in hexane (400 mL) and heated at reflux. After hot filtration to remove residual solids and cooling slowly to room temperature, 5.73 g of the titled compound was obtained as a light brown solid. The solids concentrated from the filtrate and undissolved solids from the above recrystallization were purified by a flash silica column to afford additional product (1.7 g). MS (ES): [M+1]+ 266.0. 1H NMR (400 Hz, CDCl3): δ 8.074-8.046 (dd, J=9.2 and 2.4 Hz, 1H), 7.705-7.677 (dd, J=9.2 and 2.4 Hz, 1H), 7.571 (s, 1H) ppm.


Step 5: Preparation of 4,7,8-trichloroquinolin-2(1H)-one. The product of step 4 (4.93 g) was suspended in dioxane (80 mL), conc. H2SO4 (16 mL), water (24 mL), and heated to reflux for 12 to 16 hours. A clear solution was initially formed followed by the precipitation of solids. After starting material was consumed (confirmed by LC-MS) and the reaction mixture was cooled, 100 mL of ACN was added to form a precipitate. Filtration, rinsing with CAN, and drying under vacuum afforded the titled compound (4.12 g) as colorless powder. MS (ES): [M+1]+ 248.0.


Step 6: Preparation of 7,8-dichloro-4-(1H-imidazol-1-yl) quinolin-2(1H)-one (1-8). The product of step 5 (4.12 g) was suspended in DMF (20 mL) with imidazole (4 g) and heated to 130° C. until starting material was consumed (confirmed by LC-MS). Additional imidazole can be added to push the reaction to the completion. After cooling, ACN (150 mL) was added to form a precipitate which was collected by filtration and rinsing with CAN afforded the titled compound (1.8, 3.90 g). MS (ES): [M+1]+ 280.0. 1H NMR (400 Hz, DMSO-d6): δ 8.089 (s, 1H), 7.650 (s, 1H), 7.494-7.516 (d, J=8.8 Hz, 1H), 7.316-7.338 (d, J=8.8 Hz, 1H), 7.225 (s. 1H), 6.795 (s, 1H) ppm.


Step 7: 2,7,8-trichloro-4-(1H-imidazol-1-yl) quinoline (Intermediate 1). The product of step 6 (3.90 g) was suspended in ACN (100 mL) and POCl3 (8 mL) and heated to reflux until all solids were dissolved and starting material was consumed (confirmed by LC-MS). After concentrating under vacuum, the residue was cooled in an ice bath and carefully neutralized by 5% NaOH to pH 7 to precipitate Intermediate 1. Filtration, rinsing with water, and drying under vacuum afforded Intermediate 1 (3.8 g) as light tan solid. MS (ES): [M+1]+ 298. 1H NMR (400 Hz, DMSO-d6): δ 8.200 (s, 1H), 8.005 (s, 1H), 7.925-7.948 (d, J=9.2 Hz, 1H), 7.785-7.808 (d, J=9.2 Hz, 1H), 7.772 (s. 1H), 7.286 (s, 1H) ppm.


Preparation of 2,7,8-trichloro-4-(1H-pyrazol-4-yl)quinoline (Intermediate 2)



embedded image


Step 1: Preparation of 7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2(1H)-one. 4,7,8-trichloroquinolin-2(1H)-one (300 mg, 1.21 mmol), Na2CO3 (321 mg, 3.03 mmol), and Pd(PPh3)4 (140 mg, 0.121 mmol) were added to a round-bottom flask and diluted with dioxane (10 mL) and water (5 mL). The mixture was cooled in an ice-bath, and vacuumed and purged with N2 three times. A solution of boron ester 2.1 (703 mg, 3.62 mmol) in dioxane (3 mL) was prepared and degassed. Under N2, a portion of the boron ester 2.1 (1 mL, 1.0 eq) in dioxane was added to the reaction mixture via a syringe. The reaction mixture was heated at 110° C. for 2 h while LC-MS showed that around 50% of 1.7 was consumed. The rest of the boron ester 2.1 in dioxane was added under N2. After heating and stirring for an additional 3 h, compound 1.7 was consumed (confirmed by LC-MS). After cooling to room temperature, the reaction mixture was further diluted with water (20 mL). The precipitated solids were collected by centrifuge or filtration and washed with DCM (5 mL×3). After drying under the vacuum, the titled compound was afforded (2.2, 315 mg). MS: [M+1]+ 280.


Step 2: Preparation of 2,7,8-trichloro-4-(1H-pyrazol-4-yl)quinoline (Intermediate 2). The product of step 1 (208 mg, 0.743 mmol) was suspended in ACN (2.5 mL) and POCl3 (0.42 mL). The resultant mixture was heated at 80° C. for 3 h or until less than 10% of compound 2.2 remained (confirmed by LC-MS). After the concentrating under vacuum, the residue was cooled in an ice bath and carefully neutralized by 5% NaOH to pH 7 to precipitate Intermediate 2 as a yellow solid. The solids were collected by centrifuge or filtration and washed with water (15 mL×3). After drying in vacuo, Intermediate 2 was afforded (180 mg). MS: [M+1]+ 298. 1HNMR (400 MHz, DMSO-D6) δ: 8.8.25-8.30 (m, 3H), 7.84 (d, J=9.2 Hz, 1H) and 7.72 (s, 1H) ppm.


Preparation of 2,7-dichloro-4-(1H-imidazol-1-yl) quinoline (Intermediate 3)



embedded image


Step 1: Preparation of 4,7-dichloroquinoline 1-oxide. To a solution of 4,7-dichloroquinoline (20.0 g, 101.5 mmol) in DCM (400 mL) at room temperature was added 3-chloroperoxybenzoic acid (mCPBA) (28.8 g, 85% purity, 142.1 mmol) in portions and the resulting solution was stirred at 40° C. for 2 hrs. The solution was then washed with aqueous NaHCO3 (2×50 mL), aqueous Na2S2O3 (100 mL×2), dried over anhydrous Mg2SO4, filtered, concentrated under reduced pressure, and dried under high vacuum to yield the titled product (21 g) as a white solid. MS: [M+1]+ 214.1.


Step 2: Preparation of 2,4,7-trichloroquinoline. The product of step 1 (20.0 g, 1.0 eq) was dissolved in POCl3 (74 mL) at rt and the resulting solution was stirred at 110° C. for 2 hrs. The mixture was concentrated under reduced pressure and adjusted to pH 9 with a 10% NaOH solution. The precipitate was collected, washed with water, and dried to afford the titled compound (18 g, yield 83%) as an off-white solid. MS: [M+1]+ 234.0. 1H NMR (400 Hz, CDCl3): δ 8.148-8.126 (d, J=8.8 Hz, 1H), 8.033 (s, 1H), 7.622-7.596 (dd, J=1.4 Hz, 1H), 7.510 (s, 1H) ppm.


Step 3: Preparation of 4,7-dichloroquinolin-2(1H)-one. To a solution of the product of step 2 (5.0 g, 1.0 eq) in 1,4-diaoxane (125 mL) was added 15% H2SO4 (250 mL) at 25° C. The resulting mixture was stirred at 140° C. for 12 hrs. The precipitated solids were collected by filtration, washed with water, and dried to afford the titled compound (3.9 g) as a gray solid. MS: [M+1]+ 214.1. 1H NMR (400 Hz, CDCl3): δ 12.122 (s, 1H), 7.880-7.859 (d, J=8.4 Hz, 1H), 7.406 (s, 1H), 7.368-7.343 (dd, J=1.4 Hz, 1H), 6.865 (s, 1H) ppm.


Step 4: Preparation of 7-chloro-4-(1H-imidazol-1-yl)quinolin-2(1H)-one. To a solution of 4,7-dichloroquinolin-2(1H)-one (9.0 g, 1.0 eq) in DMF (18 mL) was added imidazole (48.8 g, 17 eq). After stirring at 120° C. for 21 hrs, the reaction mixture was diluted with ACN (100 mL) and stirred for 0.5 hrs. The precipitated solid was collected, washed with ACN, and dried in vacuo to afford the crude titled compound (6.7 g) as a gray solid. MS: [M+1]+ 246.1. 1H NMR (400 MHz, DMSO): δ 12.19 (s, 1H), 8.09 (s, 1H), 7.65 (s, 1H), 7.46 (d, J=2.0 Hz, 1H), 7.39 (d, J=8.8 Hz, 1H), 7.29 (dd, J=8.8, 2.0 Hz, 1H), 7.21 (s, 1H), 6.66 (s, 1H) ppm.


Step 5: Preparation of 2,7-dichloro-4-(1H-imidazol-1-yl) quinolone (Intermediate 3). To a solution of compound 3.5 (5.0 g, 1.0 eq) in ACN (50 mL) was added POCl3 (10 mL) and the resulting mixture was stirred at 80° C. for 0.5 hrs. The mixture was concentrated in vacuo and the residue was carefully quenched with 5-10% NaOH solution to pH 7 in an ice-bath. The precipitated solid was collected, washed with water, and dried in vacuo to yield Intermediate 3 (4.1 g) as a white solid. MS (ES): [M+1]+ 264. 1H NMR (400 Hz, CDCl3): δ 8.164 (s, 1H), 7.970 (s, 1H), 7.786-7.763 (d, J=9.2 Hz, 1H), 7.631-7.608 (d, J=9.2 Hz, 1H), 7.422 (s, 1H), 7.357 (s, 1H) ppm.


Preparation of 2,4-dichloro-7-(trifluoromethyl)quinoline (Intermediate 4A) and 2,4-dichloro-5-(trifluoromethyl)quinoline (Intermediate 4B).




embedded image


Step 1: 3-Oxo-3-((3-(trifluoromethyl) phenyl) amino) propanoic acid. A mixture of 3-(trifluoromethyl) aniline (5.74 g) and diethyl malonate (14.9 g) was stirred and heated at 180° C. over 4 hours. The resultant mixture was dissolved in MeOH (30 mL) and water (10 mL). The solution was cooled in an ice-bath and treated with NaOH (7.1 g). After stirring at room temperature over 2 hours and removal of MeOH under vacuum, the resultant mixture was further diluted with water (60 mL) and acidified to pH 1 to 2 with conc. HCl. After extraction with EtOAc (30 mL×4), the combined organic layers were washed by brine and dried over Na2SO4. Evaporation of EtOAc under reduced pressure afforded 4.3 (8.76 g) as oil. MS: [M+1]+ 276.


Step 2: 7-(Trifluoromethyl) quinoline-2,4(1H,3H)-dione 4-4 and 5-(trifluoromethyl) quinoline-2,4 (1H,3H)-dione. Neat 4.3 (8.76 g) was suspended in PPA (42 g) and heated at 130° C. over 4 hours. The resultant mixture was further diluted with water (120 mL) and the aqueous layer was extracted with EtOAc (40 mL×3). The combined organic layers were washed with brine and dried over Na2SO4. Evaporation under reduced pressure afforded a mixture of two titled regioisomers (5.99 g) as sticky solids. MS: [M+1]+ 230.


Step 3: 2,4-Dichloro-7-(trifluoromethyl)quinoline (Intermediate 4A) and 2,4-dichloro-5-(trifluoromethyl)quinoline (Intermediate 4B3). The mixture of 4.4 and 4.5 (5.99 g) from the previous step were suspended in POCl3, stirred, and heated at reflux over 3 h. POCl3 was removed under reduced pressure and the resultant mixtures were carefully quenched with ice. The aqueous layer was extracted with EtOAc (30 mL×3) and the combined organic layers were washed with brine and dried over Na2SO4. Column chromatography, eluting with a gradient of DCM/Hexane from 0 to 5000, afforded Intermediate 4A (3.1 g) and Intermediate 4B (0.59 g). 4A: MS [M+1]+: 266.1. 1HNMR (400 MHz, CD3Cl) δ: 8.38 (s, 1H), 8.37 (d, J=8 Hz, 1H), 7.86 (dd, 1=8 and 4 Hz) and 7.66 (s, 1H) ppm. 4B: MS [M+1]: 266.1. 1HNMR (400 MHz, CD3Cl) δ: 8.29 (d, J=8 Hz, 1H), 8.18 (d, J=8 Hz, 1H-), 7.85 (t, J=8 Hz) and 7.72 (s, 1H) ppm.


Following the above procedures to prepare Intermediates 1 and 4, the following intermediates were prepared.


















Starting
Inter-

MS:
Inter-

MS:


Material
mediate #
Structure
[M + 1]+
mediate #
Structure
[M + 1]+









embedded image


5A


embedded image


228
5B


embedded image


260







embedded image


6A


embedded image


240
6B


embedded image


272







embedded image


7A


embedded image


254
7B


embedded image


286







embedded image


8A


embedded image


212
8B


embedded image


244







embedded image


9A


embedded image


212
9B


embedded image


244







embedded image


10A


embedded image


250
10B


embedded image


282







embedded image


11A


embedded image


266
11B


embedded image


298







embedded image


12A


embedded image


266
12B


embedded image


298







embedded image


13A


embedded image


266
13B


embedded image


298







embedded image


14A


embedded image


266
14B


embedded image


298







embedded image


15A


embedded image


262
15B


embedded image


294







embedded image





16


embedded image


298







embedded image


17


embedded image


306










embedded image


18


embedded image


266












Example 1: Synthesis of 7-chloro-4-(1H-imidazol-1-yl)-N-(pyridin-4-ylmethyl)quinolin-2-amine (I-353)



embedded image


To a solution of 2,7-dichloro-4-(1H-imidazol-1-yl) quinoline (132 mg, 0.5 mmol) in DMF (2 mL) was added pyridin-4-ylmethanamine (0.15 mL, 1.5 mmol). The solution was vigorously stirred at 120° C. for 3 h. After cooling down to room temperature, the solvent was removed by evaporation to give a crude. The crude was purified directly by column chromatography on silica gel to give the title product as a solid (115 mg, 68% yield). MS: [M+1]+ 336.1.


The following compounds are prepared essentially by the same method described above to prepare I-353.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-354


embedded image




embedded image




embedded image


358





I-126


embedded image




embedded image




embedded image


371





I-131


embedded image




embedded image




embedded image


371





I-355


embedded image




embedded image




embedded image


321





I-447


embedded image




embedded image




embedded image


371





I-356


embedded image




embedded image




embedded image


371





I-357


embedded image




embedded image




embedded image


357





I-115


embedded image




embedded image




embedded image


427





I-121


embedded image




embedded image




embedded image


347





I-95 


embedded image




embedded image




embedded image


349





I-87 


embedded image




embedded image




embedded image


397





I-91 


embedded image




embedded image




embedded image


393





I-211


embedded image




embedded image




embedded image


406





I-212


embedded image




embedded image




embedded image


420





I-213


embedded image




embedded image




embedded image


392





I-214


embedded image




embedded image




embedded image


406





I-215


embedded image




embedded image




embedded image


392





I-216


embedded image




embedded image




embedded image


482





I-358


embedded image




embedded image




embedded image


434





I-218


embedded image




embedded image




embedded image


436





I-434


embedded image




embedded image




embedded image


407





I-433


embedded image




embedded image




embedded image


393





I-437


embedded image




embedded image




embedded image


449





I-438


embedded image




embedded image




embedded image


483





I-352


embedded image




embedded image




embedded image


363





I-432


embedded image




embedded image




embedded image


407





I-359


embedded image




embedded image




embedded image


363





I-431


embedded image




embedded image




embedded image


393





I-360


embedded image




embedded image




embedded image


469





I-361


embedded image




embedded image




embedded image


469





I-362


embedded image




embedded image




embedded image


469





I-444


embedded image




embedded image




embedded image


391





I-244


embedded image




embedded image




embedded image


351





I-92 


embedded image




embedded image




embedded image


393





I-241


embedded image




embedded image




embedded image


351





I-242


embedded image




embedded image




embedded image


351





I-93 


embedded image




embedded image




embedded image


393





I-94 


embedded image




embedded image




embedded image


407





I-85 


embedded image




embedded image




embedded image


328





I-138


embedded image




embedded image




embedded image


363





I-139


embedded image




embedded image




embedded image


363





I-240


embedded image




embedded image




embedded image


337





I-239


embedded image




embedded image




embedded image


323





I-249


embedded image




embedded image




embedded image


353





I-245


embedded image




embedded image




embedded image


349





I-141


embedded image




embedded image




embedded image


363





I-243


embedded image




embedded image




embedded image


351





I-86 


embedded image




embedded image




embedded image


376





I-246


embedded image




embedded image




embedded image


399





I-247


embedded image




embedded image




embedded image


399





I-155


embedded image




embedded image




embedded image


367





I-128


embedded image




embedded image




embedded image


391





I-152


embedded image




embedded image




embedded image


462





I-363


embedded image




embedded image




embedded image


371





I-140


embedded image




embedded image




embedded image


343





I-99 


embedded image




embedded image




embedded image


359





I-97 


embedded image




embedded image




embedded image


379





I-88 


embedded image




embedded image




embedded image


342





I-446


embedded image




embedded image




embedded image


357





I-364


embedded image




embedded image




embedded image


381





I-117


embedded image




embedded image




embedded image


391





I-365


embedded image




embedded image




embedded image


342





I-366


embedded image




embedded image




embedded image


371





I-367


embedded image




embedded image




embedded image


328





I-116


embedded image




embedded image




embedded image


426





I-89 


embedded image




embedded image




embedded image


375





I-158


embedded image




embedded image




embedded image


387





I-160


embedded image




embedded image




embedded image


401





I-200


embedded image




embedded image




embedded image


386





I-201


embedded image




embedded image




embedded image


400





I-202


embedded image




embedded image




embedded image


414





I-178


embedded image




embedded image




embedded image


336





I-179


embedded image




embedded image




embedded image


350





I-204


embedded image




embedded image




embedded image


400





I-159


embedded image




embedded image




embedded image


401





I-205


embedded image




embedded image




embedded image


414





I-148


embedded image




embedded image




embedded image


426





I-149


embedded image




embedded image




embedded image


426





I-150


embedded image




embedded image




embedded image


412





I-368


embedded image




embedded image




embedded image


440





I-206


embedded image




embedded image




embedded image


400





I-207


embedded image




embedded image




embedded image


400





I-210


embedded image




embedded image




embedded image


400





I-190


embedded image




embedded image




embedded image


350





I-191


embedded image




embedded image




embedded image


364





I-192


embedded image




embedded image




embedded image


378





I-428


embedded image




embedded image




embedded image


377





I-327


embedded image




embedded image




embedded image


393





I-369


embedded image




embedded image




embedded image


393





I-370


embedded image




embedded image




embedded image


417





I-328


embedded image




embedded image




embedded image


393





I-238


embedded image




embedded image




embedded image


337





I-145


embedded image




embedded image




embedded image


349





I-203


embedded image




embedded image




embedded image


386





I-144


embedded image




embedded image




embedded image


349





I-371


embedded image




embedded image




embedded image


392





I-372


embedded image




embedded image




embedded image


379





I-373


embedded image




embedded image




embedded image


426





I-250


embedded image




embedded image




embedded image


387





I-248


embedded image




embedded image




embedded image


351





I-374


embedded image




embedded image




embedded image


377





I-375


embedded image




embedded image




embedded image


391





I-157


embedded image




embedded image




embedded image


373





I-98 


embedded image




embedded image




embedded image


379





I-443


embedded image




embedded image




embedded image


391





I-142


embedded image




embedded image




embedded image


379





I-435


embedded image




embedded image




embedded image


377





I-143


embedded image




embedded image




embedded image


377





I-436


embedded image




embedded image




embedded image


391





I-448


embedded image




embedded image




embedded image


376





I-451


embedded image




embedded image




embedded image


404





I-348


embedded image




embedded image




embedded image


452





I-429


embedded image




embedded image




embedded image


393





I-453


embedded image




embedded image




embedded image


404





I-449


embedded image




embedded image




embedded image


392





I-450


embedded image




embedded image




embedded image


390





I-147


embedded image




embedded image




embedded image


377





I-345


embedded image




embedded image




embedded image


393





I-376


embedded image




embedded image




embedded image


377





I-430


embedded image




embedded image




embedded image


395





I-439


embedded image




embedded image




embedded image


459





I-322


embedded image




embedded image




embedded image


395





I-377


embedded image




embedded image




embedded image


285





I-269


embedded image




embedded image




embedded image


360





I-268


embedded image




embedded image




embedded image


374





I-267


embedded image




embedded image




embedded image


332





I-266


embedded image




embedded image




embedded image


332





I-264


embedded image




embedded image




embedded image


386





I-263


embedded image




embedded image




embedded image


386





I-262


embedded image




embedded image




embedded image


370





I-261


embedded image




embedded image




embedded image


386





I-260


embedded image




embedded image




embedded image


386





I-259


embedded image




embedded image




embedded image


353





I-132


embedded image




embedded image




embedded image


363





I-133


embedded image




embedded image




embedded image


363





I-134


embedded image




embedded image




embedded image


363





I-455


embedded image




embedded image




embedded image


393





I-456


embedded image




embedded image




embedded image


391





I-457


embedded image




embedded image




embedded image


453





I-458


embedded image




embedded image




embedded image


387





I-459


embedded image




embedded image




embedded image


407





I-460


embedded image




embedded image




embedded image


419





I-461


embedded image




embedded image




embedded image


377





I-462


embedded image




embedded image




embedded image


459





I-463


embedded image




embedded image




embedded image


421





I-464


embedded image




embedded image




embedded image


401





I-465


embedded image




embedded image




embedded image


401





I-466


embedded image




embedded image




embedded image


395





I-467


embedded image




embedded image




embedded image


399





I-468


embedded image




embedded image




embedded image


459





I-469


embedded image




embedded image




embedded image


389





I-470


embedded image




embedded image




embedded image


389





I-471


embedded image




embedded image




embedded image


431





I-472


embedded image




embedded image




embedded image


407





I-473


embedded image




embedded image




embedded image


391





I-474


embedded image




embedded image




embedded image


389





I-475


embedded image




embedded image




embedded image


403





I-476


embedded image




embedded image




embedded image


425





I-477


embedded image




embedded image




embedded image


391





I-478


embedded image




embedded image




embedded image


403





I-479


embedded image




embedded image




embedded image


405





I-480


embedded image




embedded image




embedded image


389





I-481


embedded image




embedded image




embedded image


391





I-482


embedded image




embedded image




embedded image


391





I-483


embedded image




embedded image




embedded image


405





I-484


embedded image




embedded image




embedded image


391





I-485


embedded image




embedded image




embedded image


395





I-486


embedded image




embedded image




embedded image


395





I-487


embedded image




embedded image




embedded image


453





I-488


embedded image




embedded image




embedded image


387





I-489


embedded image




embedded image




embedded image


389





I-490


embedded image




embedded image




embedded image


389





I-491


embedded image




embedded image




embedded image


401





I-492


embedded image




embedded image




embedded image


403





I-493


embedded image




embedded image




embedded image


417





I-494


embedded image




embedded image




embedded image


403





I-495


embedded image




embedded image




embedded image


358





I-496


embedded image




embedded image




embedded image


401





I-497


embedded image




embedded image




embedded image


407





I-498


embedded image




embedded image




embedded image


393





I-499


embedded image




embedded image




embedded image


393





I-500


embedded image




embedded image




embedded image


407





I-501


embedded image




embedded image




embedded image


365





I-502


embedded image




embedded image




embedded image


392





I-503


embedded image




embedded image




embedded image


388





I-504


embedded image




embedded image




embedded image


399





I-659


embedded image




embedded image




embedded image


349





I-644


embedded image




embedded image




embedded image


433





I-660


embedded image




embedded image




embedded image


439





I-835


embedded image




embedded image




embedded image


379





I-888


embedded image




embedded image




embedded image


427





I-881


embedded image




embedded image




embedded image


434





I-883


embedded image




embedded image




embedded image


427





I-872


embedded image




embedded image




embedded image


451









Example 2: Synthesis of 2-(1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)acetic acid (I-123)



embedded image


Step 1: Ethyl 2-(1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)acetate. To a solution of 2,7-dichloro-4-(1H-imidazol-1-yl)quinoline (38 mg, 0.144 mmol) in DMF (1 mL) was added ethyl 2-(piperidin-3-yl)acetate (50 mg, 0.3 mmol). The solution was vigorously stirred at 140° C. for 3 h. After cooling down to room temperature, the solvent was removed by evaporation and the residue was purified by column chromatography on silica gel to give the titled product as an oil (MS: [M+1]+ 399.1).


Step 2: 2-(1-(7-Chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)acetic acid. To a solution of ethyl 2-(1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)acetate (20 mg, 0.05 mmol) in MeOH (2 mL) was added 10% NaOH (aq., 0.5 mL). The mixture was stirred at 50° C. for 1 h. After cooling to room temperature, the crude was acidified by 1 N HCl (2.5 mL). The titled compound was collected as a solid by filtration (MS: [M+1]+ 371.1).


The following compounds were prepared essentially by the same method as described above for I-123.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-84


embedded image




embedded image




embedded image


372





I-122


embedded image




embedded image




embedded image


357.1





I-127


embedded image




embedded image




embedded image


357.1





I-445


embedded image




embedded image




embedded image


343.0





I-331


embedded image




embedded image




embedded image


357.1





I-135


embedded image




embedded image




embedded image


373.1





I-332


embedded image




embedded image




embedded image


373





I-334


embedded image




embedded image




embedded image


406





I-130


embedded image




embedded image




embedded image


373.1





I-136




embedded image


373.1





I-124


embedded image




embedded image




embedded image


407





I-333


embedded image




embedded image




embedded image


371





I-505


embedded image




embedded image




embedded image


389





I-506


embedded image




embedded image




embedded image


373





I-507


embedded image




embedded image




embedded image


391





I-508


embedded image




embedded image




embedded image


445





I-509


embedded image




embedded image




embedded image


576





I-510


embedded image




embedded image




embedded image


576





I-511


embedded image




embedded image




embedded image


576





I-512


embedded image




embedded image




embedded image


528





I-513


embedded image




embedded image




embedded image


389





I-514


embedded image




embedded image




embedded image


393





I-515


embedded image




embedded image




embedded image


417





I-516


embedded image




embedded image




embedded image


403





I-517


embedded image




embedded image




embedded image


417





I-518


embedded image




embedded image




embedded image


462





I-519


embedded image




embedded image




embedded image


392





I-520


embedded image




embedded image




embedded image


413





I-521


embedded image




embedded image




embedded image


393





I-844


embedded image




embedded image




embedded image


427





I-845


embedded image




embedded image




embedded image


441





I-831


embedded image




embedded image




embedded image


407





I-887


embedded image




embedded image




embedded image


413





I-854


embedded image




embedded image




embedded image


393





I-830


embedded image




embedded image




embedded image


393





I-882


embedded image




embedded image




embedded image


419









Example 3: Synthesis of (7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)glycine (I-166)



embedded image


Step 1: tert-Butyl (7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)glycinate. A mixture of 2,7-dichloro-4-(1H-imidazol-1-yl)quinoline (30 mg, 0.114 mmol), tert-butyl glycinate (38 mg, 0.228 mmol), triethylamine (0.2 mL) and dioxane (2 mL) was heated at 100° C. for 16 h. After cooling to room temperature, the crude was diluted with water (5 mL) and extracted by EtOAc (2×5 mL). The combined organics were dried and concentrated. The residue was purified by column chromatography on silica gel (25-100% EtOAc/Hexanes) to give tert-butyl (7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)glycinate as a solid (MS: [M+1]+ 359.1).


Step 2: (7-Chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)glycine. Tert-butyl (7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)glycinate (20 mg, 0.056 mmol) was placed in a vial with dichloromethane (3 mL). Hydrochloric acid (2.0 M in diethyl ether) was added and the reaction was stirred at r.t. for 16 h. The volatiles were concentrated off and the resulting solid was triturated with dichloromethane, then dried in vacuo to afford (7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)glycine as a solid (MS: [M+1]+ 303.1).


The following compounds were prepared essentially by the same method as described above to prepare I-166:


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-182


embedded image




embedded image




embedded image


317.1





I-167


embedded image




embedded image




embedded image


317.1









Example 4: Synthesis of 5-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-hydroxynicotinic acid (I-57)



embedded image


5-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-methoxynicotinic acid (I-33, 15 mg, 0.04 mmol) was placed in a vial with acetic acid (1 mL). Hydrobromic acid (33% in acetic acid, 0.2 mL) was added and the reaction was stirred at r.t. for 16 h. The volatiles were concentrated off and the resulting residue was purified on a silica prep plate to afford the title compound (MS: [M+1]+ 367.0).


Example 5: Synthesis of (7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)serine (I-168)



embedded image


Step 1: Methyl (7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)serinate. To a solution of 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (30 mg, 0.1 mmol) in DMSO (0.2 mL) was added DL-Serine methyl ester HCl (39 mg, 0.25 mmol) and N,N-diisopropylethylamine (0.1 mL) and the solution was stirred at 95° C. for 16 h. After cooling to room temperature, water (5 mL) was added and the organics were extracted into 10% methanol in dichloromethane (2×5 mL). The combined organics were dried (Na2SO4) and concentrated. The resulting residue was purified by silica chromatography using 0-15% MeOH/dichloromethane to afford methyl (7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)serinate (MS: [M+1]+ 381.0).


Step 2: (7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)serine. Methyl (7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)serinate (12 mg, 0.031 mmol) was placed in a vial with THF (0.6 mL), methanol (0.2 mL), and water (0.2 mL). Lithium hydroxide monohydrate (4 mg, 0.09 mmol) was added and the reaction was allowed to stir at r.t. for 16 h. The volatiles were concentrated off and the resulting residue neutralized by the addition of 1 N hydrochloric acid. The resulting solution was lyophilized to afford the titled compound (MS: [M+1]+ 367.0).


The following compounds were prepared essentially by the same method described above to prepare I-168:


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-169


embedded image




embedded image




embedded image


443.0





I-185


embedded image




embedded image




embedded image


365.0





I-186


embedded image




embedded image




embedded image


365.0





I-189


embedded image




embedded image




embedded image


427.0





I-187


embedded image




embedded image




embedded image


393.0





I-184


embedded image




embedded image




embedded image


365.0





I-183


embedded image




embedded image




embedded image


365.1





I-188


embedded image




embedded image




embedded image


393.0









Example 6: Synthesis of N-(7,8-dichloro-4-(1H-imidazol-1-yl)quinoline-2-yl)-N-(2-morpholinoeythyl)glicine (I-172)



embedded image


Step 1: tert-Butyl (2-morpholinoethyl)glycinate. To a solution of 2-morpholinoethan-1-amine (260 mg, 2.0 mmol) in acetonitrile (5 mL) was added K2CO3 (1.38 g, 10 mmol). The reaction was cooled to 0° C. and tert-butyl 2-bromoacetate (390 mg, 2.0 mmol) in acetonitrile (5 mL) was added dropwise. The reaction was stirred at 0° C. for 1 h then filtered through Celite and concentrated. The residue was purified by column chromatography to afford tert-butyl (2-morpholinoethyl)glycinate as an oil (200 mg) (MS: [M+1]+ 245.2).


Step 2: tert-Butyl N-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-N-(2-morpholinoethyl)glycinate. To a solution of 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (30 mg, 0.1 mmol) in DMSO (0.2 mL) was added tert-butyl (2-morpholinoethyl)glycinate (200 mg) and N,N-diisopropylethylamine (0.1 mL). The solution was stirred at 95° C. for 16 h. After cooling down to room temperature, water (5 mL) was added and the organics were extracted into 10% methanol in dichloromethane (2×5 mL). The combined organics were dried (Na2SO4) and concentrated. The resulting residue was purified by silica chromatography using 0-15% MeOH/dichloromethane to afford tert-butyl N-(7,8-dichloro-4-(1H-imidazol-1-yl)294uinoline-2-yl)-N-(2-morpholinoethyl)glycinate (10 mg) (MS: [M+1]+ 506.1).


Step 3: N-(7,8-dichloro-4-(1H-imidazol-1-yl)quinoline-2-yl)-N-(2-morpholinoethyl)glycine. tert-Butyl N-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-N-(2-morpholinoethyl)glycinate (10 mg, 0.02 mmol) was placed in a vial with dichloromethane (1 mL). Trifluoroacetic acid (0.1 mL) was added and the reaction stirred at r.t. for 16 h. The volatiles were removed by rotary evaporation and the residue was dried under vacuum to afford N-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-N-(2-morpholinoethyl)glycine (MS: [M+1]+ 450.1).


The following compounds were prepared essentially by the same methods described above to prepare I-172:


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-173


embedded image




embedded image




embedded image


395.0





I-174


embedded image




embedded image




embedded image


395.1





I-175


embedded image




embedded image




embedded image


409.1





I-176


embedded image




embedded image




embedded image


428.0









Example 7: Synthesis of 2-((7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)-4-(methylsulfonyl)butanoic acid (I-170)



embedded image


Step 1: Ethyl (7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)methioninate. To a solution of 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (30 mg, 0.1 mmol) in DMSO (0.2 mL) was added ethyl methioninate (47 mg, 0.2 mmol) and N,N-diisopropylethylamine (0.1 mL). The solution was stirred at 95° C. for 16 h. After cooling down to room temperature, water (5 mL) was added and the organics were extracted into 10% methanol in dichloromethane (2×5 mL). The combined organics were dried (Na2SO4) and concentrated. The resulting residue was purified by silica chromatography using 0-100% EtOAc/hexanes to afford ethyl (7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)methioninate (20 mg) (MS: [M+1]+ 439.0).


Step 2: Ethyl 2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)-4-(methylsulfonyl)butanoate. Ethyl (7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)methioninate (20 mg, 0.045 mmol) was placed in a vial with dichloromethane (2 mL). m-CPBA (20 mg, 0.09 mmol) was added and the reaction was allowed to stir at r.t. for 16 h. Sodium sulfite (2 mL, 10% aq.) was added and the organics were dried (Na2SO4) and purified by silica chromatography using 0-10% MeOH/CH2Cl2 to afford ethyl 2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)-4-(methylsulfonyl)butanoate (10 mg) (MS: [M+1]+ 471.1).


Step 3: 2-((7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)-4-(methylsulfonyl)butanoic acid. Ethyl 2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)-4-(methylsulfonyl)butanoate (20 mg, 0.042 mmol) was placed in a vial with THF (1 mL) and water (0.25 mL). Lithium hydroxide monohydrate (5 mg, 0.13 mmol) was added and the reaction mixture was stirred at r.t. for 3 h. The volatiles were removed by rotary evaporation and the residue was neutralized by the addition of 1N HCl (aq.). The resulting solution was lyophilized to afford 2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)-4-(methylsulfonyl)butanoic acid (12 mg) (MS: [M+1]+ 443.0).


Example 8: Synthesis of 7-chloro-4-(1H-imidazol-1-yl)-2-(4-methoxyphenyl)quinoline (I-2)



embedded image


A mixture of 2,7-dichloro-4-(1H-imidazol-1-yl)quinoline (132 mg, 0.5 mmol), (4-methoxyphenyl)boronic acid (114 mg, 0.75 mmol), Na2CO3 (265 mg, 2.5 mmol), 1,4-dioxane (4 mL) and H2O (1 mL) was purged by N2 for 30 min. Tetrakis(triphenylphosphine)palladium (55 mg, 0.05 mmol) was added to the mixture above under N2. The mixture was vigorously stirred at 100° C. for 3 h. After cooling down to room temperature, the crude was diluted by EtOAc (20 mL) and washed by water (5 mL×2) and brine (5 mL×2). The organic phase was concentrated and purified by column chromatography on silica gel to give the titled product I-2 as a solid (MS: [M+1]+ 336.1).


The following compounds were prepared essentially by the same methods described above to prepare I-2.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-5


embedded image




embedded image




embedded image


364.1





I-12


embedded image




embedded image




embedded image


378.1





I-3


embedded image




embedded image




embedded image


364.1





I-35


embedded image




embedded image




embedded image


347





I-36


embedded image




embedded image




embedded image


363





I-25


embedded image




embedded image




embedded image


406.1





I-31


embedded image




embedded image




embedded image


384.0





I-18


embedded image




embedded image




embedded image


434





I-45


embedded image




embedded image




embedded image


414





I-40


embedded image




embedded image




embedded image


395





I-46


embedded image




embedded image




embedded image


419





I-41


embedded image




embedded image




embedded image


386





I-43


embedded image




embedded image




embedded image


356





I-44


embedded image




embedded image




embedded image


440





I-47


embedded image




embedded image




embedded image


356





I-20


embedded image




embedded image




embedded image


365





I-28


embedded image




embedded image




embedded image


394





I-378


embedded image




embedded image




embedded image


330.0









Example 9: Synthesis of 7-chloro-4-(1H-imidazol-1-yl)-2-(4-methoxyphenyl)quinoline (I-6)



embedded image


Step 1: Methyl 3-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)benzoate. A mixture of 2,7-dichloro-4-(1H-imidazol-1-yl)quinoline (132 mg, 0.5 mmol), (3-(methoxycarbonyl)phenyl)boronic acid (136 mg, 0.75 mmol), Na2CO3 (265 mg, 2.5 mmol), 1,4-dioxane (4 mL) and H2O (1 mL) was purged by N2 for 30 min. Tetrakis(triphenylphosphine) palladium (55 mg, 0.05 mmol) was added to the mixture above under N2. The mixture was vigorously stirred at 100° C. for 2 h. After cooling down to room temperature, the crude was diluted by EtOAc (20 mL) and washed by water (5 mL×2) and brine (5 mL×2). The organic phase was concentrated and purified by column chromatography on silica gel to give the titled product as a solid (MS: [M+1]+ 364.1).


Step 2: 3-(7-Chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)benzoic acid. To a solution of methyl 3-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)benzoate (80 mg, 0.22 mmol) in MeOH (4 mL) was added 10% NaOH (aq., 1 mL). The mixture was stirred at 50° C. for 1 h. After cooling down to room temperature, the crude was acidified by 1 N HCl (5 mL). The titled compound was collected as a solid by filtration (MS: [M+1]+ 350.1).


The following compounds are prepared essentially by the same method described above to prepare I-6:


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-13


embedded image




embedded image




embedded image


350.1





I-4


embedded image




embedded image




embedded image


350.1





I-15


embedded image




embedded image




embedded image


380





I-30


embedded image




embedded image




embedded image


380





I-55


embedded image




embedded image




embedded image


356





I-37


embedded image




embedded image




embedded image


394.0





I-38




embedded image


430.0





I-27


embedded image




embedded image




embedded image


428.0





I-17


embedded image




embedded image




embedded image


368.0





I-14


embedded image




embedded image




embedded image


364.1





I-9


embedded image




embedded image




embedded image


364.1





I-32


embedded image




embedded image




embedded image


351.0





I-33


embedded image




embedded image




embedded image


381.1









Example 10: Synthesis of 3-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)benzamide (I-7)



embedded image


To a solution of 3-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)benzoic acid (25 mg, 0.07 mmol) in THF (1 mL) was added SOCl2 (0.015 mL, 0.2 mmol). After 1 h, NH4OH (aq., 0.5 mL) was added. After 3 h, the solvent was removed by evaporation to give a crude. The crude was purified directly by column chromatography on silica gel to give the titled product as a solid (MS: [M+1]+ 349.1).


Example 11: Synthesis of 3-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-methoxybenzoic acid (I-24) and 3-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-hydroxybenzoic acid (I-26)



embedded image


Step 1: 3-(4,7-dichloroquinolin-2-yl)-5-methoxybenzoic acid was prepared essentially by the same methods described above to prepare I-2.


Step 2: 3-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-methoxybenzoic acid. To a vial were added 3-(4,7-dichloroquinolin-2-yl)-5-methoxybenzoic acid (50 mg, 0.144 mmol), imidazole (49 mg, 0.718 mmol), Cs2CO3 (70 mg, 0.216 mmol) and DMF (1.0 mL). The resulting reaction mixture was stirred at 110° C. overnight. At room temperature the reaction mixture was diluted by H2O (4 mL) and acidified by HCl (1 N) to pH about 3. A lot of white solid precipitated, centrifuged and washed by H2O (2×4 mL). The residue was dried in vacuo to afford the titled compound as white solid (30 mg). MS: [M+1]+ 380.


Step 3: 3-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-hydroxybenzoic acid. To a vial were added 3-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-methoxybenzoic acid (7.9 mg, 0.0208 mmol), NaI (20 mg, 0.133 mmol) and HBr in HOAc (33%, 1.0 mL). The resulting mixture was stirred at 50° C. overnight. The mixture was cooled to room temperature and diluted by H2O (10 mL), centrifuged, and washed with H2O (4 mL). The residue was dried under high vacuum to afford the title product as off-white solid (18 mg) (MS: [M+1]+ 366).


Example 12: Synthesis of 5-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-hydroxybenzoic acid (I-16) and 5-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-ethoxybenzoic acid (I-19)



embedded image


Step 1: Methyl 5-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-methoxybenzoate was prepared according to the procedure above to prepare compound I-2.


Step 2: 5-(7-Chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-hydroxybenzoic acid (I-16) was prepared according to the procedure above to prepare compound I-26. (MS: [M+1]+ 366).


Step 3: Ethyl 5-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-ethoxybenzoate. To a vial were added 5-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-hydroxybenzoic acid (14 mg, 0.0383 mmol), ethyl iodide (22 μl, 0.274 mmol), Cs2CO3 (62 mg, 0.191 mmol) and DMF (1.0 mL). The resulting reaction mixture was stirred at room temperature overnight. The mixture was diluted with ethyl acetate (40 mL), washed by H2O (4×20 mL) and brine (15 mL). After concentration, the crude was used in next step.


Step 4: 5-(7-Chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-ethoxybenzoic acid. To the crude product above were added MeOH (0.6 mL), THF (0.4 mL) and a solution of NaOH in H2O (7.66 mg/0.2 mL). The resulting reaction mixture was stirred at room temperature overnight. Then the mixture was diluted by H2O (3 mL) and acidified by HOAc to pH 4. The cloudy mixture was centrifuged and the residue was washed by H2O (2×1.5 mL), dried over high vacuum to afford the title product as off-white solid (6 mg) (MS: [M+1]+ 394).


Example 13: Synthesis of 3-amino-5-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)benzoic acid (I-22)



embedded image


Step 1: methyl 3-amino-5-(4,7-dichloroquinolin-2-yl)benzoate was prepared according to the procedure above to prepare compound I-2.


Step 2: 3-amino-5-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)benzoic acid was prepared according to the procedure above to prepare compound I-24 (MS: [M+1]+ 365).


Example 14: Synthesis of 4-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-1H-pyrrole-2-carboxylic acid (I-56)



embedded image


Step 1: 1-(tert-Butyl) 2-methyl 4-(4,7-dichloroquinolin-2-yl)-1H-pyrrole-1,2-dicarboxylate was prepared according to the procedure described above to prepare compound I-2.


Step 2: Methyl 4-(4,7-dichloroquinolin-2-yl)-1H-pyrrole-2-carboxylate was prepared according to the procedure described above to prepare compound I-172 (step 3).


Step 3: 4-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-1H-pyrrole-2-carboxylic acid was prepared according to the procedure described above to prepare compound I-24 (step 2). (MS: [M+1]+339).


Example 15: Synthesis of 3-(7-Chloro-4-(1H-pyrazol-5-yl)quinolin-2-yl)benzoic acid (I-74)



embedded image


Step 1: Methyl 3-(4,7-dichloroquinolin-2-yl)benzoate was prepared according to the procedure described above to prepare compound I-2.


Step 2: Methyl 3-(7-chloro-4-(1H-pyrazol-5-yl)quinolin-2-yl)benzoate was prepared according to the procedure described above to prepare compound I-2.


Step 3: 3-(7-Chloro-4-(1H-pyrazol-5-yl)quinolin-2-yl)benzoic acid was prepared according to the procedure described above to prepare compound I-24 (step 2). (MS: [M+1]+ 350).


Example 16: Synthesis of 3-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-(2-hydroxyacetamido)benzoic acid (I-23)



embedded image


Step 1: 3-(2-Acetoxyacetamido)-5-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)benzoic acid. To a vial were added 3-amino-5-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)benzoic acid (9.3 mg, 0.0255 mmol), DCM (0.5 mL) and TEA (8.9 μl, 0.0637 mmol). A solution of 2-chloro-2-oxoethyl acetate in DCM (4.18 mg/0.1 mL, 0.0306 mmol) was added and the resulting reaction mixture was stirred at room temperature for 1 h. H2O (0.5 mL) was added to quench the reaction. Most of DCM was removed under reduced pressure and the crude was used in next step.


Step 2: 3-(7-Chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-(2-hydroxyacetamido)benzoic acid. To the crude product above (3-(2-acetoxyacetamido)-5-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)benzoic acid) were added MeOH (0.5 mL), THF (1.0 mL) and a solution of LiOH—H2O in H2O (5.36 mg/0.2 mL, 0.127 mmol). The resulting reaction mixture was stirred at room temperature for 16 h. All volatiles were removed and the mixture was diluted by H2O (2.5 mL). After centrifugation, the residue was washed by H2O (2×3 mL), dried under high vacuum to afford the title product as white solid (MS: [M+1]+ 423).


Example 17: Synthesis of 5-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-(2-hydroxyethoxy)benzoic acid (I-21)



embedded image


Step 1: Methyl 5-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-hydroxybenzoate was prepared according to the procedure described above to prepare compound I-2.


Step 2: Methyl 2-(2-((tert-butyldimethylsilyl)oxy)ethoxy)-5-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)benzoate. To a vial were added methyl 5-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-hydroxybenzoate (10 mg, 0.0263 mmol), Cs2CO3 (10.3 mg, 0.0316 mmol) and a solution of (2-bromoethoxy)(tert-butyl)dimethylsilane in DMF (6.8 μl/0.5 mL, 0.0316 mmol). The resulting reaction mixture was stirred at room temperature for 1 h. Then the mixture was diluted by ethyl acetate (30 mL), washed by H2O (4×10 mL) and brine (10 mL), dried over Na2SO4. After concentration, the crude was used in next step.


Step 3: 5-(7-Chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-(2-hydroxyethoxy)benzoic acid. To the crude step 2 were added MeOH (0.5 mL), THF (1.0 mL) and a solution of LiOH—H2O in H2O (5.5 mg/0.5 mL, 0.132 mmol). The resulting reaction mixture was stirred at room temperature for 48 h. The mixture was acidized by acetic acid to pH 5. Most of volatiles were removed and the residue was diluted with H2O (1.5 mL), centrifuged, washed by H2O (2×1.5 mL), dried under high vacuum to afford the title product as off-white solid (MS: [M+1]+ 410).


The following compounds are prepared essentially by the same method described above to prepare I-21.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-29


embedded image




embedded image




embedded image


410









Example 18: Synthesis of 5-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-hydroxybenzoic acid (I-42)



embedded image


Step 1: Methyl 2-methoxy-5-(4,7,8-trichloroquinolin-2-yl)benzoate. To a suspension of 2,4,7,8-tetrachloroquinoline (200 mg, 0.75 mmol) with (4-methoxy-3-(methoxycarbonyl)phenyl) boronic acid (205.3 mg, 0.98 mmol) and Na2CO3 (178 mg, 1.68 mmol) in dioxane (8.0 mL) and water (2.0 mL) was added Pd(PPh3)4 (79.2 mg). The resultant mixture was vacuumed and purged with N2 for three cycles, then stirred and heated at 80° C. over two hours. After cooling to room temperature, the reaction mixture was dissolved in DCM (50 mL) and washed with water and brine. The resultant organic layer was separated and dried over anhydrous Na2SO4. A silica gel flash column chromatography eluting with DCM/Hexane afforded the desired colorless product (MS: [M+1]+ 396).


Step 2: 2-Hydroxy-5-(4,7,8-trichloroquinolin-2-yl)benzoic acid (I-75). Methyl 2-methoxy-5-(4,7,8-trichloroquinolin-2-yl) benzoate (17 mg) in DCM (1.5 mL) was treated with 1M BBr3 in DCM (0.1 mL) at room temperature over 8 hours. The resulting mixture was diluted with EtOAc (25 mL), washed with water (10 mL) and dried over Na2SO4. Concentration under vacuum afforded the desired light brown solid (11 mg), 2-hydroxy-5-(4,7,8-trichloroquinolin-2-yl) benzoic acid (MS: [M+1]+ 368).


Step 3: 5-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-hydroxybenzoic acid. To a solution of 2-hydroxy-5-(4,7,8-trichloroquinolin-2-yl) benzoic acid (11 mg) in DMF (0.5 mL) was added imidazole (55 mg) and K2CO3 (50 mg). The resultant solution was stirred and heated at 120° C. over 5 hours until the starting material was completely consumed. The reaction mixture was diluted with water (3 mL) and treated with Dowex resin until the pH to 3. The resultant colorless solid was isolated by filtration and washed with water (3 mL). After drying under vacuum, the desired product was obtained (MS: [M+1]+ 400).


Example 19: Synthesis of 3-(4-(1H-imidazol-1-yl)-7-(trifluoromethyl)quinolin-2-yl)benzoic acid (I-49)



embedded image


Step 1: Methyl 3-(4-chloro-7-(trifluoromethyl)quinolin-2-yl)benzoate. To a mixture of 2,4-dichloro-7-(trifluoromethyl)quinoline (217 mg, 0.82 mmol) with (3-(methoxycarbonyl)phenyl)boronic acid (190.9 mg, 1.06 mmol) in dioxane (7 mL) and water (3 mL) were added Na2CO3 (191 mg, 1.804 mmol) and Pd(PPh3)4 (95 mg, 0.082 mmol). The resultant mixture was placed under vacuum and purged with nitrogen repeatedly three times, then was stirred and heated at 90° C. over 3 hours. The reaction mixture was diluted with water (25 mL) and extracted with EtOAc (20 mL). After isolation of the organic layer and a column chromatography eluting with a gradient of DCM/Hexane from 0 to 60%), the desired product (254 mg) methyl 3-(4-chloro-7-(trifluoromethyl)quinolin-2-yl)benzoate (MS: [M+1]+366).


Step 2: 3-(4-(1H-Imidazol-1-yl)-7-(trifluoromethyl)quinolin-2-yl)benzoic acid. A mixture of methyl 3-(4-chloro-7-(trifluoromethyl)quinolin-2-yl)benzoate (42 mg, 0.12 mmol), imidazole (40 mg) and Cs2CO3 was added DMF (0.6 mL). The suspended solution was stirred and heated at 100° C. over 6 hours. The reaction mixture was diluted with water (2 mL) and acidified with HOAc to pH 3 to precipitate the product. The product was isolated by centrifuge and rinsed with water and 50% acetonitrile/water. The wet solid was dried in vacuo to afford the title compound (30 mg) ([M+1]+ : 384).


Example 20: Synthesis of 3-(7-Bromo-4-(1H-imidazol-1-yl)-8-methoxyquinolin-2-yl)benzoic acid (I-48)



embedded image


Following step 1 in the preparation of I-49, tert-butyl 3-(7-bromo-4-chloro-8-methoxyquinolin-2-yl) benzoate was prepared from Intermediate 17.


Step 1: tert-Butyl 3-(7-bromo-4-(1H-imidazol-1-yl)-8-methoxyquinolin-2-yl)benzoate. To a mixture of tert-butyl 3-(7-bromo-4-chloro-8-methoxyquinolin-2-yl) benzoate (125 mg) and Cs2CO3 (136.8 mg) in DMF (2 mL) was added imidazole (96 mg). The suspended solution was stirred and heated at 130° C. over 2 h. Aqueous work-up with EtOAc and a column chromatography eluting with EtOAc/Hexane afforded the desired product tert-butyl 3-(7-bromo-4-(1H-imidazol-1-yl)-8-methoxyquinolin-2-yl) benzoate (120 mg) (MS: [M+1]+ 480).


Step 2: 3-(7-Bromo-4-(1H-imidazol-1-yl)-8-methoxyquinolin-2-yl)benzoic acid. To a solution of tert-butyl 3-(7-bromo-4-(1H-imidazol-1-yl)-8-methoxyquinolin-2-yl)benzoate (65 mg) in DCM (0.2 mL) and MeOH (0.2 mL) was added TFA (0.4 mL). The resultant solution was stirred over 5 h and concentrated to dryness. The resultant oily residue was suspended in water (0.5 mL) and lyophilized to afford the title compound 3-(7-bromo-4-(1H-imidazol-1-yl)-8-methoxyquinolin-2-yl) benzoic acid (60 mg) as light brown powder (MS: [M+1]+ 424).


Example 21: Synthesis of 7-bromo-4-(1H-imidazol-1-yl)-8-methoxy-2-phenylquinoline (I-54)



embedded image


7-Bromo-4-chloro-8-methoxy-2-phenylquinoline was prepared following step 1 in the preparation procedure of methyl 3-(4-chloro-7-(trifluoromethyl)quinolin-2-yl)benzoate.


To a solution of 7-bromo-4-chloro-8-methoxy-2-phenylquinoline (63 mg) in DMF (2.0 mL) were added imidazole (129 mg) and Cs2CO3 (62 mg). The resultant mixture was heated at 80° C. overnight. The reaction mixture was diluted with water (4 mL) to precipitate the desired product. Isolation of the product and rinsing with water (2 mL) afforded the title compound (55 mg) 7-bromo-4-(1H-imidazol-1-yl)-8-methoxy-2-phenylquinoline (MS: [M+1]+ 380).


Example 22: Synthesis of 7-bromo-4-(1H-imidazol-1-yl)-2-phenylquinolin-8-ol (I-53)



embedded image


To a solution of 7-bromo-4-chloro-8-methoxy-2-phenylquinoline (20 mg) in DMF (0.5 mL) were added imidazole (50 mg) and Cs2CO3 (50 mg). The resultant mixture was stirred at 120° C. overnight. The reaction mixture was diluted with water (3 mL) to precipitate the desired product. Isolation of the product and rinsing with water (1 mL) afforded the title compound (MS: [M+1]+ 366).


Example 23: Synthesis of 2-((7-bromo-4-(1H-imidazol-1-yl)-2-phenylquinolin-8-yl)oxy)acetic acid (I-379)



embedded image


Step 1: tert-Butyl 2-((7-bromo-4-(1H-imidazol-1-yl)-2-phenylquinolin-8-yl)oxy)acetate. To a solution of 7-bromo-4-(1H-imidazol-1-yl)-2-phenylquinolin-8-ol (10 mg) in DMSO (2 mL) were added tert-butyl 2-bromoacetate (40 mg) and K2CO3 (40 mg). The resultant solution was stirred at room temperature for 3 hrs and diluted with water/EtOAc (20 mL). The organic layer was separated, washed with brine, and dried over Na2SO4, then a column chromatography eluting with hexane/EtOAc afforded the desired product (9 mg). MS:[M+1]+ 480.


Step 2: 2-((7-Bromo-4-(1H-imidazol-1-yl)-2-phenylquinolin-8-yl)oxy)acetic acid. To a solution of tert-butyl 2-((7-bromo-4-(1H-imidazol-1-yl)-2-phenylquinolin-8-yl)oxy)acetate (9 mg) in DCM (0.8 mL) was added TFA (0.2 mL). The resultant solution was stirred overnight and concentrated to dryness. Lyophilization afforded the desired product (6.6 mg) (MS: [M+1]+ 424).


Example 24: Synthesis of 3-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)aniline (I-8)



embedded image


2,7-Dichloro-4-(1H-imidazol-1-yl)quinoline (26 mg, 0.10 mmol) was placed in a vial with dioxane 2.3 mL) and water (0.5 mL) under N2. (3-Aminophenyl)boronic acid (21 mg, 0.15 mmol) and potassium carbonate (70 mg, 0.50 mmol) were added followed by tetrakis(triphenylphosphine)palladium(0) (6 mg, 0.005 mmol). The mixture was stirred at 100° C. for 16 h. Water (5 mL) was added and the organics were extracted into 10% methanol in dichloromethane (2×5 mL). The combined organics were dried (Na2SO4) and concentrated. Purification by silica chromatography afforded 3-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)aniline (MS: [M+1]+ 321.1).


The following compounds are prepared essentially by the same method described above to prepare I-8.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-34


embedded image




embedded image




embedded image


365.0





I-58


embedded image




embedded image




embedded image


323.1





I-59


embedded image




embedded image




embedded image


323.0









Example 25: Synthesis of N-(3-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)phenyl)methanesulfonamide (I-10)



embedded image


3-(7-Chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)aniline (12 mg, 0.04 mmol) was placed in a vial with dichloromethane (2 mL) and TEA (0.1 mL) and cooled to 0° C. Methanesulfonyl chloride (6 mg, 0.06 mmol) was added and the solution was stirred at rt for 16 h. Water (3 mL) was added and the aqueous extracted with ethyl acetate (2×5 mL). The combined organics were dried and concentrated and the residue purified by silica chromatography to afford N-(3-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)phenyl)methanesulfonamide (MS: [M+1]+ 399.0).


Example 26: Synthesis of 1-(3-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)phenyl)urea (I-11)



embedded image


3-(7-Chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)aniline (10 mg, 0.03 mmol) was placed in a vial with acetic acid (0.2 mL). Potassium cyanate (25 mg, 0.31 mmol) in water was then added dropwise to the vial and the solution was stirred at rt for 1.5 h. Water (3 mL) was added and the aqueous extracted with 10% methanol/dichloromethane (2×5 mL). The combined organics were dried and concentrated and the residue was purified by silica chromatography to afford 1-(3-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)phenyl)urea (MS: [M+1]+ 364.1).


Example 27: Synthesis of 7-bromo-4-(1H-imidazol-1-yl)-2-(thiophen-2-yl)quinoline (I-380)



embedded image


Step 1: N-(2-Acetyl-5-bromophenyl)thiophene-2-carboxamide. 1-(2-Amino-4-bromophenyl)ethan-1-one (214 mg, 1.00 mmol) was placed in a flask with dichloromethane (5 mL) and triethylamine (0.15 mL, 1.10 mmol) then cooled to 0° C. Thiophene-2-carbonyl chloride (147 mg, 1.10 mmol) was added dropwise. The reaction was warmed to r.t. and stirred for 16 h. The volatiles were removed and the resulting solids were triturated with dichloromethane, filtered and vacuum dried to afford N-(2-acetyl-5-bromophenyl)thiophene-2-carboxamide as a solid (MS: [M+1]+ 323.9).


Step 2: 7-Bromo-2-(thiophen-2-yl)quinolin-4-ol. N-(2-acetyl-5-bromophenyl)thiophene-2-carboxamide (250 mg, 0.77 mmol) was placed in a flask with dioxane (10 mL). Sodium hydroxide (108 mg, 2.7 mmol) was added and the mixture heated to 110° C. for 2 h. Ethanol (2 mL) was added and the resulting solids filtered off. The filtrate was concentrated to dryness. Water (4 mL) and hexanes (1 mL) were added and the mixture was stirred for 5 minutes. The solution was acidified with HCl (1.0 N aq.) and the resulting solids filtered and vacuum dried to afford 7-bromo-2-(thiophen-2-yl)quinolin-4-ol as a solid (MS: [M+1]+ 306.0).


Step 3: 7-Bromo-4-chloro-2-(thiophen-2-yl)quinoline. 7-Bromo-2-(thiophen-2-yl)quinolin-4-ol (180 mg, 0.59 mmol) was placed in a flask with phosphorus oxychloride (3 mL). The reaction was heated to 110° C. for 3 h. After cooling to r.t., ice was added. The aqueous portion was extracted with ethyl acetate (2×5 mL) and the combined organics dried (Na2SO4) then concentrated to afford 7-bromo-4-chloro-2-(thiophen-2-yl)quinoline (MS: [M+1]+ 323.9).


Step 4: 7-bromo-4-(1H-imidazol-1-yl)-2-(thiophen-2-yl)quinoline. 7-Bromo-4-chloro-2-(thiophen-2-yl)quinoline (65 mg, 0.2 mmol) was placed in a flask with imidazole (34 mg, 0.50 mmol), potassium t-butoxide (34 mg, 0.30 mmol), Bis(triphenylphosphine)palladium(II) dichloride (7 mg, 0.01 mmol) and DMA (3 mL) under N2. The mixture was heated at 110° C. for 2 h. After cooling down to room temperature, the crude was diluted by EtOAc (20 mL) and washed by water (5 mL×2) and brine (5 mL×2). The organic phase was concentrated and purified by column chromatography on silica gel to give 7-bromo-4-(1H-imidazol-1-yl)-2-(thiophen-2-yl)quinoline as a solid (MS: [M+1]+ 356.0).


The following compounds are prepared essentially by the same method described above to prepare I-380.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-381


embedded image




embedded image




embedded image


380.0





I-382


embedded image




embedded image




embedded image


380.0





I-1


embedded image




embedded image




embedded image


306.1





I-275


embedded image




embedded image




embedded image


382.1





I-383


embedded image




embedded image




embedded image


351.0





I-384


embedded image




embedded image




embedded image


351.0





I-385


embedded image




embedded image




embedded image


364.0





I-50


embedded image




embedded image




embedded image


340.0









Example 28: Synthesis of(S)-(2-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)acetyl)glycine (I-156)



embedded image


Step 1: tert-Butyl (S)-(2-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)acetyl)glycinate. To a mixture of (S)-2-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)acetic acid (9.4 mg), tert-butyl glycinate (8.05 mg), EDC HCl (9.2 mg), HOBt (6.5 mg) and DMAP (8.7 mg) was added DMF (0.2 mL) and TEA (0.05 mL). The resulting mixture was stirred overnight at room temperature and diluted with EtOAc (20 mL). The organic solution was washed with water, brine and dried over anhy. Na2SO4. A column chromatography eluting with a gradient of hexanes and EtOAc afforded the desired product (9.0 mg) as yellow solids (MS: [M+1]+ 544.2).


Step 2: (S)-(2-(1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)acetyl)glycine. To a solution of tert-butyl (S)-(2-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)acetyl)glycinate (9.0 mg) in DCM (0.4 mL) was added TFA (0.1 mL). After stirring over 4 hours, the reaction mixture was concentrated to dryness under reduced pressure and diluted with water for lyophilization. A total of 9.2 mg of the title compound, (S)-(2-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)acetyl)glycine, was afforded (MS [M+1]+ : 448).


The following compounds are prepared essentially by the same method described above to prepare I-156.


















MS


I-#
Starting Materials
Structure
[M + 1]+



















I-325


embedded image




embedded image




embedded image


488





I-326


embedded image




embedded image




embedded image


448





I-454


embedded image




embedded image




embedded image


474





I-386


embedded image




embedded image




embedded image


488





I-339


embedded image




embedded image




embedded image


488





I-340


embedded image




embedded image




embedded image


476





I-338


embedded image




embedded image




embedded image


490





I-387


embedded image




embedded image




embedded image


476





I-388


embedded image




embedded image




embedded image


  524.1





I-196


embedded image




embedded image




embedded image


422





I-389


embedded image




embedded image




embedded image


462





I-321


embedded image




embedded image




embedded image


422





I-320


embedded image




embedded image




embedded image


436





I-323


embedded image




embedded image




embedded image


434





I-324


embedded image




embedded image




embedded image


448





I-341


embedded image




embedded image




embedded image


450





I-342


embedded image




embedded image




embedded image


464





I-343


embedded image




embedded image




embedded image


450





I-344


embedded image




embedded image




embedded image


464





I-522


embedded image




embedded image




embedded image


544





I-524


embedded image




embedded image




embedded image


488





I-525


embedded image




embedded image




embedded image


476





I-526


embedded image




embedded image




embedded image


490





I-527


embedded image




embedded image




embedded image


476





I-528


embedded image




embedded image




embedded image


524





I-529


embedded image




embedded image




embedded image


502





I-530


embedded image




embedded image




embedded image


502





I-531


embedded image




embedded image




embedded image


462





I-532


embedded image




embedded image




embedded image


502





I-533


embedded image




embedded image




embedded image


502





I-534


embedded image




embedded image




embedded image


537





I-535


embedded image




embedded image




embedded image


516





I-536


embedded image




embedded image




embedded image


512





I-537


embedded image




embedded image




embedded image


523





I-538


embedded image




embedded image




embedded image


504





I-539


embedded image




embedded image




embedded image


477





I-540


embedded image




embedded image




embedded image


504





I-541


embedded image




embedded image




embedded image


473





I-542


embedded image




embedded image




embedded image


420





I-543


embedded image




embedded image




embedded image


584





I-544


embedded image




embedded image




embedded image


487





I-452


embedded image




embedded image




embedded image


420





I-545


embedded image




embedded image




embedded image


490





I-546


embedded image




embedded image




embedded image


488





I-547


embedded image




embedded image




embedded image


488





I-548


embedded image




embedded image




embedded image


488





I-549


embedded image




embedded image




embedded image


490





I-550


embedded image




embedded image




embedded image


476





I-551


embedded image




embedded image




embedded image


486





I-552


embedded image




embedded image




embedded image


474





I-553


embedded image




embedded image




embedded image


488





I-554


embedded image




embedded image




embedded image


448





I-555


embedded image




embedded image




embedded image


464





I-608


embedded image




embedded image




embedded image


433





I-562


embedded image




embedded image




embedded image


422





I-563


embedded image




embedded image




embedded image


436





I-564


embedded image




embedded image




embedded image


450





I-565


embedded image




embedded image




embedded image


436





I-566


embedded image




embedded image




embedded image


497





I-567


embedded image




embedded image




embedded image


471





I-568


embedded image




embedded image




embedded image


421









The following compounds were prepared essentially by the same method as step 1 described above for I-156.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-556


embedded image




embedded image




embedded image


475





I-557


embedded image


NH4Cl


embedded image


392





I-558


embedded image


NH2OH


embedded image


408





I-603


embedded image


NH3


embedded image


390









The following compounds were prepared essentially by the same method as step 2 described above for I-156.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-559


embedded image


LiOH


embedded image


407





I-560


embedded image


LiOH


embedded image


407





I-561


embedded image


LiOH


embedded image


451









Example 29: Synthesis of N-benzyl-3-((7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)(methyl)amino)propenamide (I-193)



embedded image


Step 1: tert-Butyl 3-((7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)(methyl)amino)propanoate. To a solution of 2,7,8-trichloro-4-(1H-pyrazol-4-yl)quinoline (125 mg, 0.42 mmol) in DMSO (0.5 mL) was added tert-butyl 3-(methylamino)propanoate (133 mg, 0.84 mmol) and N,N-diisopropylethylamine (0.22 mL, 1.26 mmol). The solution is stirred at 95° C. for 16 h. After cooling down to room temperature, water (5 mL) was added and the organics were extracted into 10% methanol in dichloromethane (2×5 mL). The combined organics are dried (Na2SO4) and concentrated. The resulting residue was purified by silica chromatography using 20-80% EtOAc/hexanes to afford tert-butyl 3-((7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)(methyl)amino)propanoate (120 mg) (MS: [M+1]+ 421).


Step 2: 3-((7,8-Dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)(methyl)amino)propanoic acid. To a solution of tert-butyl 3-((7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)(methyl)amino)propanoate (100 mg, 0.24 mmol) in dichloromethane (3 mL) was added hydrochloric acid (0.5 mL, 4.0 M in dioxane). The reaction is stirred at rt for 16 h. The volatiles are removed by rotary evaporation and the resulting solids were used without further purification (MS: [M+1]+ 365).


Step 3: N-Benzyl-3-((7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)(methyl)amino)propenamide. To a solution of 3-((7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)(methyl)amino)propanoic acid (20 mg, 0.055 mmol) in dimethylformamide (0.3 mL) was added HATU (31 mg, 0.082 mmol), N,N-diisopropylethylamine (0.1 mL, 0.57 mmol) and benzylamine (9 mg, 0.082 mmol). The reaction is stirred at rt for 16 hrs. Water (5 mL) was added and the organics were extracted into 10% methanol in dichloromethane (2×5 mL). The combined organics were dried (Na2SO4) and concentrated. The resulting residue was purified by silica chromatography using 0-15% MeOH/CH2Cl2 to afford the titled compound (MS: [M+1]+ 454).


The following compounds are prepared essentially by the same method described above to prepare I-193.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-194


embedded image




embedded image




embedded image


408





I-195


embedded image




embedded image




embedded image


421





I-197


embedded image




embedded image




embedded image


392





I-198


embedded image




embedded image




embedded image


448





I-199


embedded image




embedded image




embedded image


462









Example 30: Synthesis of (S)-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl)carbamate (I-153)



embedded image


To a solution of tert-butyl (S)-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl)carbamate (174 mg) in DCM (2 mL) was added TFA (0.4 mL). After the resultant solution was stirred over 6 hours, evaporation under reduced pressure and lyophilization afforded the title compound (200 mg) as brown powder-tert-butyl (S)-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl)carbamate (MS: [M+1]+ 362).


Example 31: Synthesis of (S)—N-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl)-2-hydroxyacetamide (I-154)



embedded image


To a solution of (S)-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methanamine TFA salt (50 mg) in DCM (0.4 mL) and TEA (0.05 mL) was added 2-chloro-2-oxoethyl acetate (17 mg). After stirred over 4 hours, the reaction mixture was diluted with EtOAc (20 mL) and the organic layer was washed with NaHCO3 and brine. A column chromatography with a gradient of hexane and 5% MeOH in EtOAc afforded the desired intermediate 13.4 mg (MS: [M+1]+ 462.). The intermediate (13.4 mg) was dissolved in MeOH (0.4 mL) and water (0.1 mL) and treated with LiOH·H2O (4 mg) over 2 hours. The reaction mixture was diluted with water (1 mL) and acidified with HOAc (0.02 mL) to precipitate out the product which was further lyophilized to the final product 2260 (7.8 mg) as a powder (MS: [M+1]+ 420).


The following compounds are prepared essentially by the same method described above to prepare I-154.


















MS


I-#
Starting Materials
Structure
[M + 1]+



















I-151


embedded image




embedded image




embedded image


440





I-390


embedded image




embedded image




embedded image


512





I-335


embedded image




embedded image




embedded image


498





I-391


embedded image




embedded image




embedded image


546





I-392


embedded image




embedded image




embedded image


586





I-393


embedded image




embedded image




embedded image


573





I-637


embedded image




embedded image




embedded image


538





I-638


embedded image




embedded image




embedded image


546





I-639


embedded image




embedded image




embedded image


484





I-640


embedded image




embedded image




embedded image


546





I-641


embedded image




embedded image




embedded image


473









Example 32: Synthesis of N-(2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)ethyl)aminosulfonamide (I-223)



embedded image


Step 1: tert-Butyl 2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)ethylcarbamate. To a vial were added 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (100 mg, 0.335 mmol), tert-butyl 2-(methylamino)ethylcarbamate (175 mg, 1.01 mmol), DMF (2.0 mL), and TEA (187 μl, 1.34 mmol). The resulting reaction mixture was stirred at 70° C. for 3 h and cooled to room temperature, followed by adding H2O (20 mL). The cloud mixture was centrifuged and the residue was extracted by DCM (2×10 mL). The organic phase was washed by H2O (10 mL), brine (10 mL), dried over Na2SO4, concentrated to afford an off-white solid (132 mg) (MS: [M+1]+ 436).


Step 2: N-(2-aminoethyl)-7,8-dichloro-4-(1H-imidazol-1-yl)-N-methylquinolin-2-amine. To a vial were added tert-butyl 2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)ethylcarbamate (131 mg, 0.300 mmol), DCM (1.5 mL) and TFA (1.5 mL). The resulting mixture was stirred at room temperature for 2 hrs. Then all volatile solvents were removed under reduced pressure and the residue was dried under high vacuum to afford the title product. The crude was used in next step.


Step 3: N-(2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)ethyl)aminosulfonamide. To a vial were added N-(2-aminoethyl)-7,8-dichloro-4-(1H-imidazol-1-yl)-N-methylquinolin-2-amine (12.5 mg, 0.0373 mmol), DMF (0.5 mL), sulfamoyl chloride (26 mg, 0.224 mmol) and DIPEA (52 μl, 0.296 mmol). The resulting reaction mixture was stirred at 50° C. overnight. The reaction mixture was diluted by ethyl acetate (15 mL). The organic phase was washed by H2O (3×5 mL), brine (5 mL), dried over Na2SO4. After concentration, 30% of the crude was purified by PTLC to afford the title product as an off-white solid (1.3 mg) (MS: [M+1]+ 415).


The following compounds are prepared essentially by the same method described above to prepare I-223.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-224


embedded image




embedded image




embedded image


429





I-346


embedded image




embedded image




embedded image


450





I-347


embedded image




embedded image




embedded image


434





I-349


embedded image




embedded image




embedded image


532





I-350


embedded image




embedded image




embedded image


496





I-351


embedded image




embedded image




embedded image


470









The following compounds were prepared essentially by the same method to prepare I-223. Some analogues were prepared from a general amide formation and a following hydrolysis of an ester to form the corresponding acid.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-609


embedded image




embedded image




embedded image


534





I-610


embedded image




embedded image




embedded image


458





I-611


embedded image




embedded image




embedded image


534





I-612


embedded image




embedded image




embedded image


520





I-613


embedded image




embedded image




embedded image


394





I-614


embedded image




embedded image




embedded image


408





I-615


embedded image




embedded image




embedded image


476





I-616


embedded image




embedded image




embedded image


436





I-617


embedded image




embedded image




embedded image


472





I-618


embedded image




embedded image




embedded image


458





I-619


embedded image




embedded image




embedded image


456





I-620


embedded image




embedded image




embedded image


484





I-621


embedded image




embedded image




embedded image


502





I-622


embedded image




embedded image




embedded image


476





I-623


embedded image




embedded image




embedded image


520









Example 33: Synthesis of (S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-ol (I-119)



embedded image


Step 1: 1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-ol was the same as that for compound I-353 (MS: [M+1]+ 263).


Step 2: (S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl sulfamate. To a vial were added 1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-ol (20 mg, 0.0551 mmol), DMF (0.3 mL) and TEA (50 μl, 0.364 mmol). A stock solution of sulfamoyl chloride in DMF (38 mg/100 μl, 0.33 mmol) was added. The resulting reaction mixture was stirred at 100° C. for 6 hrs and cooled to room temperature followed by adding 4 mL of water. The mixture was centrifuged and the residue was purified by PTLC (30% MeOH/DCM) to afford the desired product as white solid (1 mg) (MS: [M+1]+ 442).


The following compounds are prepared essentially by the same methods as for I-223.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-118


embedded image




embedded image




embedded image


456





I-96


embedded image




embedded image




embedded image


458









Example 34: Synthesis of N-(2-((7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)ethyl)cyclopropanesulfonamide (I-226)



embedded image


Step 1: tert-Butyl (2-((7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)ethyl)carbamate. To a solution of 2,7-dichloro-4-(1H-imidazol-1-yl)quinoline (528 mg, 2.0 mmol) in DMF (2 mL) was added tert-butyl (2-(methylamino)ethyl)carbamate (871 mg, mmol) and Hunig's base (0.5 mL). The solution was stirred at 95° C. for 16 h. After cooling down to room temperature, water (10 mL) was added and the organics were extracted into 10% Methanol/dichloromethane (2×5 mL). The combined organics were dried (Na2SO4) and concentrated. The resulting residue was purified by silica column chromatography using 50-100% EtOAc/Hex to afford tert-butyl (2-((7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)ethyl)carbamate as a solid (MS: [M+1]-Boc 302).


Step 2: N1-(7-Chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-N1-methylethane-1,2-diamine. To a vial was added tert-butyl (2-((7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)ethyl)carbamate (120 mg, 0.30 mmol). Hydrochloric acid (4.0M in dioxane (1.0 mL) was added and the reaction allowed to stir at rt for 16 h. The resulting solids were filtered off and dried to afford N1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-N1-methylethane-1,2-diamine hydrochloride as a solid (MS: [M+1]+ 302).


Step 3: N-(2-((7-Chloro-4-(1H-imidazol-1-yl) quinolin-2-yl) (methyl) amino)ethyl) cyclopropanesulfonamide. To a vial was added N1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-N1-methylethane-1,2-diamine hydrochloride (20 mg, 0.06 mmol) in DMF (0.5 mL) and Hunig's base (0.1 mL). Cyclopropanesulfonyl chloride (17 mg, 0.12 mmol) was added and the reaction allowed to stir at rt for 16 h. Water (5 mL) was added and the aqueous extracted with ethyl acetate (2×5 mL). The combined organics were dried (Na2SO4) then purified by silica chromatography using 0-10% MeOH/CH2Cl2 to afford 12 mg N-(2-((7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)ethyl)cyclopropanesulfonamide (MS: [M+1]+ 406).


The following compounds are prepared essentially by the same method described above to prepare I-226.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-227


embedded image




embedded image




embedded image


442





I-228


embedded image




embedded image




embedded image


510





I-394


embedded image




embedded image




embedded image


510





I-232


embedded image




embedded image




embedded image


446





I-229


embedded image




embedded image




embedded image


476





I-230


embedded image




embedded image




embedded image


432





I-231


embedded image




embedded image




embedded image


443





I-236


embedded image




embedded image




embedded image


344





I-237


embedded image




embedded image




embedded image


406





I-234


embedded image




embedded image




embedded image


380





I-235


embedded image




embedded image




embedded image


414





I-225


embedded image




embedded image




embedded image


414









Example 35: Synthesis of (1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-amine) (I-108)



embedded image


To a vial was added tert-butyl (1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)carbamate (90 mg, 0.21 mmol) with dichloromethane (5 mL). Trifluoromethanesulfonic acid (0.5 mL) was added and the reaction was stirred at rt for 16 h. The volatiles were removed by rotary evaporation and the crude product (1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-amine) (di-TFA salt) was used directly in the next step (MS: [M+1]+ 328).


Example 36: Synthesis of N-(1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)methanesulfonamide (I-105)



embedded image


To a solution of 1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-amine (di TFA salt) (55 mg, 0.10 mmol) in dichloromethane (3 mL) and DMF (0.3 mL) was added triethylamine (0.3 mL) followed by methanesulfonyl chloride (23 mg, 0.20 mmol). The mixture is stirred at rt for 3 h. Water (5 mL) was added and the organics were extracted into 10% methanol in dichloromethane (2×5 mL). The combined organics were dried (Na2SO4) and concentrated. Purification by silica chromatography afforded N-(1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)methanesulfonamide (33 mg) (MS: [M+1]+ 406).


The following compounds are prepared essentially by the same method described above to prepare I-105.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-395


embedded image




embedded image




embedded image


428





I-100


embedded image




embedded image




embedded image


432





I-103


embedded image




embedded image




embedded image


472





I-107


embedded image




embedded image




embedded image


468





I-102


embedded image




embedded image




embedded image


469





I-109


embedded image




embedded image




embedded image


472





I-101


embedded image




embedded image




embedded image


458









Example 37: Synthesis of 1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl hydrogen phosphonate (I-111)



embedded image


Step 1: 1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-ol was prepared according to the procedure described above to prepare compound I-353.


Step 2: 2-Cyanoethyl 1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl phosphonate. To a vial were added 1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-ol (20 mg, 0.0551 mmol), DCM (1.0 mL), pyridinium trifluoroacetate (10.6 mg, 0.0551 mmol). Then 3-(bis(diisopropylamino)phosphinooxy) propanenitrile (26 μl, 0.0826 mmol) was added. The resulting reaction mixture was stirred at room temperature overnight. The reaction was monitored by LC-MS. H2O (0.1 mL) was added and the reaction mixture was stirred for 1 hr. All volatiles were removed under reduced pressure. The crude was used in next step.


Step 3: 1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl hydrogen phosphonate. To a vial containing crude product 2-cyanoethyl 1-(7,8-dichloro-4-(1H-imidazol-1-yl)345uinoline-2-yl)piperidin-3-yl phosphonate in step 2 were added MeOH (0.5 mL), THF (1.0 mL) and a solution of LiOH·H2O in H2O (4.63 mg/0.5 mL, 0.110 mmol). The resulting reaction mixture was stirred overnight, acidified to pH 5, and concentrated. The crude was purified by HPLC to afford the title product as a white solid (8 mg) (MS: [M+1]+ 427).


The following compounds are prepared essentially by the same method described above to prepare I-111.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-110


embedded image




embedded image




embedded image


441





I-255


embedded image




embedded image




embedded image


401





I-396


embedded image




embedded image




embedded image


494





I-329


embedded image




embedded image




embedded image


441





I-257


embedded image




embedded image




embedded image


468





I-256


embedded image




embedded image




embedded image


415





I-330


embedded image




embedded image




embedded image


455





I-258


embedded image




embedded image




embedded image


415









Example 38: Synthesis of 1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl dihydrogen phosphate (I-112)



embedded image


To a vial were added product I-111 (6.2 mg, 0.0145 mmol) and pyridine (0.3 mL). Trimethylsilyl 2,2,2-trifluoro-N-(trimethylsilyl)acetimidate (0.019 mL, 0.0726 mmol) was then added. The resulting reaction mixture was stirred for 5 min. A solution of I2 in pyridine (4.4 mg/0.1 mL) was then added dropwise and the reaction mixture was stirred for 5 min. The volatiles were removed under reduced pressure. The crude was purified by prep HPLC to afford the title product as a white solid (2.6 mg) (MS: [M+1]+ 443).


Example 39: Synthesis of (1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)methyl dihydrogen phosphate (I-113)



embedded image


(1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)methyl dihydrogen phosphate was prepared according to the procedure described above to prepare compound I-112 (MS: [M+1]+ 457).


Example 40: Synthesis of Ethyl hydrogen (7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-ylamino)methylphosphonate (I-253) and (7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-ylamino)methylphosphonic acid (I-254)



embedded image


Step 1: Ethyl hydrogen (7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-ylamino)methylphosphonate. To a vial were added 2,7,8-trichloro-4-(1H-pyrazol-4-yl)quinoline (30 mg, 0.10 mmol), DMSO (0.3 mL), diethyl aminomethylphosphonate (50 mg, 0.3 mmol), and DIPEA (52 μl, 0.3 mmol). The resulting reaction mixture was stirred at 130° C. for 20 hrs. The crude was purified by prep HPLC to afford product 2157 as a white solid (5.2 mg) (MS: [M+1]+ 401).


Step 2: (7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-ylamino)methylphosphonic acid. To a vial were added ethyl hydrogen (7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-ylamino)methylphosphonate (3.7 mg, 0.0092 mmol) and hydrochloric acid (37%, 0.5 mL). The resulting reaction mixture was stirred at 70° C. overnight. All starting material was converted to desired product. After concentration, the title product was obtained as a white solid (3.5 mg) (MS: [M+1]+ 373).


Example 41: Synthesis of (3-((7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)amino)propyl)phosphonic acid (I-397)



embedded image


Step 1: Diethyl 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-ylamino)propylphosphonate. To a vial were added 2,7,8-trichloro-4-(1H-pyrazol-4-yl)quinoline (30 mg, 0.10 mmol), DMSO (0.3 mL), diethyl 3-aminopropylphosphonate (58.6 mg, 0.30 mmol) and DIPEA (52 μl, 0.30 mmol). The resulting reaction mixture was stirred at 110° C. for 5 hrs followed by addition of H2O (4 mL). The precipitated white solid was collected by a centrifuge, and the crude was purified by silica gel chromatography (eluted by 5% MeOH/DCM) to afford the desired product as a white solid (9 mg) (MS: [M+1]+ 457).


Step 2: 3-(7,8-Dichloro-4-(1H-pyrazol-4-yl)quinolin-2-ylamino)propylphosphonic acid. To a vial were added diethyl 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-ylamino)propylphosphonate (4.5 mg, 0.0098 mmol) and hydrochloric acid (37%, 0.5 mL). The resulting reaction mixture was stirred at 70° C. overnight. All starting material was converted to desired product. After concentration, the title product was obtained as a white solid (4.0 mg) (MS: [M+1]+ 401).


Example 42: Synthesis of diethyl (1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)methylphosphonate (I-114)



embedded image


Step 1: (1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)methanol was prepared according to the procedure described above to prepare compound I-353. (MS: [M+1]+ 377).


Step 2: 2-(3-(Bromomethyl)piperidin-1-yl)-7,8-dichloro-4-(1H-imidazol-1-yl)quinoline. To a vial were added (1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)methanol (100 mg, 0.265 mmol), CBr4 (176 mg, 0.53 mmol), and DCM (5 mL). The mixture was cooled in ice-water bath for 20 min followed by addition of a solution of Ph3P in DCM (105 mg/l mL, 0.398 mmol). The resulting mixture was stirred at 0° C. for 1 hr. The mixture was concentrated, purified by silica gel chromatography eluting by 50% to 60% of ethyl acetate in hexanes to afford desired product as an off-white solid (85 mg) (MS: [M+1]+ 439).


Step 3: Diethyl (1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)methylphosphonate. To a vial were added 2-(3-(bromomethyl)piperidin-1-yl)-7,8-dichloro-4-(1H-imidazol-1-yl)quinoline (20 mg, 0.0455 mmol) and triethyl phosphate (0.5 mL). The resulting mixture was stirred at 150° C. for 10 hrs. The reaction mixture (30%) was purified by PTLC to afford the desired product as a white solid (2.0 mg) (MS: [M+1]+ 497).


Step 4: (1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)methylphosphonic acid was prepared according to the procedure described above to prepare compound I-397 (step 2) (MS: [M+1]+ 441).


Example 43: Synthesis of Ethyl hydrogen (7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)methylphosphonate (I-252)



embedded image


Step 1: 2-(Bromomethyl)-4,7,8-trichloroquinoline. To a flask were added 4,7,8-trichloro-2-methylquinoline (493 mg, 2.0 mmol), NBS (356 mg, 2.0 mmol), AIBN (66 mg, 0.40 mmol) and CCl4 (6 mL). The reaction mixture was degassed by bubbling N2 flow for 20 min and stirred at 65° C. for 16 hrs. under N2. After being cooled to room temperature, the mixture was diluted by ethyl acetate (50 mL), washed by H2O (20 mL) and brine (20 mL), dried over Na2SO4. After concentration, the crude was used in next step.


Step 2: Diethyl (4,7,8-trichloroquinolin-2-yl)methylphosphonate was prepared according to the procedure described above to prepare compound 2060 step 3 (MS: [M+1]+ 382).


Step 3: Ethyl hydrogen (7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)methylphosphonate was prepared according to the procedure described above to prepare compound I-56 (step 3) (MS: [M+1]+386).


Example 44: Synthesis of ((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)methyl)phosphonic acid (I-251)



embedded image


((7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)methyl)phosphonic acid was prepared according to the procedure described above to prepare compound I-397 (step 2) (MS: [M+1]+ 358).


Example 45: Synthesis of 2-(benzyloxy)-7-chloro-4-(1H-imidazol-1-yl)quinoline (I-277)



embedded image


To a vial were added 2,7-dichloro-4-(1H-imidazol-1-yl)quinoline (132 mg, 0.5 mmol), DMSO (1 mL), benzyl alcohol (62 μl, 0.6 mmol) and NaOH (24 mg, 0.6 mmol). The mixture was stirred at 100° C. overnight. The 10% of the reaction mixture was purified by PTLC, eluted by 7.5% MeOH/DCM to afford the title product as white solid (4.5 mg, 27% yield) (MS: [M+1]+ 336.1).


The following compounds are prepared essentially by the same method described above to prepare I-277.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-278


embedded image




embedded image




embedded image


322





I-282


embedded image




embedded image




embedded image


366





I-281


embedded image




embedded image




embedded image


380





I-284


embedded image




embedded image




embedded image


373





I-283


embedded image




embedded image




embedded image


366





I-280


embedded image




embedded image




embedded image


274





I-279


embedded image




embedded image




embedded image


359









Example 46: Synthesis of (7,8-Dichloro-2-(1H-pyrazol-3-yl)quinolin-4-yl)glycine (I-287)



embedded image


Step 1: tert-Butyl (4,7,8-trichloroquinolin-2-yl)glycinate and tert-butyl (2,7,8-trichloroquinolin-4-yl)glycinate. To a solution of 2,4,7,8-tetrachloroquinoline (270 mg) and tert-butyl glycinate HCl (270 mg) in DMSO (0.8 mL) was added K2CO3 (276 mg). The resultant reaction mixture was heated at 85° C. over 4 h. Aqueous work up with EtOAc (30 mL) and a column chromatography eluting with a gradient of DCM/Hexane from 0 to 50% afforded two colorless compounds. The earlier eluted fraction is 4-substituted product (120 mg) (MS: [M+1]+ 361) and the later fraction with 2-substituted product (62 mg) (MS: [M+1]+ 361).


Step 2: tert-Butyl (7,8-dichloro-2-(1H-pyrazol-4-yl)quinolin-4-yl)glycinate. To a mixture of tert-butyl (2,7,8-trichloroquinolin-4-yl)glycinate (52 mg), 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (50 mg), K2CO3 (48 mg) and Pd(PPh3)4 (16 mg) were added dioxane (2 mL) and water (0.5 mL). The resultant suspended solution was vacuumed and purged with nitrogen repeatedly three time, then stirred and heated at 85° C. under nitrogen over 4 hours. An aqueous work-up with EtOAc and a column chromatography gave the desired colorless product (27 mg) (MS: [M+1]+ 393).


Step 3: (7,8-Dichloro-2-(1H-pyrazol-3-yl)quinolin-4-yl)glycine. To a solution of tert-butyl (7,8-dichloro-2-(1H-pyrazol-3-yl) quinolin-4-yl) glycinate (27 mg) in DCM (0.5 mL) was added TFA (0.2 mL). The resultant solution was stirred overnight. After removal of DCM and TFA under reduced pressure, the resultant was mixed with 0.4 mL water and lyophilized to afford the title product (MS: [M+1]+ 337).


The following compounds are prepared essentially by the same method described above to prepare I-287 and some analogues are prepared from intermediates by additional deprotection.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-285


embedded image




embedded image




embedded image


351





I-290


embedded image




embedded image




embedded image


407





I-286


embedded image




embedded image




embedded image


351





I-300


embedded image




embedded image




embedded image


337





I-288


embedded image




embedded image




embedded image


337





I-301


embedded image




embedded image




embedded image


386





I-401


embedded image




embedded image




embedded image


319





I-295


embedded image




embedded image




embedded image


354





I-296


embedded image




embedded image




embedded image


320





I-161


embedded image




embedded image




embedded image


334





I-306


embedded image




embedded image




embedded image


305





I-294


embedded image




embedded image




embedded image


305





I-171


embedded image




embedded image




embedded image


305





I-307


embedded image




embedded image




embedded image


330





I-312


embedded image




embedded image




embedded image


316









Example 47: Synthesis of (7,8-dichloro-4-((2-sulfamoylethyl)amino)quinolin-2-yl)glycine (I-319)



embedded image


Step 1: tert-Butyl (7,8-dichloro-4-((2-sulfamoylethyl)amino)quinolin-2-yl)glycinate. To a mixture of tert-butyl (4,7,8-trichloroquinolin-2-yl) glycinate (28 mg), 2-aminoethane-1-sulfonamide (20 mg) and K2CO3 (22 mg) was added DMSO (0.4 mL). After stirring at 130° C. overnight, the reaction mixture was diluted with water and extracted with EtOAc (15 mL). A column chromatography afforded the desired product (6 mg) (MS: [M+1]+ 449).


Step 2: (7,8-Dichloro-4-((2-sulfamoylethyl)amino)quinolin-2-yl)glycine. The acidic deprotection of tert-butyl (7,8-dichloro-4-((2-sulfamoylethyl) amino) quinolin-2-yl) glycinate (6 mg) in DCM (0.4 mL) with TFA (0.1 mL) afforded the desired product (5 mg) (MS: [M+]1+ 393).


Example 48: Synthesis of (7,8-dichloro-2-((2-sulfamoylethyl)amino)quinolin-4-yl)glycine (I-318)



embedded image


Following the similar preparation procedure of I-319, tert-butyl (4,7,8-trichloroquinolin-2-yl)glycinate (27.8 mg) was treated with 2-aminoethane-1-sulfonamide (18.5 mg) in DMSO and the following deprotection afforded the desired product (5.9 mg) (MS: [M+1]+ 393).


Example 49: Synthesis of (7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)glycine (I-177)



embedded image


(7,8-Dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)glycine was prepared essentially by the same method described above to prepare I-287.


The following compounds are prepared essentially by the same method described above to prepare I-177.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-402


embedded image




embedded image




embedded image


407





I-180


embedded image




embedded image




embedded image


351





I-181


embedded image




embedded image




embedded image


351





I-265


embedded image




embedded image




embedded image


386





I-403


embedded image




embedded image




embedded image


319









Example 50: Synthesis of 4,7,8-trichloro-2-(1H-pyrazol-4-yl)quinoline (I-404) and 2-((7,8-dichloro-2-(1H-pyrazol-4-yl)quinolin-4-yl)amino)ethan-1-ol (I-289)



embedded image


Step 1: 4,7,8-Trichloro-2-(1H-pyrazol-4-yl)quinoline. To a mixture of 2,4,7,8-tetrachloroquinoline (430 mg), 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (345 mg), Na2CO3 (513 mg) and Pd(PPh3)4 (186 mg) were added dioxane (6 mL) and water (3 mL). The resultant mixture was vacuumed and purged with N2 repeatedly three time, then stirred and heated at 90° C. over 2 hours. A solution of 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (300 mg) in dioxane (2 mL) was added to the reaction mixture. After 1.5 h, the second portion of 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (300 mg) in dioxane (2 mL) was added. After 1.5 hour, the starting material-2,4,7,8-tetrachloroquinoline was completely consumed and only one major product was observed by LC-MS. The reaction mixture was diluted with water (4 mL) to precipitate out the desired product. After isolation by a filtration, the isolated solid was washed with water (4 mL) and EtOAc/Hexane (1:1, 6 mL) to afford the desired product I-404 (350 mg) (MS: [M+1]+ 298).


Step 2: 2-((7,8-Dichloro-2-(1H-pyrazol-4-yl)quinolin-4-yl)amino)ethan-1-ol. To a mixture of 4,7,8-trichloro-2-(1H-pyrazol-4-yl) quinoline (45 mg) and 2-aminoethan-1-ol (46 mg) were added Na2CO3 (32 mg) and DMSO (0.4 mL). The resultant suspension was stirred at 110° C. overnight until the starting material was consumed. Dilution with water (3 mL) precipitated the desired product. Isolation by centrifuge, rinsing with water and drying under vacuum afforded the title product (27.2 mg) (MS: [M+1]+ 323).


The following compounds are prepared essentially by the same method described above to prepare I-289.


















MS


I-#
Starting Materials
Structure
[M + 1]+



















I-297


embedded image




embedded image




embedded image


337





I-292


embedded image




embedded image




embedded image


377





I-291


embedded image




embedded image




embedded image


377





I-298


embedded image




embedded image




embedded image


363





I-299


embedded image




embedded image




embedded image


362









The following compounds are prepared essentially by the same method described above to prepare I-289 (Suzuki coupling, nucleophilic substitution, and deprotection).


















MS


Example
Starting Materials
Structure
[M + 1]+




















I-425


embedded image




embedded image




embedded image




embedded image


351





I-426


embedded image




embedded image




embedded image




embedded image


337









Example 51: Synthesis of 2-(benzyloxy)-7-chloro-4-(1H-imidazol-1-yl)quinoline (I-405)



embedded image


To a solution of 4,7,8-trichloro-2-methylquinoline (0.5 g) in DMF (4 mL) were added K2CO3 (0.2 g) and imidizaole (0.54 g). The resultant suspension was heated at 120° C. over 2 h. The reaction mixture was diluted with water (8 mL) to precipitate out the title compound. Filtration and rinsing with water afforded the title compound (0.55 g)-7,8-dichloro-4-(1H-imidazol-1-yl)-2-methylquinoline (MS: [M+1]+ 278).


The following compounds are prepared essentially by the same method described above to prepare I-405. Some analogues were isolated in pure form by precipitation from water and others were purified by column chromatography.


















MS


I-#
Starting Materials
Structure
[M + 1]+



















I-316


embedded image




embedded image




embedded image


353





I-315


embedded image




embedded image




embedded image


310





I-313


embedded image




embedded image




embedded image


284





I-314


embedded image




embedded image




embedded image


298





I-305


embedded image




embedded image




embedded image


324





I-310


embedded image




embedded image




embedded image


324





I-302


embedded image




embedded image




embedded image


310





I-311


embedded image




embedded image




embedded image


325





I-317


embedded image




embedded image




embedded image


374.0





I-308


embedded image




embedded image




embedded image


334.0





I-303


embedded image




embedded image




embedded image


345.0





I-293


embedded image




embedded image




embedded image


285.0





I-304


embedded image




embedded image




embedded image


271.0





I-309


embedded image




embedded image




embedded image


299.0





I-66


embedded image




embedded image




embedded image


348





I-65


embedded image




embedded image




embedded image


346





I-64


embedded image




embedded image




embedded image


345





I-62


embedded image




embedded image




embedded image


331





I-61


embedded image




embedded image




embedded image


334





I-63


embedded image




embedded image




embedded image


336





I-60


embedded image




embedded image




embedded image


320





I-52


embedded image




embedded image




embedded image


406





I-39


embedded image




embedded image




embedded image


414









Example 52: Synthesis of 7,8-dichloro-2-methyl-4-(1H-pyrazol-4-yl)quinoline (I-406)



embedded image


To a mixture of 4,7,8-trichloro-2-methylquinoline (0.2 g), 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (281 mg), K2CO3 (278 mg) and Pd(PPh3)4 (85 mg) were added dioxane (4 mL) and water (2 mL). The resultant mixture was vacuumed and purged with N2 repeatedly three time, then stirred and heated at 90° C. over 2 hours. A solution of 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (150 mg) in dioxane (2 mL) was degassed and added to the reaction mixture. After 2 hours, the second portion of 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (150 mg) in dioxane (2 mL) was added. After stirring overnight, 2,4,7,8-tetrachloroquinoline was completely consumed. Aqueous work-up and column purification eluting with hexane/EtOAc afforded the title compound (108 mg)-7,8-dichloro-2-methyl-4-(1H-pyrazol-4-yl) quinoline-MS: [M+1]+ 278.


The following compounds are prepared essentially by the same method described above to prepare I-406 and the Suzuki coupling in I-42.


















MS


I-#
Starting Materials
Structure
[M + 1]+



















I-407


embedded image




embedded image




embedded image


278





I-67


embedded image




embedded image




embedded image


306





I-68


embedded image




embedded image




embedded image


306





I-73


embedded image




embedded image




embedded image


333





I-72


embedded image




embedded image




embedded image


305





I-70


embedded image




embedded image




embedded image


317





I-69


embedded image




embedded image




embedded image


317





I-71


embedded image




embedded image




embedded image


318









Example 53: Synthesis of 8-chloro-4-(1H-imidazol-1-yl)quinoline (I-408)



embedded image


4,8-Dichloroquinoline (50 mg, 0.25 mmol) was placed in a vial with dioxane (2 mL). Imidazole (68 mg, 1.0 mmol) was added and the reaction was heated to 130° C. for 16 h. Water (10 mL) was added to the reaction and then the organics were extracted into ethyl acetate (2×5 mL). The organic phase was dried (Na2SO4) and concentrated. The residue was purified by silica chromatography using 30-100% (EtOAc/Hexanes) to afford 8-chloro-4-(1H-imidazol-1-yl)quinoline as a solid (MS: [M+1]+ 230).


The following compounds are prepared essentially by the same method described above to prepare I-408.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-409


embedded image




embedded image




embedded image


274





I-410


embedded image




embedded image




embedded image


196





I-411


embedded image




embedded image




embedded image


210





I-412


embedded image




embedded image




embedded image


274





I-413


embedded image




embedded image




embedded image


196





I-414


embedded image




embedded image




embedded image


274





I-415


embedded image




embedded image




embedded image


274





I-416


embedded image




embedded image




embedded image


214





I-417


embedded image




embedded image




embedded image


226.1





I-418


embedded image




embedded image




embedded image


275.0





I-419


embedded image




embedded image




embedded image


2





I-420


embedded image




embedded image




embedded image


74.9









Example 54: Synthesis of 7-chloro-4-(1H-imidazol-1-yl)-8-methylquinoline (I-421)



embedded image


4,7-Dichloro-8-methylquinoline (53 mg, 0.25 mmol), imidazole (43 mg, 0.63 mmol), potassium t-butoxide (42 mg, 0.38 mmol), Bis(triphenylphosphine)palladium(II) dichloride (9 mg, 0.013 mmol) and DMF (3 mL) were placed in a vial under N2. The mixture was heated at 110° C. for 2 h. After cooling down to room temperature, the crude is diluted by EtOAc (20 mL) and washed by water (5 mL×2) and brine (5 mL×2). The organic phase is concentrated and purified by column chromatography on silica gel to give 7-chloro-4-(1H-imidazol-1-yl)-8-methylquinoline as a solid. (MS: [M+1]+ 244.0)


The following compounds are prepared essentially by the same method described above to prepare I-421.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-422


embedded image




embedded image




embedded image


298









Example 55: Synthesis of 7-chloro-4-(1H-imidazol-1-yl)-8-methylquinoline (I-82)



embedded image


Step 1: 1-tert-Butyl 2-methyl 4-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperazine-1,2-dicarboxylate was prepared essentially by the same method described above to prepare I-79.


Step 2: Methyl 4-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperazine-2-carboxylate. To a vial were added 1-tert-butyl 2-methyl 4-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperazine-1,2-dicarboxylate (276 mg), DCM (1.0 mL) and TFA (1.0 mL). The resulting reaction mixture was stirred at room temperature for 2 hrs. All solvents were removed under reduced pressure. The residue was dried under high vacuum and the crude was used in next step (MS: [M+1]+ 506).


Step 3: Methyl 4-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-1-methylpiperazine-2-carboxylate. To a vial were added methyl 4-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperazine-2-carboxylate (0.025 mmol), DMF (0.5 mL), Cs2CO3 (16 mg, 0.05 mmol), a solution of CH3I in DMF (1.87 μl/0.5 mL, 0.03 mmol). The resulting reaction mixture was stirred at room temperature for 3 hrs. The reaction mixture was diluted with ethyl acetate (20 mL), washed by water (5 mL×2) and brine (5 mL), and dried over Na2SO4. The crude was purified by silica gel chromatography to afford 4 mg of the title compound (MS: [M+1]+ 420).


Step 4: 4-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-1-methylpiperazine-2-carboxylic acid. To a vial were added methyl 4-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-1-methylpiperazine-2-carboxylate (4.4 mg, 0.0105 mmol), MeOH (0.25 mL), THF (0.5 mL), a solution of LiOH—H2O in H2O (2.2 mg/0.25 mL, 0.0523 mmol). The resulting reaction mixture was stirred at room temperature overnight, acidified to pH 4. The cloudy mixture was centrifuged and the residue was dried under high vacuum to afford 4 mg the titled product as a white solid (MS: [M+1]+ 406).


The following compounds were prepared essentially by the same method described above to prepare I-82.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-80


embedded image




embedded image




embedded image


420





I-81


embedded image




embedded image




embedded image


436









Example 56: Synthesis of 4-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperazine-2-carboxylic acid (I-79)



embedded image


Step 1: 1-(tert-Butyl) 2-methyl 4-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperazine-1,2-dicarboxylate. A solution of 2,7,8-trichloro-4-(1H-imidazol-1-yl) quinoline (120 mg), 1-(tert-butyl) 2-methyl piperazine-1,2-dicarboxylate (230 mg), and DIPEA (0.1 mL) in DMF (0.6 mL) was heated at 90° C. overnight until the starting material was consumed. Aqueous work-up with EtOAc (25 mL)/water (10 mL) and a column chromatography eluting with a gradient of hexanes and EtOAc afforded the title compound (85 mg) (MS: [M+1]+ 506).


Step 2: 4-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperazine-2-carboxylic acid. To a solution of 1-(tert-butyl) 2-methyl 4-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperazine-1,2-dicarboxylate (85 mg) in dioxane (2 mL) was added 2 N HCl (2 mL). The resultant mixture was heated at 80° C. over 4 hours until the Boc protecting group and the methyl ester were removed. Evaporation under reduced pressure and lyophilization afforded the title compound (80 mg) as a di-HCl salt (MS: [M+1]+392).


Example 57: Synthesis of 4-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-1-(2-hydroxyacetyl)piperazine-2-carboxylic acid (I-83)



embedded image


Step 1: Methyl 1-(2-acetoxyacetyl)-4-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperazine-2-carboxylate. Methyl 4-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperazine-2-carboxylate TFA salt (20 mg) in DMF (0.5 mL) and TEA (0.1 mL) was treated with 2-chloro-2-oxoethyl acetate (20 mg) over 4 hours. Aqueous work up with EtOAc/water/sat NaHCO3/brine and purification by column chromatography afforded methyl 1-(2-acetoxyacetyl)-4-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperazine-2-carboxylate (12 mg) (MS: [M+1]+ 506).


Step 2: 4-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-1-(2-hydroxyacetyl)piperazine-2-carboxylic acid. The intermediate was dissolved in MeOH (0.8 mL) and water (0.2 mL) and treated with LiOH—H2O (20 mg) overnight. The reaction mixture was diluted with water (2 mL) and acidified with HOAc (0.02 mL) to precipitate 4-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-1-(2-hydroxyacetyl)piperazine-2-carboxylic acid (7 mg) (MS: [M+1]+ 450).


Example 58: Synthesis of 1-(7,8-dichloro-4-(1H-imidazol-1-yl) quinolin-2-yl)-3-(2-hydroxyacetamido) piperidine-3-carboxylic acid (I-125)



embedded image


Step 1: 3-Amino-1-(7,8-dichloro-4-(1H-imidazol-1-yl) quinolin-2-yl)piperidine-3-carboxylic acid. A solution of 2,7,8-trichloro-4-(1H-imidazol-1-yl) quinoline (30 mg) with 3-aminopiperidine-3-carboxylic acid (65 mg) and triethylamine (0.2 mL) in DMF (0.5 mL) was heated at 90° C. over 5 hours until the starting material was consumed. The resultant mixture was diluted with water (2 mL), frozen and lyophilized to afford a mixture containing the title compound, which was used in the next step directly (MS: [M+1]+ 406).


Step 2: Methyl 1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-(2-hydroxyacetamido)piperidine-3-carboxylate. The mixture from the above step was suspended in anhydrous DCM (2 mL) and mixed with 2-chloro-2-oxoethyl acetate (0.06 mL) and TEA (0.1 mL). The resultant mixture was stirred at room temperature over 3 hours until the starting was completely consumed. The reaction mixture was concentrated to dryness under reduced pressure. The resultant mixture was dissolved in anhydrous MeOH (5 mL) and treated with a couple of drops of SOCl2. After stirring overnight, the acid was converted to the methyl ester. After solvent evaporation under reduced pressure and aqueous work up with EtOAc, column chromatography afforded the desired methyl ester (12 mg). (MS: [M+1]478)


Step 3: 1-(7,8-dichloro-4-(1H-imidazol-1-yl) quinolin-2-yl)-3-(2-hydroxyacetamido) piperidine-3-carboxylic acid. Methyl 1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-(2-hydroxyacetamido)piperidine-3-carboxylate (12 mg) was treated with LiOH—H2O (8 mg) in MeOH (2 mL) and water (0.5 mL) over 4 hours. After evaporation of the organic solvents under reduced pressure, the resultant solid mixtures were carefully suspended in water (1 mL) and neutralized with HOAc (0.020 mL). The solid was isolated by centrifuge and rinsed with water (1 mL). The resultant wet cake was lyophilized to afford the title compound (7 mg) as a colorless powder (MS: [M+1]+ 464).


Example 59: Synthesis of 7-chloro-4-(1H-imidazol-1-yl)-2-(piperazin-1-yl)quinoline (I-76)



embedded image


A solution of 2,7-dichloro-4-(1H-imidazol-1-yl)quinoline (46 mg) in EtOH (5 mL) was treated with tert-butyl piperazine-1-carboxylate (60 mg) in the presence of K2CO3 (48 mg) overnight at 80° C. Aqueous work-up with EtOAc and water removed inorganic bases. The separation of the organic layer and evaporation of organic solvents afforded the desired tert-butyl 4-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperazine-1-carboxylate intermediate. Deprotection with DCM (3 mL) and TFA (1 mL) in 4 hours at room temperature gave the TFA salt of the desired product. The TFA salt were dissolved in DMF (1 mL) and precipitated out as neutral product (40 mg) after adding an aqueous solution of NaHCO3 to afford the title compound (30 mg) (MS: [M+1]+ 314).


Example 60: Synthesis of 1-(4-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperazin-1-yl)-2-hydroxyethan-1-one (I-77)



embedded image


A solution of 7-chloro-4-(1H-imidazol-1-yl)-2-(piperazin-1-yl)quinoline (10 mg, 1981) in DMF (1 mL) was added to a mixture of 2-chloro-2-oxoethyl acetate (56 mg) and TEA (0.1 mL). After stirring over 2 hours, aqueous work-up with EtOAc (10 mL) and evaporation under reduced pressure afforded a residue with the desired intermediate 2-(4-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperazin-1-yl)-2-oxoethyl acetate (20 mg). (MS: [M+1]+ 414). This residue was treated with LiOH (60 mg) in MeOH (4 mL) and water (1 mL) over 4 hours. Aqueous work-up with EtOAc and a column chromatography afforded the desired product (7 mg) (MS: [M+1]+ 372).


Example 61: Synthesis of 7-chloro-4-(1H-imidazol-1-yl)-2-(4-(methylsulfonyl) piperazin-1-yl) quinoline (I-78)



embedded image


To a solution of 7-chloro-4-(1H-imidazol-1-yl)-2-(piperazin-1-yl) quinoline (10 mg) in DMF (1 mL) were added methanesulfonyl chloride (30 mg) and TEA (0.1 mL). After stirring overnight, aqueous work-up with EtOAc/water and a column chromatography afforded the titled product (8 mg) (MS: [M+1]+ 392).


Example 62: Synthesis of 2-(4-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-oxo-1,4-diazepan-1-yl)acetic acid (I-90)



embedded image


Step 1: tert-butyl 2-(4-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-oxo-1,4-diazepan-1-yl)acetate. To a solution of 4-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-1,4-diazepan-2-one (20 mg) and tert-butyl 2-bromoacetate (30 mg) in anhydrous DMF was added NaH (10 mg, 65% in mineral oil). After stirring 3 hours, the reaction mixture was diluted with EtOAc (10 mL) and carefully quenched with water (5 mL). Isolation of the organic layer and a column chromatography eluting with a gradient of hexanes and EtOAc afforded the desired intermediate tert-butyl 2-(4-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-oxo-1,4-diazepan-1-yl)acetate (20 mg) (MS: [M+1]+ 456).


Step 2: 2-(4-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-oxo-1,4-diazepan-1-yl)acetic acid. tert-butyl 2-(4-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-oxo-1,4-diazepan-1-yl)acetate was further treated with TFA (0.4 mL) in DCM (0.8 mL). Removal of DCM and TFA under reduced pressure and lyophilization afforded the desired product (10 mg)-2-(4-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-oxo-1,4-diazepan-1-yl)acetic acid (MS: [M+1]+ 400).


Example 63: Synthesis of (R)-1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-N-hydroxypiperidine-3-carboxamide (I-137)



embedded image


To a solution of (R)-1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidine-3-carboxylic acid (25 mg, 0.05 mmol, TEA salt) in DMF (2 mL) was added (COCl)2 (0.017 mL, 0.2 mmol) at 0° C. The solution was stirred at rt for 1 h. The mixture was evaporated to give a crude as white solid.


Hydroxylamine hydrochloride (35 mg, 0.5 mmol) and TEA (0.1 mL) was dissolved in THF (0.5 mL) and water (0.1 mL) at 0° C. A solution of the crude from the first step in DCM (1 mL) was added. The mixture was stirred at rt overnight. The crude was purified directly by column chromatography on silica gel to give (R)-1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-N-hydroxypiperidine-3-carboxamide as a solid. (11 mg, 59% yield) (MS: [M+1]+ 372).


Example 64: Synthesis of (1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)boronic acid (I-120)



embedded image


Step 1: 7,8-Dichloro-4-(1H-imidazol-1-yl)-2-(3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)piperidin-1-yl)quinoline. To a solution of 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (30 mg, 0.1 mmol) and 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)piperidine (38 mg, 0.15 mmol) in DMF (1 mL) was added NaHCO3 (42 mg, 0.5 mmol). The solution was vigorously stirred at 100° C. for 2 h. After cooling down to room temperature, water (2 mL) was added. The crude was collected by filtration (50 mg) which is used for next step without purification (MS: [M+1]+ 472).


Step 2: (1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)boronic acid. To a mixture of 7,8-dichloro-4-(1H-imidazol-1-yl)-2-(3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)piperidin-1-yl)quinoline (50 mg, 0.1 mmol), NaIO4 (43 mg, 0.2 mmol) and NH4OAc (15 mg, 0.2 mmol) was added water (1 mL) and acetone (1 mL). The mixture was stirred at rt overnight. After evaporation, the crude was purified directly by column chromatography on silica gel to give the titled product as a solid (35 mg) (MS: [M+1]+ 391).


Example 65: Synthesis of tert-butyl ((2-(((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)methyl)morpholino)sulfonyl)carbamate (I-398) and 2-(((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)methyl)morpholine-4-sulfonamide (I-219)



embedded image


Step 1: tert-Butyl 2-(((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)methyl)morpholine-4-carboxylate. To a solution of 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (300 mg, 1 mmol) in DMF (2 mL) was added tert-butyl 2-(aminomethyl)morpholine-4-carboxylate (320 mg, 1.5 mmol) and NaHCO3 (336 mg, 4 mmol). The mixture was vigorously stirred at 120° C. for 2 h. After cooling down to rt, DMF was removed by evaporation. The residue was dissolved in DCM (20 mL) and washed by H2O (10 mL) and brine (10 mL×2). The organic phase was collected and concentrated to give white solid (500 mg) which was used directly without purification (MS: [M+1]+478).


Step 2: 7,8-Dichloro-4-(1H-imidazol-1-yl)-N-(morpholin-2-ylmethyl)quinolin-2-amine. A solution of tert-butyl 2-(((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)methyl)morpholine-4-carboxylate (500 mg crude from step 1) in 50% TFA in DCM (5 mL) was stirred at rt for 1 h. After evaporation, the crude was purified directly by column chromatography on silica gel to give the titled product as a solid. (110 mg) (MS: [M+1]+ 378).


Step 3: tert-Butyl ((2-(((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)methyl)morpholino)sulfonyl)carbamate. To a solution of chlorosulfonyl isocyanate (0.1 mL, 1.15 mmol) in DCM (1 mL) was added t-BuOH (0.085 mL, 1.15 mmol) at 0° C. for 30 min. To a solution of 7,8-dichloro-4-(1H-imidazol-1-yl)-N-(morpholin-2-ylmethyl)quinolin-2-amine (48 mg, 0.1 mmol) and triethylamine (0.04 mL, 0.3 mmol) in DCM (1 mL) was added the N-chlorosulfonyl carbamate solution above (0.1 mL, 0.115 mmol) at 0° C. After 2 h, the mixture was evaporated and purified by column chromatography on silica gel to give the titled product as a solid (25 mg) (MS: [M+1]+ 557).


Step 4: 2-(((7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)methyl)morpholine-4-sulfonamide. A solution of tert-butyl ((2-(((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)methyl)morpholino)sulfonyl)carbamate (11 mg, 0.02 mmol) in 50% TFA in DCM (1 mL) was stirred at rt for 1 h. After evaporation, the crude was purified directly by column chromatography on silica gel to give the titled product as a solid (3 mg) (MS: [M+1]+ 457.0).


The following compounds are prepared essentially by the same method described above to prepare I-398 and I-219.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-399


embedded image




embedded image




embedded image


478.2





I-220


embedded image




embedded image




embedded image


378.1





I-217


embedded image




embedded image




embedded image


378.1









Example 66: Synthesis of 7-chloro-4-(1H-imidazol-1-yl)-8-methylquinoline (I-221)



embedded image


To a solution of 7,8-dichloro-N-(morpholin-2-ylmethyl)-4-(1H-pyrazol-4-yl)quinolin-2-amine (20 mg, 0.053 mmol) in THF (1 mL) was added triethylamine (0.074 mL, 0.53 mmol) and acetyl chloride (0.0057 mL, 0.08 mmol) at 0° C. After 1 h at rt, the reaction was quenched by MeOH (0.1 mL). After evaporation, the crude was purified directly by column chromatography on silica gel to give 1-(2-(((7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)amino)methyl)morpholino)ethan-1-one as a solid (7 mg) (MS: [M+1]+ 420.0).


The following compounds are prepared essentially by the same method described above to prepare I-221.


















MS


I-#
Starting Material
Structure
[M + 1]+







I-400


embedded image




embedded image


478.1





I-222


embedded image




embedded image


436.1









Example 67: Synthesis of 7-bromo-4-(1H-imidazol-1-yl)-2-phenylquinoline (I-424)



embedded image


Step 1: 7-Bromo-2-phenylquinolin-4-ol (250 mg, 0.83 mmol) was placed in a flask with phosphorus oxychloride (8 mL) and heated to 100° C. for 4 h. After removal of the volatiles under reduced pressure, the residue was dried in vacuo. The crude 7-bromo-4-chloro-2-phenylquinoline was taken onward without further purification.


Step 2: 7-Bromo-4-chloro-2-phenylquinoline (50 mg, 0.16 mmol), imidazole (27 mg, 0.39 mmol), potassium t-butoxide (26 mg, 0.23 mmol), Bis(triphenylphosphine)palladium(II) dichloride (6 mg, 0.008 mmol) and DMA (5 mL) were placed in a vial under N2. The mixture was heated at 110° C. for 2 h. After cooling down to room temperature, ice was added and then the aqueous was extracted with EtOAc (2×10 mL) and washed by water (5 mL×2) and brine (5 mL×2). The organic phase was concentrated and purified by column chromatography on silica gel to give 7-bromo-4-(1H-imidazol-1-yl)-2-phenylquinoline as a solid (37 mg) (MS: [M+1]+ 350).


Example 68: Synthesis of 1-(1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)urea (I-104)



embedded image


To a solution of 1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-amine (di TFA salt) (I-108, 23 mg, 0.041 mmol) in THF (3 mL) and triethylamine (0.03 mL, 0.212 mmol) was added (trimethylsilyl) isocyanate (8 mg, 0.071 mmol). The solution was stirred at rt for 3 h. Methanol (1 mL) was added and then the volatiles were removed by rotary evaporation. Water (3 mL) was added to the residue and the resulting solids were filtered and dried to afford 1-(1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)urea (12 mg) (MS: [M+1]+ 371).


Example 69: Synthesis of 2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)-3-phenylpropane-1-sulfonic acid (I-162)



embedded image


Step 1: 2-((tert-Butoxycarbonyl)amino)-3-phenylpropyl 4-methylbenzenesulfonate. A solution of tert-butyl (1-hydroxy-3-phenylpropan-2-yl)carbamate (1.25 g, 5.0 mmol) in pyridine (2 mL) was cooled to −10° C. Tosyl chloride (0.95 g, 5.0 mmol) in pyridine (2 mL) was added dropwise. The reaction was stirred at 0° C. for 1 h then warmed to rt and stirred for 16 h. The reaction was poured over ice, then extracted with (4:1) hexanes/Ethyl acetate (2×10 mL). The combined organics were dried (Na2SO4) and concentrated. The crude 2-((tert-butoxycarbonyl)amino)-3-phenylpropyl 4-methylbenzenesulfonate (MS: [M+1−Boc]+306) was taken onward directly.


Step 2: S-(2-((tert-Butoxycarbonyl)amino)-3-phenylpropyl) ethanethioate. 2-((tert-Butoxycarbonyl)amino)-3-phenylpropyl 4-methylbenzenesulfonate (1.6 g, 3.95 mmol) was placed in a flask with DMF (10 mL). Potassium thioacetate (677 mg, 5.93 mmol) in DMF (5 mL) was added to the flask then allowed to stir at rt for 16 h. Water (25 mL) was added and the organics were extracted into (4:1) hexanes/EtOAc (2×20 mL). The combined organics were dried (Na2SO4) and concentrated to afford S-(2-((tert-butoxycarbonyl)amino)-3-phenylpropyl) ethanethioate (MS: [M+1−Boc]+210).


Step 3: 2-Amino-3-phenylpropane-1-sulfonic acid hydrochloride. S-(2-((tert-Butoxycarbonyl)amino)-3-phenylpropyl) ethanethioate (309 mg, 1.0 mmol) was dissolved in formic acid (1.0 mL) and added dropwise at 0° C. to a solution of hydrogen peroxide (1 mL, 30% aq) in formic acid. The reaction was stirred at rt for 16 h. The volatiles were concentrated off to afford 2-amino-3-phenylpropane-1-sulfonic acid hydrochloride (MS: [M+1]+ 216).


Step 4: 2-((7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)-3-phenylpropane-1-sulfonic acid. 2-amino-3-phenylpropane-1-sulfonic acid hydrochloride (62 mg, 0.25 mmol) was placed in a vial with 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (30 mg, 0.1 mmol) in DMSO (0.2 mL) and N,N-diisopropylethylamine (0.1 mL). The solution is stirred at 95° C. for 16 h. After cooling down to room temperature, water (5 mL) was added and the organics were extracted into 10% methanol in dichloromethane (2×5 mL). The combined organics were dried (Na2SO4) and concentrated. The resulting residue was purified by silica chromatography using 0-25% methanol/dichloromethane to afford 2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)-3-phenylpropane-1-sulfonic acid (MS: [M+1]+ 477).


The following compounds are prepared essentially by the same method described above to prepare I-162:


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-163


embedded image




embedded image




embedded image


395









Example 70: Synthesis of 2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)-3-phenylpropane-1-sulfonamide (I-208)



embedded image


Step 1: 2-((tert-Butoxycarbonyl)amino)-3-phenylpropyl 4-methylbenzenesulfonate. A solution of tert-butyl (1-hydroxy-3-phenylpropan-2-yl)carbamate (1.25 g, 5.0 mmol) in pyridine (2 mL) was cooled to −10° C. Tosyl chloride (0.95 g, 5.0 mmol) in pyridine (2 mL) was added dropwise. The reaction was stirred at 0° C. for 1 h then warmed to rt and stirred for 16 h. The reaction was poured over ice, then extracted with (4:1) hexanes/Ethyl acetate (2×10 mL). The combined organics were dried (Na2SO4) and concentrated. The crude 2-((tert-butoxycarbonyl)amino)-3-phenylpropyl 4-methylbenzenesulfonate (MS: [M+1−Boc]+306) was taken onward directly.


Step 2: S-(2-((tert-Butoxycarbonyl)amino)-3-phenylpropyl) ethanethioate. 2-((tert-butoxycarbonyl)amino)-3-phenylpropyl 4-methylbenzenesulfonate (1.6 g, 3.95 mmol) was placed in a flask with DMF (10 mL). Potassium thioacetate (677 mg, 5.93 mmol) in DMF (5 mL) was added to the flask then allowed to stir at rt for 16 h. Water (25 mL) was added and the organics were extracted into (4:1) hexanes/EtOAc (2×20 mL). The combined organics were dried (Na2SO4) and concentrated to afford S-(2-((tert-butoxycarbonyl)amino)-3-phenylpropyl) ethanethioate (MS: [M+1−Boc]+210).


Step 3: tert-Butyl (1-(chlorosulfonyl)-3-phenylpropan-2-yl)carbamate. S-(2-((tert-Butoxycarbonyl)amino)-3-phenylpropyl) ethanethioate (300 mg, 0.97 mmol) was placed in a vial with acetonitrile (3 mL) and water (52 L) then cooled to 0° C. tert-butyl hypochlorite (0.33 mL, 2.9 mmol) was added and the reaction allowed to stir at 0° C. for 20 mins. The volatiles were removed by rotary evaporation and the crude tert-butyl (1-(chlorosulfonyl)-3-phenylpropan-2-yl)carbamate taken onward without further purification.


Step 4: tert-Butyl (1-phenyl-3-sulfamoylpropan-2-yl)carbamate. Tert-butyl (1-(chlorosulfonyl)-3-phenylpropan-2-yl)carbamate was placed in a vial with acetonitrile (5 mL) and cooled to 0° C. Ammonium hydroxide (2 mL) was added and the reaction was stirred at 0° C. for 10 minutes then warmed to rt and stirred for 1 h. Dichloromethane (25 mL) was added followed by water (10 mL) and the mixture stirred vigorously for 5 mins. The organics were separated, dried (Na2SO4) and concentrated to afford tert-butyl (1-phenyl-3-sulfamoylpropan-2-yl)carbamate (MS: [M+1−Boc]+215).


Step 5: 2-amino-3-phenylpropane-1-sulfonamide hydrochloride. tert-butyl (1-phenyl-3-sulfamoylpropan-2-yl)carbamate (100 mg, 0.32 mmol) was placed in a vial with dichloromethane (2 mL). Hydrochloric acid (4.0M in dioxane) (0.5 mL) was added and the reaction allowed to stir at rt for 1 h. Acetonitrile was added and the resulting solids were filtered off and vacuum dried to afford 2-amino-3-phenylpropane-1-sulfonamide hydrochloride (MS: [M+1]+ 215).


Step 6: 2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)-3-phenylpropane-1-sulfonamide. 2-amino-3-phenylpropane-1-sulfonamide hydrochloride (55 mg, 0.23 mmol) was placed in a vial with 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (30 mg, 0.1 mmol) in DMSO (0.2 mL) and N,N-diisopropylethylamine (0.1 mL). The solution is stirred at 95° C. for 16 h. After cooling down to room temperature, water (5 mL) was added and the organics were extracted into 10% methanol in dichloromethane (2×5 mL). The combined organics are dried (Na2SO4) and concentrated. The resulting residue was purified by Combiflash using 0-10% Methanol/Dichloromethane to afford 2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)-3-phenylpropane-1-sulfonamide (MS: [M+1]+ 476)


The following compounds are prepared essentially by the same method described above to prepare I-208:


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-209


embedded image




embedded image




embedded image


428.0









Example 71: Synthesis of 2-(benzyl(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)ethane-1-sulfonic acid (I-164)



embedded image


Step 1: Isopropyl ethenesulfonate. 2-Chloroethane-1-sulfonyl chloride (0.65 mL, 6.1 mmol) was placed in a flask with dichloromethane (3.0 mL) and isopropanol (0.47 mL, 6.1 mmol) and cooled to −10° C. A solution of pyridine (0.98 mL, 6.1 mmol) in dichloromethane (1.4 mL) was added dropwise and the reaction was allowed to stir at −10° C. for 2 h and warmed to rt over 30 mins. The mixture was quenched by the addition of 1M HCl (20 mL). The aqueous layer was extracted with ethyl acetate (2×10 mL). The combined extracts were dried (Na2SO4) and concentrated to afford isopropyl ethenesulfonate as an oil and was taken onward without further purification.


Step 2: 2-(Benzylamino)ethane-1-sulfonic acid hydrochloride. A solution of isopropyl ethenesulfonate (440 mg, 0.2 mmol) in methanol (1.5 mL) was added to a solution of benzyl amine (0.31 g, 2.9 mmol) in methanol (1.0 mL) at 0° C. The reaction was stirred at 0° C. for 1.5 h then warmed to rt. The solution was acidified by the addition of HCl (LOM in MeOH) then heated at 90° C. for 16 h. The resulting precipitate was filtered off and dried to afford 2-(benzylamino)ethane-1-sulfonic acid hydrochloride (MS: [M+1]+ 216).


Step 3: 2-(benzyl(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)ethane-1-sulfonic acid. 2-(Benzylamino)ethane-1-sulfonic acid hydrochloride (50 mg, 0.2 mmol) was placed in a vial with 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (30 mg, 0.1 mmol) in DMSO (0.2 mL) and N,N-diisopropylethylamine (0.1 mL). The solution is stirred at 95° C. for 16 h. After cooling down to room temperature, water (5 mL) was added and the organics were extracted into 10% methanol in dichloromethane (2×5 mL). The combined organics are dried (Na2SO4) and concentrated. The resulting residue was purified by Combiflash using 0-10% methanol/dichloromethane to afford 2-(benzyl(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)ethane-1-sulfonic acid (33 mg) (MS: [M+1]+ 477).


The following compounds are prepared essentially by the same method described above to prepare I-164:


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-165


embedded image




embedded image




embedded image


429









Example 72: Synthesis of N-(1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)-2-hydroxyacetamide (I-106)



embedded image


To a solution of 2-((1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)amino)-2-oxoethyl acetate (I-395) (17 mg, 0.04 mmol) in THF (2 mL) and water (0.5 mL) was added lithium hydroxide monohydrate (5 mg, 0.12 mmol). The solution was stirred at rt for 16 h. The volatiles were removed, and the residue was neutralized with 1M HCl (aq) to pH ˜7. The organics were extracted into ethyl acetate (2×5 mL). The combined organics were dried and concentrated to afford N-(1-(7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)piperidin-3-yl)-2-hydroxyacetamide (9 mg) (MS: [M+1]+ 386).


Example 73: Synthesis of (E)-5-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)methylene)thiazolidine-2,4-dione (I-276)



embedded image


Step 1: 7,8-Dichloro-4-(1H-imidazol-1-yl)quinoline-2-carbaldehyde. To a solution of 7,8-dichloro-4-(1H-imidazol-1-yl)-2-methylquinoline (104 mg) in dioxane (5 mL) was added selenium dioxide (82 mg). After the reaction mixture was stirred at 85° C. over 2 hours, a filtration through a Celite pad and evaporation under reduced pressure afforded the desired 7,8-dichloro-4-(1H-imidazol-1-yl)quinoline-2-carbaldehyde (90 mg) as a brown solid.


Step 2: (E)-5-((7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)methylene)thiazolidine-2,4-dione. The aldehyde (20 mg) was suspended in HOAc (1 mL) and treated with thiazolidine-2,4-dione (65 mg) and beta-analine (33 mg) at 100° C. over 4 hours. After removal of HOAc under reduced pressure, the residue was diluted with water (1 mL) and saturated NaHCO3 solution (1 mL). The solid was isolated by centrifuge and rinsed with water and 50% acetonitrile/water. Drying in vacuo afforded the desired product (24 mg), (E)-5-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)methylene)thiazolidine-2,4-dione, as a brown color solid (MS: [M+1]+ 391).


Example 74: Synthesis of 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)acrylic acid (I-423)



embedded image


Step 1: tert-Butyl 3-(4,7,8-trichloroquinolin-2-yl)acrylate. To a solution of 2,4,7,8-tetrachloroquinoline (1.3 g) in dry DMF (1 mL) were added tert-butyl acrylate (3.4 g), TEA (1.96 g) and PdCl2(PPh3)4 (0.34 g). The resultant mixture was stirred at 110° C. overnight and diluted with water. The aqueous layer was extracted with EtOAc (80 mL), and the isolated organic layer was rinsed with sat NH4Cl and dried over anhydrous Na2SO4. Silica gel column chromatography (eluting with a gradient of petroleum ether and EtOAc) afforded the desired product (1.4 g) (MS: [M+1]+ 358).


Step 2: tert-Butyl 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)acrylate. To a solution of tert-butyl 3-(4,7,8-trichloroquinolin-2-yl)acrylate (358 mg) in dioxane (5 mL) and water (2 mL) were added 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (580 mg), Na2CO3 (650 mg) and Pd(PPh3)4 (115 mg). The resultant mixture was purged with N2 via 3 cycles of vacuum and purging) and stirred at 120° C. over 4 hours under N2. After dilution with water (20 mL), the aqueous layer was extracted with EtOAc (30 mL). The isolated organic layer was washed with brine and dried over anhy. Na2SO4. A column chromatography gave the desired product (130 mg) (MS: [M+1]+ 390).


Step 3: 3-(7,8-Dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)acrylic acid. To a solution of tert-butyl 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)acrylate (15 mg) in DCM (0.4 mL) was added TFA (0.2 mL). The resultant solution was stirred over 1 hour and concentrated to dryness. The residue was purified by a preparative thin layer chromatography to afford the title compound (1.4 mg) (MS: [M+1]+334).


Example 75: Synthesis of 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propanoic acid (I-270)



embedded image


Step 1: tert-butyl 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propanoate. To a solution of tert-butyl 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)acrylate (60 mg) in EtOH (5 mL) were added LiCl (23 mg) and NaBH4 (9 mg) at 0° C. After stirring at 0° C. over 1 hour, the reaction mixture was quenched with 0.5 N HCl (0.5 mL). The mixture was concentrated to dryness to afford the crude of tert-butyl 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propanoate.


Step 2: 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propanoic acid. To a solution of tert-butyl 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propanoateresidue in DCM (2 mL) was added TFA (0.8 mL) and the reaction stirred at rt over 4 hours. The crude product was purified by preparative HPLC to afford the desired 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propanoic acid (19 mg). MS: [M+1]+ 336. 1H NMR (400 MHz, DMSO-D6): δ 8.193-8.171 (m, 3H), 7.735-7.712 (d, J=8.8 Hz, 1H), 7.626 (s, 1H), 3.216-3.252 (t, J=7.2 Hz, 2H), 2.863-2.899 (t, J=7.2 Hz, 2H) ppm.


Example 76: Synthesis of 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propan-1-ol (I-271)



embedded image


Step 1: Methyl 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propanoate. To a solution of tert-butyl 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propanoate (235 mg) in MeOH HCl (5 mL). The resultant solution was stirred at rt overnight. After concentration under reduced pressure, the residue was purified by preparative thin layer chromatography to afford the title compound (150 mg) as a powder (MS: [M+1]+ 322).


Step 2: 3-(7,8-Dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propan-1-ol. To a solution of methyl 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propanoate (100 mg) in anhydrous THF (4 mL) was added DIABL-H (1.1 mL, 1M in THF) at −60° C. After stirring over one hour at 60° C., the reaction mixture was slowly warmed up to room temperature, quenched by adding MeOH (1 mL) and diluted with EtOAc (25 mL). The combined organic layers were washed with brine and dried over anhydrous Na2SO4. Concentration under reduced pressure and purification by preparative HPLC afforded the title compound (6 mg). MS: [M+1]+ 322. 1H NMR (400 MHz, DMSO-D6): δ 8.195-8.173 (m, 3H), 7.735-7.712 (d, J=8.8 Hz, 1H), 7.594 (s, 1H), 3.520-3.536 (m, 2H), 2.996-3.034 (t, J=7.6 Hz, 2H), 1.965-2.002 (m, 2H) ppm.


Example 77: Synthesis of 7-chloro-4-(1H-imidazol-1-yl)-8-methylquinoline (I-274)



embedded image


Step 1: tert-Butyl 4-(7,8-dichloro-2-(3-methoxy-3-oxopropyl)quinolin-4-yl)-1H-pyrazole-1-carboxylate. To a solution of methyl 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propanoate (30 mg) in DMF (1 mL) were added DMAP (12 mg), (BOC)2O (28 mg) and TEA (35 mg) under N2. After stirring over 2 h at room temperature, the reaction mixture was diluted with water (10 mL) and extracted with EtOAc (25 mL). The separated organic layer was washed with saturated NH4Cl and concentrated to dryness. The residual was purified by preparative thin layer chromatography to afford the title compound (33 mg) as a solid. (MS: [M+1]+ 450)


Step 2: tert-Butyl 4-(7,8-dichloro-2-(3-oxopropyl)quinolin-4-yl)-1H-pyrazole-1-carboxylate. To a solution of tert-butyl 4-(7,8-dichloro-2-(3-methoxy-3-oxopropyl)quinolin-4-yl)-1H-pyrazole-1-carboxylate (190 mg) in DCM (2 mL) was added DIBAL-H (0.56 mL) at −60° C. The reaction mixture was stirred and slowly warmed to room temperature in 2 hours. The reaction was quenched with MeOH (1 mL) and water (100 mL). Extraction with EtOAc, washing with brine and purification by preparative thin layer chromatography afforded the title compound (57 mg) as a solid. (MS: [M+1]+ 420)


Step 3 and Step 4: (E)-5-(3-(7,8-Dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propylidene)thiazolidine-2,4-dione. To a solution of tert-butyl 4-(7,8-dichloro-2-(3-oxopropyl)quinolin-4-yl)-1H-pyrazole-1-carboxylate (30 mg) in EtOH was added thiazolidine-2,4-dione (18 mg) and piperidine (8 mg). The reaction mixture was stirred at 85° C. for 1 hour. After concentration under reduced pressure, the residue was diluted EtOAc (10 mL) and washed with water (10 mL). After removal of organic solvents, the crude intermediate was treated with TFA (0.3 mL) in DCM (0.9 mL) over 2 hour. After concentrated under reduced pressure, the crude product was purified by preparative HPLC to afford the title compound (7.3 mg) as a powder. MS: [M+1]+ 419.1. 1H NMR (400 MHz, DMSO-D6): δ 8.186-8.209 (d, J=9.2 Hz, 1H), 8.168 (s, 1H), 7.733-7.756 (d, J=9.2 Hz, 1H), 7.636 (s, 1H), 7.073-7.111 (t, J=7.6 Hz, 1H), 3.3 (m, 2H), 2.768-2.822 (dd, J=14.4 and 7.2 Hz, 2H) ppm.


Example 78: Synthesis of 5-(3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propyl)thiazolidine-2,4-dione (I-272)



embedded image


Crude (E)-5-(3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propylidene)thiazolidine-2,4-dione (40 mg) in DMF (1 mL) was treated with NaBH4 (7 mg) and LiCl (5 mg) at 0° C. for 3 hours. The product was purified by preparative HPLC to afford the title product (4.5 mg). MS: [M+1]+ 421.0. 1H NMR (400 MHz, CD3OD): δ 8.202-8.224 (d, J=9.2 Hz, 1H), 8.143 (s, 1H), 7.733-7.710 (d, J=9.2 Hz, 1H), 7.643-7.650 (d, J=2.8 Hz, 1H), 4.532-7.552 (d, J=8 Hz, 1H), 3.154-3.171 (m, 2H), 2.259-2.291 (m, 2H) and 2.014-2.062 (m, 2H) ppm.


Example 79: Synthesis of 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)cyclohex-3-ene-1-carboxylic acid (I-273)



embedded image


Step 1: Ethyl 3-(4,7,8-trichloroquinolin-2-yl)cyclohex-3-ene-1-carboxylate. Ethyl 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)cyclohex-3-ene-1-carboxylate was synthesized according to WO2020/112706. To a mixture of 2,4,7,8-tetrachloroquinoline, ethyl 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)cyclohex-3-ene-1-carboxylate (50 mg, 0.19 mmol), Na2CO3 (53 mg, 0.38 mmol), Pd(Ph3P)4 (23 mg, 0.02 mmol) in dioxane/H2O (5/1, 5 mL) was purged with N2 via 3 cycles of vacuum/purging. Then the reaction mixture was stirred at 100° C. overnight. After cooling to rt, the reaction mixture was taken up with EtOAc, washed with brine, and concentrated under reduced pressure. The crude was purified by prep-TLC with pet. ether/DCM (1/1) to afford the desired product of ethyl 3-(4,7,8-trichloroquinolin-2-yl)cyclohex-3-ene-1-carboxylate (36 mg) as light brown semi-solid. MS (ES+): [M+1]+, m/z 384.1, 386.1


Step 2: Ethyl 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)cyclohex-3-ene-1-carboxylate. To a mixture of ethyl 3-(4,7,8-trichloroquinolin-2-yl)cyclohex-3-ene-1-carboxylate, 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (36 mg, 0.093 mmol), Na2CO3 (38.6 mg, 0.28 mmol), Pd(Ph3P)4 (10.7 mg, 0.009 mmol) dioxane/H2O (2/1, 15 mL) was purged with N2 via 3 cycles of vacuum/purging. Then the reaction was stirred at 100° C. overnight. After cooling to rt, it was taken up with EtOAc, washed with brine, concentrated. The crude was purified by prep-TLC with pet. ether/DCM (1/1) to afford the desired product of ethyl 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)cyclohex-3-ene-1-carboxylate (5.1 mg) as an off-white solid. MS: (ES+): [M+1]+m/z 416.1, 418.1


Step 3: 3-(7,8-Dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)cyclohex-3-ene-1-carboxylic acid. To a mixture of ethyl 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)cyclohex-3-ene-1-carboxylate (10 mg, 0.024 mmol) in CH3OH/H2O/THF (1/1/1, 3 mL) was LiOH (6 mg). The reaction mixture was stirred at rt overnight. After quenched with aq. HCl, it was taken up with DCM/iPrOH, washed with brine, and concentrated. The crude was purified by prep-HPLC to afford the desired product of 3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)cyclohex-3-ene-1-carboxylic acid (3.4 mg) as an off-white solid (MS: (ES+): m/z 388.1 [M+1]+). 1H NMR (400 MHz, CD3OD, ppm): δ 8.38 (s, 1H), 8.00 (d, J=9.2 Hz, 1H), 7.70 (s, 1H), 7.56 (d, J=9.2 Hz, 1H), 5.89 (s, 1H), 2.90 (s, 1H), 2.71-2.63 (m, 2H), 2.42 (s, 2H), 2.01-1.94 (s, 2H).


Example 80: Synthesis of (3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propyl)glycine (I-336)



embedded image


Step 1: tert-Butyl 4-(2-(3-((2-(tert-butoxy)-2-oxoethyl)amino)propyl)-7,8-dichloroquinolin-4-yl)-1H-pyrazole-1-carboxylate. To a solution of tert-butyl 4-(7,8-dichloro-2-(3-oxopropyl)quinolin-4-yl)-1H-pyrazole-1-carboxylate (100 mg) and tert-butyl glycinate (62 mg) in DCM (0.5 mL) and EtOH (2.5 mL) were added NaBH3CN (100 mg) and acetic acid (1 drop). After stirring overnight, quenching with water, extraction with DCM, and concentration under reduced pressure, the residue was purified by a preparative thin layer chromatography (eluting with 15% MeOH in DCM) to afford the title compound (58 mg). MS: [M+1]+ 479.


Step 2: (3-(7,8-Dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propyl)glycine. To a solution of tert-butyl 4-(2-(3-((2-(tert-butoxy)-2-oxoethyl)amino)propyl)-7,8-dichloroquinolin-4-yl)-1H-pyrazole-1-carboxylate (15 mg) in DCM (3 mL) was added TFA (0.5 mL). The resultant solution was stirred over 6 hours at room temperature. After evaporation under reduced pressure, the residue was purified by preparative HPLC to afford the tile compound (6.4 mg) (MS: [M+1]+ 379.1). 1H NMR (400 MHz, CD3OD): δ 8.179-8.202 (d, J=9.2 Hz, 1H), 8.140 (s, 1H), 7.690-7.713 (d, J=9.2 Hz, 1H), 7.596 (s, 1H), 4.019 (S, 2H), 3.324-3.365 (m, 4H), 2.338-2.391 (m, 2H) ppm


Example 81: Synthesis of N-(3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propyl)-N-(methylsulfonyl) glycine (I-337)



embedded image


Step 1: tert-Butyl 4-(2-(3-(N-(2-(tert-butoxy)-2-oxoethyl)methylsulfonamido)propyl)-7,8-dichloroquinolin-4-yl)-1H-pyrazole-1-carboxylate. A solution of tert-butyl 4-(2-(3-((2-(tert-butoxy)-2-oxoethyl)amino)propyl)-7,8-dichloroquinolin-4-yl)-1H-pyrazole-1-carboxylate (15 mg) in DCM (0.5 mL) were added MsCl (6.4 mg) and TEA (14.8 mg). After stirring at room temperature over 1 hour, aqueous work up with DCM, washing with brine, condensation under reduced pressure, and purification by preparative thin layer chromatography afforded the title compound (8 mg). MS: [M+1]+ 613.


Step 2: N-(3-(7,8-Dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propyl)-N-(methylsulfonyl) glycine. To a solution of N-(3-(7,8-dichloro-4-(1H-pyrazol-4-yl)quinolin-2-yl)propyl)-N-(methylsulfonyl) glycine (8 mg) in DCM (5 mL) was added TFA (1 mL). The reaction mixture was stirred over 5 hours at room temperature. Evaporation under reduced pressure and purification by preparative HPLC afforded the title compound (3 mg) as a powder (MS: [M+1]+ 457.1). 1H NMR (400 MHz, DMSO-D6): δ 8.192-8.215 (d, J=9.2 Hz, 1H), 8.140 (s, 1H), 7.697-7.720 (d, J=9.2 Hz, 1H), 7.664 (s, 1H), 4.149 (S, 2H), 3.468-3.503 (t, J=7.2 Hz, 2H), 3.160-3.196 (t, J=7.2 Hz, 2H), 3.011 (S, 3H), 2.202-2.237 (m, 2H) ppm.


Example 82: Synthesis of (2S,4S)-4-(carboxymethoxy)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidine-2-carboxylic acid (I-569)



embedded image


Step 1: To a mixture of NaH (6 mg, 0.15 mmol), tert-butyl 2-bromoacetate (80 uL, 0.4 mmol) and TBAI (5 mg, 0.01 mmol) and THF (1 mL) was added a solution of methyl (2S,4S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)naphthalen-2-yl)-4-hydroxypyrrolidine-2-carboxylate (41 mg, 0.1 mmol) in THF (1 mL) at 0° C. The resulting mixture was stirred 2 h at room temperature and quenched by NH4Cl (aq, sat., 1 mL). The crude was diluted by EtOAc and the organic phase was washed with water, brine and dried over anhy. Na2SO4. A column chromatography eluting with a gradient of hexanes and EtOAc afforded the desired product (25 mg) as white solids (MS: [M+1]+ 520.1).


Step 2: methyl (2S,4S)-4-(2-(tert-butoxy)-2-oxoethoxy)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)naphthalen-2-yl)pyrrolidine-2-carboxylate (25 mg, 0.05 mmol) was placed in a vial with THF (1.2 mL), methanol (0.4 mL), and water (0.4 mL). Lithium hydroxide monohydrate (8 mg, 0.18 mmol) was added and the reaction was allowed to stir at r.t. for 16 h. The volatiles were concentrated off and the resulting residue neutralized by the addition of 1 N hydrochloric acid. The resulting solution was lyophilized to afford the titled compound (MS: [M+1]+ 450.1).


Synthesis of (2S,4S)-4-(((carboxymethyl)carbamoyl)oxy)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)naphthalen-2-yl)pyrrolidine-2-carboxylic acid (I-570)



embedded image


Step 1: To a solution of methyl (2S,4S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)naphthalen-2-yl)-4-hydroxypyrrolidine-2-carboxylate (40 mg, 0.1 mmol) in THF (1 mL) was added CDI (32 mg, 0.2 mmol). After 1 h, a solution of tert-butyl glycinate (25 mg, 0.15 mmol) and DIPEA (52 uL, 0.3 mmol) in THF (1 mL) was added. The mixture was stirred at 60° C. overnight. After cooling down to r.t., the solvent was evaporated. The crude was dissolved in EtOAc (5 mL) and washed with water, brine and dried over anhy. Na2SO4. A column chromatography eluting with a gradient of hexanes and EtOAc afforded the desired product (40 mg) as white solids (MS: [M+1]+ 563.1).


Step 2: methyl (2S,4S)-4-(((2-(tert-butoxy)-2-oxoethyl)carbamoyl)oxy)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)naphthalen-2-yl)pyrrolidine-2-carboxylate (40 mg, 0.07 mmol) was placed in a vial with THF (1.2 mL), methanol (0.4 mL), and water (0.4 mL). Lithium hydroxide monohydrate (8 mg, 0.18 mmol) was added, and the reaction was allowed to stir at r.t. for 16 h. The volatiles were concentrated off and the resulting residue neutralized by the addition of 1 N hydrochloric acid. The resulting solution was lyophilized to afford the titled compound (MS: [M+1]+ 493.1).


The following compounds were prepared essentially by the same method described above to prepare I-570.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-571


embedded image




embedded image




embedded image


506









Example 83: Synthesis of methyl (S)-2-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)naphthalen-2-yl)pyrrolidin-2-yl)acetate (I-572)



embedded image


At 0° C., oxalyl chloride (60 uL, 0.5 mmol) or thionyl chloride was added dropwise to MeOH (1 mL). After 30 min, a solution of (S)-2-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)naphthalen-2-yl)pyrrolidin-2-yl)acetic acid (40 mg, 0.1 mmol) in MeOH (0.1 mL) was added. The solution was stirred at r.t. overnight. After evaporation, the crude was dissolved in EtOAc (5 mL) and washed with saturated NaHCO3 (aq.), water, brine and dried over anhy. Na2SO4. A column chromatography eluting with a gradient of hexanes and EtOAc afforded the desired product (40 mg) as white solids (MS: [M+1]+ 404.1).


The following compounds were prepared essentially by the same method described above to prepare I-572.




















MS


I-#
Starting Material

Structure
[M + 1]+







I-573


embedded image




embedded image




embedded image


447





I-574


embedded image


MeOH


embedded image


405





I-575


embedded image


MeOH


embedded image


409





I-576


embedded image


MeOH


embedded image


403





I-577


embedded image


MeOH


embedded image


403





I-578


embedded image


MeOH


embedded image


405





I-579


embedded image


MeOH


embedded image


403





I-580


embedded image


MeOH


embedded image


417





I-581


embedded image


MeOH


embedded image


439





I-582


embedded image


MeOH


embedded image


417





I-583


embedded image


MeOH


embedded image


419





I-584


embedded image


MeOH


embedded image


403





I-585


embedded image


MeOH


embedded image


405





I-586


embedded image


MeOH


embedded image


405





I-587


embedded image


MeOH


embedded image


419





I-588


embedded image


MeOH


embedded image


439





I-589


embedded image


MeOH


embedded image


406





I-590


embedded image


MeOH


embedded image


405





I-591


embedded image


MeOH


embedded image


403





I-592


embedded image


MeOH


embedded image


409





I-593


embedded image


MeOH


embedded image


391





I-594


embedded image


CH3CH2OH


embedded image


405





I-595


embedded image




embedded image




embedded image


419





I-596


embedded image


CH3OH


embedded image


407





I-597


embedded image


CH3OH


embedded image


407





I-598


embedded image


CH3OH


embedded image


407





I-599


embedded image


CH3CH2OH


embedded image


421





I-600


embedded image




embedded image




embedded image


435





I-601


embedded image




embedded image




embedded image


449





I-602


embedded image




embedded image




embedded image


449





I-662


embedded image


MeOH


embedded image


421





I-663


embedded image




embedded image




embedded image


543









Example 84: Synthesis of methyl (S)-2-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)naphthalen-2-yl)azetidin-2-yl)acetate (I-604)



embedded image


Step 1: At 0° C., oxalyl chloride (0.1 mL) was added dropwise to MeOH (1 mL). After 30 min, a solution of (S)-2-(1-(tert-butoxycarbonyl)azetidin-2-yl)acetic acid (43 mg, 0.2 mmol) in MeOH (0.1 mL) was added. The solution was stirred at r.t. overnight. After evaporation, the crude was dissolved in EtOAc (5 mL) and washed with saturated NaHCO3 (aq.), water, brine and dried over anhy. Na2SO4. The crude was used in the next step without further purification. (MS: [M+1]+ 230.1).


Step 2: A solution of tert-butyl (S)-2-(2-methoxy-2-oxoethyl)azetidine-1-carboxylate (50 mg crude, 0.2 mmol) in 50% TFA/DCM was stirred at r.t. for 30 min. After evaporation, the crude was dissolved in DCM (2 mL). The solvent was removed by evaporation. The process was repeated twice. The crude was used in the next step without further purification. (MS: [M+1]+ 130.1).


Step 3: I-604 was prepared essentially by the same method as I-353. (MS: [M+1]+ 390.1).


Example 85: Synthesis of (S)-5-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)-1,2,4-oxadiazol-3(2H)-one (I-605)



embedded image


To a mixture of (S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidine-2-carboxamide (100 mg, 0.27 mmol) and DCE (2 mL) was added (COCl)2 (35 uL, 0.4 mmol) at r.t. The mixture was stirred at 70° C. overnight and then TMSN3 (1 mL) was added. The solution was stirred at 80° C. for 2 days. After cooling down to r.t., the crude was purified directly by a column chromatography on silica gel to give the titled product as a solid (MS: [M+1]+ 417.1).


Example 86: Synthesis of (2S,4R)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-4-((morpholine-4-carbonyl)oxy)pyrrolidine-2-carboxylic acid (I-606)



embedded image


embedded image


Step 1: methyl (2S,4R)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-4-hydroxypyrrolidine-2-carboxylate. To a vial were added 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (90 mg, 0.302 mmol), methyl (2S,4R)-4-hydroxypyrrolidine-2-carboxylate (146 mg, 0.603 mmol), DMSO (0.20 mL), and N,N-diisopropylethylamine (0.10 mL). The resulting reaction mixture was stirred at 90° C. for 16 h and cooled to room temperature, followed by adding H2O (20 mL). The cloudy mixture was centrifuged and the residue was extracted with 10% MeOH/DCM (2×10 mL). The organic phase was washed with H2O (10 mL), brine (10 mL), dried over Na2SO4, and concentrated. The crude residue was purified by silica chromatography to afford (104 mg) (MS: [M+1]+ 407).


Step 2: (3R,5S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-(methoxycarbonyl)pyrrolidin-3-yl 1H-imidazole-1-carboxylate. To a vial were added methyl (2S,4R)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-4-hydroxypyrrolidine-2-carboxylate (100 mg, 0.246 mmol), THF (5 mL) and carbonyldiimidazole (CDI) (80 mg, 0.493 mmol). The resulting mixture was stirred at room temperature for 4 hrs. All volatile solvents were then removed under reduced pressure and the residue was dried at high vacuum to afford the titled product. The crude was used in next step.


Step 3: (3R,5S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-(methoxycarbonyl)pyrrolidin-3-yl morpholine-4-carboxylate. To a vial were added (3R,5S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-(methoxycarbonyl)pyrrolidin-3-yl 1H-imidazole-1-carboxylate (125 mg, 0.250 mmol), THF (2 mL), and morpholine (44 mg, 0.505 mmol). The resulting reaction mixture was stirred at rt for 3 h. The volatiles were concentrated off. The reaction mixture was diluted by ethyl acetate (15 mL). The organic phase was washed by H2O (3×5 mL), brine (5 mL), and dried over Na2SO4. After concentration, the crude was purified by silica gel chromatography to afford the titled product as an off-white solid (60 mg) (MS: [M+1]+ 520).


Step 4: (2S,4R)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-4-((morpholine-4-carbonyl)oxy)pyrrolidine-2-carboxylic acid. To a vial were added (3R,5S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-(methoxycarbonyl)pyrrolidin-3-yl morpholine-4-carboxylate (50 mg, 0.096 mmol), THF (0.8 mL), methanol (0.2 mL), water (0.2 mL) and morpholine. Lithium hydroxide monohydrate (12 mg, 0.288 mmol) was added and the resulting reaction mixture was stirred at rt for 16 h. The volatiles were concentrated off. The reaction mixture was diluted by water (2 mL) then acidified with 1M HCl to afford the titled product as an off-white solid (38 mg) (MS: [M+1]+ 506).


The following compound was synthesized in a similar manner as I-606:


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-607


embedded image




embedded image




embedded image


494









Example 87: Synthesis of 2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)-N-(2-hydroxyethyl)ethane-1-sulfonamide (I-624)



embedded image


Step 1: I-159 was heated in the excess of thionyl chloride at 60° C. over 4 hours. The resultant solution was concentrated under vacuum to afford 2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)ethane-1-sulfonyl chloride, which was used in the following step without further purification.


Step 2: 2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)ethane-1-sulfonyl chloride was mixed with 2-aminoethan-1-ol in pyridine. Aqueous work-up and purification by prep-HPLC afforded the titled compound (I-624). MS [M+1]+ 444.


Following the preparation of I-624 described above, the following compounds were prepared.


















MS


I-#
Starting Materials
Structure
[M + 1]+



















I-625


embedded image




embedded image




embedded image


512





I-626


embedded image




embedded image




embedded image


498





I-627


embedded image




embedded image




embedded image


498





I-628


embedded image




embedded image




embedded image


526





I-629


embedded image




embedded image




embedded image


512





I-630


embedded image




embedded image




embedded image


472





I-631


embedded image




embedded image




embedded image


512





I-632


embedded image




embedded image




embedded image


458





I-633


embedded image




embedded image




embedded image


458









Example 88: Synthesis of 3-(N-(2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)ethyl)methylsulfonamido) propanoic acid (I-634)



embedded image


A mixture of N-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-N1-methylethane-1,2-diamine (100 mg, 0.3 mmol), tert-butyl acrylate (41.9 mg, 0.33 mmol) and DBU (135 mg, 0.9 mmol) in DMF was stirred at 40° C. until completion by LCMS. After cooling to 0° C., MsCl (103 mg, 0.9 mmol) was added and the mixture was stirred for another 3 h, monitored by TLC. The mixture was then quenched with aq.NaHCO3, taken up with DCM, washed with brine, dried, concentrated, and purified by prep-TLC to afford tert-butyl 3-(N-(2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)ethyl)methylsulfonamido)propanoate which was dissolved in DCM/TFA (1/1, 2 mL) and stirred for 2 h, monitored by TLC. The solvent was then removed under vacuum and the residue was purified by prep-HPLC to the titled product (I-634) as a light yellow solid. 1H NMR (400 MHz, CD3OH, ppm): δ 9.22 (s, 1H), 7.98 (s, 1H), 7.79 (s, 1H), 7.41-7.33 (m, 3H), 4.05 (s, 2H), 3.59-3.58 (m, 4H), 3.31 (s, 3H), 2.92 (s, 3H), 2.65 (s, 2H). LCMS: (ES+): m/z 486.1 [M+1]+.


Example 89: Synthesis of N-(2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)ethyl)-N-(methylsulfonyl)glycine (I-635)



embedded image


To a mixture of N-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-N1-methylethane-1,2-diamine (100 mg, 0.3 mmol) and ethyl 2-oxoacetate (74 mg, ˜50% in toluene, 0.36 mmol) in DCM/DCM/CH3OH (2/1, 3 mL) and cat. AcOH (2 drops) was added NaBH3CN (38 mg, 0.6 mmol) at rt. and stirred for 10 h until completion by LCMS. The mixture was then quenched with aq. NaHCO3, taken up with DCM, washed with brine, dried, and concentrated to afford crude desired product of ethyl 2-oxoacetate, which was used into next step without further purification. The residue was dissolved into DCM/TEA (5/1, 5 mL), then MsCl (150 μL) was added into the mixture at rt, and stirred for another 2 h, monitored by TLC. The mixture was then quenched with aq. NH4Cl, taken up with DCM, washed with brine, dried, and concentrated. The residue was purified by prep-TLC to afford ethyl N-(2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)ethyl)-N-(methylsulfonyl)glycinate which was dissolved in CH3OH/H2O (2/1, 3 mL), added LiOH, and stirred for 1 h, monitored by TLC. The reaction was then quenched with TFA, concentrated, and purified by prep-TLC (DCM/CH3OH=8/1) to afford the titled product (I-635) as a light yellow solid. 1H NMR (400 MHz, CD3OD, ppm): δ 8.09 (s, 1H), 7.56 (s, 1H), 7.36-7.20 (m, 4H), 4.02-3.97 (m, 4H), 3.65 (br, 2H), 3.31 (s, 3H), 3.02 (s, 3H). LCMS: (ES+): m/z 473.1 [M+1]+.


Example 90: Synthesis of 3-((N-(2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(methyl)amino)ethyl)methylsulfonamido) methyl)benzoic acid (I-636)



embedded image


I-636 was prepared according to the same method to prepare I-635. 1H NMR (400 MHz, CD3OD, ppm): δ 9.20 (s, 1H), 8.00 (s, 1H), 7.81 (s, 1H), 7.64 (s, 1H), 757-7.47 (m, 2H), 7.34 (d, J=8.8 Hz, 1H), 7.25 (d, J=8.8 Hz, 1H), 4.33 (s, 2H), 3.84-3.72 (m, 4H), 3.06 (m, 3H), 3.02 (s, 3H). LCMS: (ES+): m/z 548.1 [M+1]+.


Example 91: Synthesis of methyl (2S,3S)-3-acetoxy-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidine-2-carboxylate (I-642) and (2S,3S)-3-acetoxy-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidine-2-carboxylic acid (I-643)

I-642 and I-643 were prepared according to the follow synthetic scheme:




embedded image


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-642


embedded image




embedded image




embedded image


449





I-643


embedded image




embedded image




embedded image


435









Example 92: Synthesis of tert-butyl (2S,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidine-2-carboxylate (I-645)



embedded image


Step 1: (2S,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidine-2-carboxylic acid was prepared similar to the preparation of I-123, step 1.


Step 2: The mixture of (2S,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidine-2-carboxylic acid (380 mg, 0.083 mmol) and 1,1-di-tert-butoxy-N,N-dimethylmethanamine (200 mg, 0.983 mmol) was stirred at 80° C. overnight. The crude was purified by silica gel chromatography to afford I-645 (MS: [M+1]+ 449)


Example 93: Synthesis of methyl (2S,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-methoxypyrrolidine-2-carboxylate (I-646)



embedded image


Step 1: (2S,3S)-methyl 3-hydroxypyrrolidine-2-carboxylate HCl salt was prepared similar to the preparation of I-593.


Step 2: To a solution of (2S,3S)-3-hydroxypyrrolidine-2-carboxylic acid HCl salt (268 mg, 1.47 mmol) in DCN (6 mL) was added Boc2O (385 mg, 1.76 mmol) and TEA (0.612 mL, 4.39 mmol). The resulting reaction solution was stirred overnight. The mixture was diluted with DCM (20 mL), washed with H2O (2×10 mL), brine (10 mL), and dried over Na2SO4. After concentration, the crude (2S,3S)-1-tert-butyl 2-methyl 3-hydroxypyrrolidine-1,2-dicarboxylate was used in next step.


Step 3: To a solution of (2S,3S)-1-tert-butyl 2-methyl 3-hydroxypyrrolidine-1,2-dicarboxylate (0.5 mmol) and iodomethane (0.093 mL, 1.5 mmol) in DCM (1.5 mL) was added 700 mg of Ag2O (700 mg, 3.02 mmol). After being stirred overnight, the mixture was filtered through a pad of Celite and purified by silica gel chromatography to afford (2S,3S)-1-tert-butyl 2-methyl 3-methoxypyrrolidine-1,2-dicarboxylate as an oil.


Step 4: To a solution of (2S,3S)-1-tert-butyl 2-methyl 3-methoxypyrrolidine-1,2-dicarboxylate (56.5 mg, 0.218 mmol) in DCM (0.5 mL) was added TFA (0.25 mL). After being stirred for 4 hours, the mixture was concentrated to remove all volatiles. The residue (2S,3S)-methyl 3-methoxypyrrolidine-2-carboxylate was used in next step.


Step 5: (2S,3S)-methyl 1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-methoxypyrrolidine-2-carboxylate was prepare by a procedure similar to that of I-123, step 1. (MS: [M+1]+ 421).


The following compounds were prepared essentially by the same method to prepare I-646.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-647


embedded image




embedded image




embedded image


421





I-648


embedded image




embedded image

embedded image



embedded image


465









Example 94: Synthesis of methyl (2R,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-fluoropyrrolidine-2-carboxylate (I-649)



embedded image


To a mixture of methyl (2S,3R)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidine-2-carboxylate (40.7 mg, 0.1 mmol) in DCM (0.5 mL) was added a solution of DAST (0.040 mL, 0.30 mmol) in DCM (0.2 mL) at −10° C. The mixture was stirred at −10° C. for 2 hours. After aqueous work-up, the crude was purified by PTLC to afford the titled product as a white solid (MS: [M+1]+ 409).


The following compounds were prepared essentially by the same method to prepare I-649.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-650


embedded image


DAST


embedded image


409





I-651


embedded image


DAST


embedded image


395









Example 95: Synthesis of methyl 1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-4,5-dihydro-1H-pyrrole-2-carboxylate (I-652)



embedded image


To the solution of methyl (2R,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-fluoropyrrolidine-2-carboxylate (10 mg, 0.0244 mmol) in methanol (0.5 mL) was added a solution of LiOH—H2O (1.57 mg, 0.0366 mmol) in H2O (0.10 mL) with stirring. After 5 min, the reaction was quenched with HOAc (0.00209 mL, 0.0366 mmol) in H2O (0.10 mL). The resulting mixture was purified by HPLC to afford the titled product as a white solid. (MS: [M+1]+ 389).


Example 96: Synthesis of methyl (2S,3R)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-(((2-methoxy-2-oxoethyl)carbamoyl)oxy)pyrrolidine-2-carboxylate (I-653)



embedded image


To a solution of methyl (2S,3R)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidine-2-carboxylate (30 mg, 0.0737 mmol) in THF (0.5 mL) was added TEA (0.051 mL, 0.369 mmol) and CDI (40 mg, 0.247 mmol). The resulting mixture was stirred for 2 hours. Methyl glycinate HCl salt (18.5 mg, 0.147 mmol) was then added. After 2 hours of stirring, the mixture was diluted with DCM (20 mL), washed with H2O (2×10 mL), brine (10 mL), and dried over Na2SO4. The crude was purified by silica gel chromatography to afford the title product as a white solid. (MS: [M+1]+ 522).


Example 97: Synthesis of(S)-1-((S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)-2,2,2-trifluoroethan-1-ol (I-654) and (R)-1-((S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)-2,2,2-trifluoroethan-1-ol (I-655)



embedded image


Step 1: To a flask were added tert-butyl (S)-2-formylpyrrolidine-1-carboxylate (1.992 g, 10 mmol) and THF (25 mL). The solution was cooled in ice bath for 20 min. After the slow addition of a solution of trimethyl(trifluoromethyl)silane in THF (2.0 M, 7.5 mL, 15 mmol) at 0° C., a solution of TBAF in THF (1.0 M, 0.5 mL, 0.5 mmol) was added dropwise. The resultant mixture was stirred at the same temperature for 2 hours. More TBAF solution in THF (1.0 M, 2.0 mL, 2.0 mmol) was added and the mixture was stirred at room temperature for another 2 hours. After being quenched by saturated NH4Cl and concentrated, the resultant mixture was diluted with DCM (150 mL), washed with H2O, brine, dried over Na2SO4 and then concentrated. The crude product was purified by silica gel chromatography to afford tert-butyl (S)-2-((S)-2,2,2-trifluoro-1-hydroxyethyl)pyrrolidine-1-carboxylate and tert-butyl (S)-2-((R)-2,2,2-trifluoro-1-hydroxyethyl)pyrrolidine-1-carboxylate.


Step 2: To a vial were added tert-butyl (S)-2-((S)-2,2,2-trifluoro-1-hydroxyethyl)pyrrolidine-1-carboxylate (50 mg, 0.186 mmol), DCM (0.5 mL) and TFA (0.25 mL). The resulting solution was stirred for 2 hours. After removing all volatiles under vacuo, the crude (S)-2,2,2-trifluoro-1-((S)-pyrrolidin-2-yl)ethan-1-ol was used directly in next step. The same procedure was used to prepare the (R, S)-isomer.


Step 3: Both of (S)-1-((S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)-2,2,2-trifluoroethan-1-ol I-654 (MS: [M+1]+ 431) and (R)-1-((S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)-2,2,2-trifluoroethan-1-ol I-655 (MS: [M+1]+ 431) were prepared following the synthetic procedure to prepare I-123, step 1.


Example 98: Synthesis of (S)-2-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)-1,1,1,3,3,3-hexafluoropropan-2-ol (I-656)



embedded image


Step 1: To a vial were added DCM (3 mL) and oxalyl chloride (0.220 mL, 2.6 mmol) followed being cooled in dry ice/acetone bath. A solution of DMSO (0.383 mL, 5.4 mmol) in DCM (2.5 mL) was added dropwise. The mixture was stirred at −78° C. for 30 min. Then a solution of tert-butyl (S)-2-((R)-2,2,2-trifluoro-1-hydroxyethyl)pyrrolidine-1-carboxylate (582 mg, 2.16 mmol) in DCM (2.5 mL) was added dropwise at same temperature. After 30 min, TEA (1.5 mL, 10.8 mmol) was added dropwise and the mixture was stirred at −78° C. for 30 min. The resulting mixture was stirred at room temperature for 1 hour, then quenched by adding H2O (10 mL). The aqueous phase was extracted by DCM (2×15 mL). The combined organic phase was concentrated, purified by silica gel chromatography to afforded tert-butyl (S)-2-(2,2,2-trifluoroacetyl)pyrrolidine-1-carboxylate.


Step 2: tert-butyl (S)-2-(1,1,1,3,3,3-hexafluoro-2-hydroxypropan-2-yl)pyrrolidine-1-carboxylate was prepared similar to the procedure to prepare I-654, step 1.


Step 3: (S)-1,1,1,3,3,3-hexafluoro-2-(pyrrolidin-2-yl)propan-2-ol was prepared similar to the procedure to prepare I-654, step 2.


Step 4: (S)-2-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)-1,1,1,3,3,3-hexafluoropropan-2-ol was prepared similar to the procedure to prepare I-123, step 1. (MS: [M+1]+ 499)


Example 99: Synthesis of methyl (2S,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidine-2-carboxylate (I-657)



embedded image


To a vial were added methyl (2S,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidine-2-carboxylate (20 mg, 0.0491 mmol) THF (0.5 mL) and ethanol (1.0 mL). At 0° C., LiBH4 (9.5 mg, 0.436 mmol) was added and the resulting mixture was stirred overnight. The mixture was diluted with DCM (20 mL), washed with saturated NH4Cl (5 mL), H2O (5 mL), brine (5 mL), and dried over Na2SO4. The crude was purified by silica gel chromatography to afford the titled compound as white solid (MS: [M+1]+ 379).


Example 100: Synthesis of ((S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl hydrogen phosphonate (I-658)

I-658 was prepared essentially by the same method to prepare I-111.


















MS


I-#
Starting Material
Structure
[M + 1]+



















I-658


embedded image




embedded image




embedded image


427









Example 101: Synthesis of (R)-2-((S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)-2-hydroxyacetic acid. hexafluoropropan-2-ol (I-661)



embedded image


Step 1: To a vial were added DCM (10 mL), TEA (1 mmol, 0.1 mL) and tert-butyl (S)-2-formylpyrrolidine-1-carboxylate (1 g, 5 mmol) and the mixture was stirred at r.t overnight. After evaporation, the crude tert-butyl (S)-2-((R)-cyano(hydroxy)methyl)pyrrolidine-1-carboxylate was used in the next step without further purification. (MS: [M+1]+ 227.1).


Step 2: A mixture of tert-butyl (S)-2-((R)-cyano(hydroxy)methyl)pyrrolidine-1-carboxylate (230 mg, 1 mmol), HCl (con., 1 mL) and 1,4-dioxane (1 mL) was refluxed for 2 h. After evaporation, the crude (R)-2-hydroxy-2-((S)-pyrrolidin-2-yl)acetic acid was used in the next step without further purification. (MS: [M+1]+ 146.1).


Step 3: (R)-2-((S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)-2-hydroxyacetic acid (I-661) was prepared similar to step 1 for the preparation of I-123. (MS: [M+1]+407.1).


Example 102: Synthesis of methyl (S)-3-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoate (I-665) and (S)-3-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoic acid (I-664)



embedded image


Step 1: tert-Butyl (S)-2-((3-(tert-butoxy)-3-oxopropoxy)methyl)pyrrolidine-1-carboxylate. To a vial were added tert-butyl (S)-2-(hydroxymethyl)pyrrolidine-1-carboxylate (805 mg, 4.1 mmol, 1.0 eq.), Cs2CO3 (1.3 g, 4.1 mmol, 1.0 eq.), t-BuOH (5 mL) and tert-butyl acrylate (1.8 mL, 20 mmol, 5 eq.). The resulting reaction mixture was stirred over night. Then the mixture was diluted by ethyl acetate (50 mL), washed by H2O (3×15 mL), brine (15 mL) and dried over Na2SO4. After concentration, the crude was purified by a silica gel chromatography to afford the title product (1.22 g).


Step 2: (S)-3-(Pyrrolidin-2-ylmethoxy)propanoic acid. The mixture of tert-butyl (S)-2-((3-(tert-butoxy)-3-oxopropoxy)methyl)pyrrolidine-1-carboxylate (400 mg, 1.21 mmol), TFA (2 mL) and DCM (2 mL) was stirred overnight at room temperature. The mixture was concentrated to remove all volatiles. The crude was used in the next step.


Step 3: (S)-3-((1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoic acid (I-664). To a vial were added 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (227 mg, 0.759 mmol, 1.0 eq.), the crude product in step 2 (0.911 mmol), DMSO (4.0 mL), DIPEA (0.66 mL, 3.8 mmol, 5.0 eq.) and NaHCO3. The reaction mixture was srirred at 90° C. overnight. Then the mixture was diluted by ethyl acetate (30 mL) and acidified by HOAc (1 M, 5 mL), washed by H2O (3×10 mL), brine (10 mL) and dried over Na2SO4. After concentration, the crude was used in the next step. The acid product was purified by silica gel chromatography. MS: [M+1]+ 435.


Step 4: Methyl (S)-3-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoate (I-665). To a vial were added the crude product in step 3 (0.345 mmol) and anhydrous methanol (2.0 mL). The reaction solution was cooled to 0° C. and was treated with SOCl2 (0.100 mL, 1.38 mmol, 4.0 eq.) at 0° C. After being stirred at room temperature for 2 hours, the reaction mixture was diluted by ethyl acetate (30 mL), washed by NaHCO3/H2O (5%, 10 mL), H2O (3×10 mL), brine (10 mL) and dried over Na2SO4. After concentration, the crude was purified by a silica gel column to afford the title product (135 mg). MS: [M+1]+ 449.


The following compounds are prepared essentially by the same methods as described above for I-664 and 665.















Ex-





am-


MS


ple
Starting Material
Structure
[M + 1]+



















I-670


embedded image




embedded image




embedded image


435





I-671




embedded image


449





I-674


embedded image




embedded image




embedded image


435





I-688


embedded image




embedded image




embedded image


421





I-696


embedded image




embedded image




embedded image


449





I-698


embedded image




embedded image




embedded image


451





I-701




embedded image


465





I-699


embedded image




embedded image




embedded image


471





I-705


embedded image




embedded image




embedded image


453





I-708


embedded image




embedded image




embedded image


449





I-709


embedded image




embedded image




embedded image


461





I-719


embedded image




embedded image




embedded image


449





I-720




embedded image


463





I-717


embedded image




embedded image




embedded image


449





I-718




embedded image


463





I-741


embedded image




embedded image




embedded image


451





I-747


embedded image




embedded image




embedded image


449





I-745


embedded image




embedded image




embedded image


449





I-750


embedded image




embedded image




embedded image


449





I-752


embedded image




embedded image




embedded image


465





I-748


embedded image




embedded image




embedded image


465





I-749


embedded image




embedded image




embedded image


467





I-721


embedded image




embedded image




embedded image


503





I-722


embedded image




embedded image




embedded image


503





I-711


embedded image




embedded image




embedded image


465





I-704


embedded image




embedded image




embedded image


453





I-702


embedded image




embedded image




embedded image


451





I-703


embedded image




embedded image




embedded image


451





I-695


embedded image




embedded image




embedded image


485





I-672


embedded image




embedded image




embedded image


449





I-673


embedded image




embedded image




embedded image


463





I-677


embedded image




embedded image




embedded image


463





I-713


embedded image




embedded image




embedded image


527





I-712


embedded image




embedded image




embedded image


541





I-767


embedded image




embedded image




embedded image


421





I-769




embedded image


435





I-771


embedded image




embedded image




embedded image


421





I-707


embedded image




embedded image




embedded image


447





I-710


embedded image




embedded image




embedded image


447





I-689


embedded image




embedded image




embedded image


421





I-772


embedded image




embedded image




embedded image


435





I-773


embedded image




embedded image




embedded image


435





I-789


embedded image




embedded image




embedded image


449





I-693


embedded image




embedded image




embedded image


395





I-692


embedded image




embedded image




embedded image


423





I-694


embedded image




embedded image




embedded image


409





I-790


embedded image




embedded image




embedded image


449





I-792


embedded image




embedded image




embedded image


449





I-804


embedded image




embedded image




embedded image


447





I-808


embedded image




embedded image




embedded image


489





I-809


embedded image




embedded image




embedded image


447





I-806


embedded image




embedded image




embedded image


483





I-810


embedded image




embedded image




embedded image


449





I-832


embedded image




embedded image




embedded image


423





I-833


embedded image




embedded image




embedded image


437





I-834


embedded image




embedded image




embedded image


435





I-805


embedded image




embedded image




embedded image


447





I-836


embedded image




embedded image




embedded image


477





I-837


embedded image




embedded image




embedded image


485





I-838


embedded image




embedded image




embedded image


439





I-839


embedded image




embedded image




embedded image


511





I-873


embedded image




embedded image




embedded image


425





I-874


embedded image




embedded image




embedded image


497





I-816


embedded image




embedded image




embedded image


475





I-796


embedded image




embedded image




embedded image


507





I-815


embedded image




embedded image




embedded image


489





I-818


embedded image




embedded image




embedded image


479





I-858


embedded image




embedded image




embedded image


423





I-859


embedded image




embedded image




embedded image


397





I-860


embedded image




embedded image




embedded image


431





I-861


embedded image




embedded image




embedded image


445





I-862


embedded image




embedded image




embedded image


479





I-856


embedded image




embedded image




embedded image


431





I-857


embedded image




embedded image




embedded image


513





I-875


embedded image




embedded image




embedded image


431





I-876


embedded image




embedded image




embedded image


469





I-793


embedded image




embedded image




embedded image


503





I-794


embedded image




embedded image




embedded image


463





I-795


embedded image




embedded image




embedded image


449





I-827


embedded image




embedded image




embedded image


369





I-800


embedded image




embedded image




embedded image


447





I-801


embedded image




embedded image




embedded image


447





I-802


embedded image




embedded image




embedded image


511





I-803


embedded image




embedded image




embedded image


517





I-797


embedded image




embedded image




embedded image


465





I-798


embedded image




embedded image




embedded image


465





I-853


embedded image




embedded image




embedded image


435





I-855


embedded image




embedded image




embedded image


435





I-867


embedded image




embedded image




embedded image


465





I-799


embedded image




embedded image




embedded image


520





I-865


embedded image




embedded image




embedded image


463





I-807


embedded image




embedded image




embedded image


461





I-864


embedded image




embedded image




embedded image


435





I-866


embedded image




embedded image




embedded image


463





I-868


embedded image




embedded image




embedded image


465





I-811


embedded image




embedded image




embedded image


461





I-829


embedded image




embedded image




embedded image


435





I-869


embedded image




embedded image




embedded image


479





I-870


embedded image




embedded image




embedded image


479





I-871


embedded image




embedded image




embedded image


479





I-814


embedded image




embedded image




embedded image


449









Example 103: Synthesis of methyl (S)-2-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)acetate (I-667) and (S)-2-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)acetic acid (I-666)



embedded image


Step 1: tert-Butyl (S)-2-((2-(tert-butoxy)-2-oxoethoxy)methyl)pyrrolidine-1-carboxylate. To a flask were added tert-butyl (S)-2-(hydroxymethyl)pyrrolidine-1-carboxylate (1.0 g, 4.98 mmol, 1.0 eq.), Bu4NI (920 mg, 2.49 mmol, 0.50 eq.), toluene (20 mL) and tert-butyl 2-bromoacetate (1.94 g, 9.95 mmol, 2.0 eq.). The reaction mixture was cooled in an ice bath. A solution of NaOH/H2O (30%, 12 mL) was added slowly at 0° C. The resulting reaction mixture was stirred at 0° C. for 3 hours. Then the mixture was diluted by ethyl acetate (50 mL), washed by H2O (3×15 mL), brine (15 mL), and dried over Na2SO4. After concentration, the crude was purified by silica gel chromatography to afford the title product (1.14 g).


Step 2: (S)-2-(Pyrrolidin-2-ylmethoxy)acetic acid. The mixture of tert-butyl (S)-2-((2-(tert-butoxy)-2-oxoethoxy)methyl)pyrrolidine-1-carboxylate (300 mg, 0.951 mmol), TFA (1.5 mL) and DCM (1.5 mL) was stirred overnight at room temperature. The mixture was concentrated to remove all volatiles. The crude was used in the next step.


Step 3: (S)-2-((1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)acetic acid (I-666). The procedure was the same as that in the synthesis of I-665. MS: [M+1]+ 421.


Step 4: Methyl (S)-2-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)acetate. The procedure was the same as that in the synthesis of I-665. MS: [M+1]+ 435.


The following compounds are prepared essentially by the same methods described above for I-666.















Exam-


MS


ple
Starting Material
Structure
FIX



















I-697


embedded image




embedded image




embedded image


437





I-706


embedded image




embedded image




embedded image


439





I-740


embedded image




embedded image




embedded image


437





I-743


embedded image




embedded image




embedded image


435





I-744


embedded image




embedded image




embedded image


435





I-751


embedded image




embedded image




embedded image


451





I-746


embedded image




embedded image




embedded image


435





I-742


embedded image




embedded image




embedded image


437





I-669


embedded image




embedded image




embedded image


449





I-668




embedded image


435





I-766


embedded image




embedded image




embedded image


407





I-768




embedded image


421





I-770


embedded image




embedded image




embedded image


407





I-820


embedded image




embedded image




embedded image


449









Example 104: Synthesis of 3-(((S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)-2-fluoropropanoic acid (I-716) and methyl 3-(((S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)-2-fluoropropanoate (I-700)



embedded image


embedded image


Step 1: tert-Butyl (2S)-2-((3-(tert-butoxy)-2-fluoro-3-oxopropoxy)methyl)pyrrolidine-1-carboxylate and tert-butyl (2S)-2-((2-fluoro-3-methoxy-3-oxopropoxy)methyl)pyrrolidine-1-carboxylate. The procedure was the same as that in the synthesis of I-665. The two title compounds were obtained.


Step 2: 2-Fluoro-3-(((S)-pyrrolidin-2-yl)methoxy)propanoic acid. The procedure was the same as that in the synthesis of I-665.


Step 3: 3-(((S)-1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)-2-fluoropropanoic acid. The procedure was the same as that in the synthesis of I-665. (MS: [M+1]+ 453)


Step 4: Methyl 2-fluoro-3-(((S)-pyrrolidin-2-yl)methoxy)propanoate. The procedure was the same as that in the synthesis of I-665.


Step 5: Methyl 3-(((S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)-2-fluoropropanoate. The procedure was the same as that in the synthesis of I-665. MS: [M+1]+ 467.


Example 105: Synthesis of (2S,5S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-(2-hydroxyethyl)pyrrolidine-2-carboxylic acid (I-783)



embedded image


Step 1: di-tert-Butyl (2S)-5-hydroxypyrrolidine-1,2-dicarboxylate. To a vial were added di-tert-butyl (S)-5-oxopyrrolidine-1,2-dicarboxylate (1.14 g, 4.0 mmol, 1.0 eq.) and anhydrous THF (20 mL). The solution was cooled in a dry ice/acetone bath for 20 min. Then a solution of LiEt3BH (1 M, 4.8 mL, 4.8 mmol) was added at −78° C. dropwise. After addition, the resulting reaction mixture was stirred at −78° C. for 30 min. A saturated NaHCO3 in H2O (6 mL) was added to quench the reaction and the reaction mixture was warmed to 0° C. slowly followed by addition of H2O2/H2O (30%, 0.050 mL). After being stirred at room temperature for 20 min, the reaction mixture was concentrated, diluted by H2O (20 mL), extracted by DCM (3×20 mL), and dried over Na2SO4. After concentration, the crude was used in the next step.


Step 2: di-tert-Butyl (2S,5S)-5-(2-methoxy-2-oxoethyl)pyrrolidine-1,2-dicarboxylate. To a stirred suspension of sodium hydride (60%, 100 mg, 2.5 mmol, 1.2 eq.) in anhydrous DMF (5 mL) was added methyl 2-(diethoxyphosphoryl)acetate (529 mg, 2.5 mmol, 1.2 eq.). The mixture was stirred at room temperature for 1 hour and then a solution of the crude in step 1 (602 mg, 2.09 mmol) in DMF (5 mL) was added. The reaction was stirred overnight at room temperature, quenched with saturated aqueous NH4Cl solution, extracted with ethyl acetate (3×25 mL), and dried over Na2SO4. After concentration, the residue was purified by silica gel chromatography to afford the title product (150 mg).


Step 3: di-tert-Butyl (2S,5S)-5-(2-hydroxyethyl)pyrrolidine-1,2-dicarboxylate. To a vial were added di-tert-butyl (2S,5S)-5-(2-methoxy-2-oxoethyl)pyrrolidine-1,2-dicarboxylate (150 mg, 0.437 mmol, 1.0 eq.) and diethyl ether (2.5 mL). Then LiBH4 (19 mg, 0.874 mmol, 2 eq.) was added and the resulting reaction mixture was stirred overnight at room temperature. After addition of HOAc/H2O (1 M, 2 mL) and H2O (10 mL), the reaction mixture was extracted with ethyl acetate (2×20 mL) and dried over Na2SO4. After concentration, the residue was purified by silica gel chromatography to afford the title product (103 mg).


Step 4: (2S,5S)-5-(2-Hydroxyethyl)pyrrolidine-2-carboxylic acid. The procedure was the same as that in the synthesis of I-665.


Step 5: (2S,5S)-1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-(2-hydroxyethyl)pyrrolidine-2-carboxylic acid. The procedure was the same as that in the synthesis described above for I-665. MS: [M+1]+ 421.


Example 106: Synthesis of (2S,5S)-5-(2-(2-carboxyethoxy)ethyl)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidine-2-carboxylic acid (I-714)



embedded image


embedded image


Step 1: di-tert-Butyl (2S,5S)-5-(2-(3-(tert-butoxy)-3-oxopropoxy)ethyl)pyrrolidine-1,2-dicarboxylate. The procedure was the same as that in the synthesis described above for I-665.


Step 2: (2S,5S)-5-(2-(2-Carboxyethoxy)ethyl)pyrrolidine-2-carboxylic acid. The procedure was the same as that in the synthesis of I-665.


Step 3: Methyl (2S,5S)-5-(2-(3-methoxy-3-oxopropoxy)ethyl)pyrrolidine-2-carboxylate. The procedure was the same as that in the synthesis described above for I-665.


Step 4: Methyl (2S,5S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-(2-(3-methoxy-3-oxopropoxy)ethyl)pyrrolidine-2-carboxylate. The procedure was the same as that in the synthesis described above for I-665.


Step 5: (2S,5S)-5-(2-(2-Carboxyethoxy)ethyl)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidine-2-carboxylic acid (I-714). To a vial were added methyl (2S,5S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-(2-(3-methoxy-3-oxopropoxy)ethyl)pyrrolidine-2-carboxylate (3.7 mg, 0.0071 mmol, 1.0 eq.), THF (0.5 mL), MeOH (0.3 mL), and a solution of LiOH·H2O (1.5 mg, 0.0355 mmol, 5.0 eq.)/H2O (0.1 mL). The resulting solution was stirred at 40° C. for 6 h and acidized by aqueous HOAc (1 M, 0.040 mL). After concentration, the title product was obtained. MS: [M+1]+ 493.


Example 107: Synthesis of (2S,5S)-5-(2-(carboxymethoxy)ethyl)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidine-2-carboxylic acid (I-715)



embedded image


embedded image


Step 1: di-tert-Butyl (2S,5S)-5-(2-(2-(tert-butoxy)-2-oxoethoxy)ethyl)pyrrolidine-1,2-dicarboxylate. The procedure was the same as that in the synthesis described above for I-667.


Step 2: (2S,5S)-5-(2-(Carboxymethoxy)ethyl)pyrrolidine-2-carboxylic acid. The procedure was the same as that in the synthesis described above for I-665.


Step 3: Methyl (2S,5S)-5-(2-(2-methoxy-2-oxoethoxy)ethyl)pyrrolidine-2-carboxylate. The procedure was the same as that in the synthesis described above for I-665.


Step 4: Methyl (2S,5S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-(2-(2-methoxy-2-oxoethoxy)ethyl)pyrrolidine-2-carboxylate. The procedure was the same as that in the synthesis described above for I-665.


Step 5: (2S,5S)-5-(2-(Carboxymethoxy)ethyl)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidine-2-carboxylic acid. The procedure was the same as that in the synthesis described above for I-714. MS: [M+1]+ 479.


Example 108: Synthesis of (S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-3-amine (I-765)



embedded image


Step 1: tert-Butyl (S)-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-3-yl)carbamate. The procedure was the same as that in the synthesis of I-665.


Step 2: (S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-3-amine. The procedure was the same as that in the synthesis described above for I-665. MS: [M+1]+ 348.


Example 109: Synthesis of(S)—N-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-3-yl)acetamide (I-764)



embedded image


The procedure used to prepare I-764 was the same as that described above to prepare I-665. MS: [M+1]+ 390.


Example 110: Synthesis of (S)-2-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)acetamide (I-760)



embedded image


To a vial were added (S)-2-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)acetic acid (30 mg, 0.0712 mmol, 1.0 eq.), NH4Cl (19 mg, 0.356 mmol, 5.0 eq.), EDC-HCl (27 mg, 0.142 mmol, 2.0 eq.), HOBt (9.6 mg, 0.0712 mmol, 1.0 eq), DMF (0.5 mL). With stirring, DIPEA (0.062 mL, 0.356 mmol, 5.0 eq.) was added and the reaction mixture was stirred overnight at room temperature. The reaction mixture was diluted with ethyl acetate (20 mL), washed with H2O (3×10 mL), brine (10 mL), dried over Na2SO4. After concentration, the residue was purified by silica gel chromatography to afford the title product. MS: [M+1]+ 420.


The following compounds are prepared essentially by the same methods as described above for I-760.


















MS


Example
Starting Material
Structure
[M + 1]+



















I-761


embedded image


NH2OH•HCl


embedded image


436





I-762


embedded image


NH2CN


embedded image


445









Example 111: Synthesis of (S)-(2-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)ethyl)phosphonic acid (I-763)



embedded image


Step 1: tert-Butyl (S)-2-((2-(diethoxyphosphoryl)ethoxy)methyl)pyrrolidine-1-carboxylate. To a vial were added t-BuOK (247 mg, 2.2 mmol, 2.2 eq.) and anhydrous DCM (2.0 mL). The mixture was cooled in an ice bath for 20 min, followed by addition of tert-butyl (S)-2-(hydroxymethyl)pyrrolidine-1-carboxylate (201 mg, 1.0 mmol, 1.0 eq.). After the mixture was stirred at 0° C. for 30 min, diethyl (2-bromoethyl)phosphonate (294 mg, 1.2 mmol, 1.2 eq.) was added and the resultant reaction mixture was stirred at room temperature overnight. After being quenched by addition of ice (5 g), the mixture was diluted with ethyl acetate (30 mL), washed with H2O (2×10 mL), brine (10 mL), and dried over Na2SO4. After concentration, the residue was purified by silica gel chromatography to afford the title product (155 mg).


Step 2: Diethyl (S)-(2-(pyrrolidin-2-ylmethoxy)ethyl)phosphonate. The procedure was the same as that in the synthesis described above for I-665.


Step 3: Diethyl (S)-(2-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)ethyl)phosphonate. The procedure was the same as that in the synthesis described above for I-665.


Step 4: (S)-(2-((1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)ethyl)phosphonic acid. The mixture of diethyl (S)-(2-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)ethyl)phosphonate (9.5 mg, 0.018 mmol) and a solution of HCl/H2O (37%, 1.0 mL) was stirred at 75° C. over two days. The mixture was concentrated to give the title product. (MS: [M+1]+ 471)


Example 112: Synthesis of 3-((2S,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidine-2-carboxamido)propanoic acid (I-784) and methyl 3-((2S,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidine-2-carboxamido)propanoate (I-791)



embedded image


Step 1: tert-Butyl 3-((2S,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidine-2-carboxamido)propanoate. The procedure was the same as that in the synthesis described above for I-760.


Step 2: 3-((2S,3S)-1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidine-2-carboxamido)propanoic acid. The procedure was the same as that in the synthesis described above for I-665. MS: [M+1]+ 464.


Step 3: Methyl 3-((2S,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidine-2-carboxamido)propanoate. The procedure was the same as that in the synthesis described above for I-665. MS: [M+1]+ 478.


Example 113: Synthesis of 2-((2R,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidin-2-yl)acetic acid (I-785) and methyl 2-((2R,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidin-2-yl)acetate (I-786)



embedded image


Step 1: (S)-4-((tert-Butyldimethylsilyl)oxy)-3,4-dihydro-2H-pyrrole 1-oxide. (S)-Pyrrolidin-3-ol (1.5 g, 17.2 mmol) was treated with TBSCl (3.37 g, 22.3 mol) and imidazole (3.51 g) in DCM (25 mL) overnight. After dilution with DCM (50 mL), the organic layer was washed with water (15 mL), saturated NaHCO3 (15 mL) and brine. The isolated organic layer was dried over anhydrous Na2SO4. Concentration under reduced pressure afforded the desired crude (S)-3-((tert-butyldimethylsilyl)oxy)pyrrolidine (4.3 g), which was used in the following step without further purification. To an ice-water chilled solution of (S)-3-((tert-butyldimethylsilyl)oxy)pyrrolidine (3.5 g) in MeOH (40 mL) was added urea hydrogen peroxide (4.94 g). Then a solution of CH3ReO3 (21.8 mg) in MeOH (3 mL) was stepwise added to the above reaction mixture over 3 hours. After being stirred at room temperature over 4 h, the reaction mixture was quenched with Na2S2O3 (4 g, 27 mmol) cautiously. After aqueous work up with EtOAc (80 mL), the reaction mixture was purified by silica gel column chromatography to afford two products, the less polar fraction is the title product (0.72 g), eluting with a gradient between DCM and EtOAc. MASS: m/z: [M+1]+ : 216.


Step 2: Methyl 2-((2R,3S)-1,3-bis((tert-butyldimethylsilyl)oxy)pyrrolidin-2-yl)acetate. To a solution of (S)-4-((tert-butyldimethylsilyl)oxy)-3,4-dihydro-2H-pyrrole 1-oxide (0.72 g) in DCM were added 4 Å sieves (1.1 g) and ZnI2 (0.26 g). After the suspended solution was chilled with a bath of dry ice-acetone under N2, tert-butyl((1-methoxyvinyl)oxy)dimethylsilane (640 mg) was added stepwise via a syringe. The reaction mixture was stirred overnight as the temperature slowly rose back to room temperature. After aqueous work-up with DCM and concentration under reduced pressure, the resultant mixture was purified by silica gel column chromatography, eluting with 5% EtOAc in Hexane, to afford the title product (245 mg) as colorless solids. MASS: m/z: [M+1]+ : 404.


Step 3: Methyl 2-((2R,3S)-3-((tert-butyldimethylsilyl)oxy)pyrrolidin-2-yl)acetate. A solution of methyl 2-((2R,3S)-1,3-bis((tert-butyldimethylsilyl)oxy)pyrrolidin-2-yl)acetate (109 mg) with 5% Pd/C in AcOH (3.5 mL) was degassed (vacuum/purge) three times with hydrogen and stirred under hydrogen overnight. After filtration, rinsing with EtOAc, and concentration under reduced pressure, the resultant mixture was dissolved in EtOAc (15 mL). The organic layer was washed with sat. NaHCO3, brine, and dried over anhydrous Na2SO4. Concentration under reduced pressure afforded the title product (I-786-4, 76.8 mg). MASS: m/z: [M+1]+ : 274.


Step 4: 2-((2R,3S)-1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidin-2-yl)acetate (I-786). To a solution of methyl 2-((2R,3S)-3-((tert-butyldimethylsilyl)oxy)pyrrolidin-2-yl)acetate (76.8 mg) and DIPEA (0.12 mL) in DMSO (0.4 mL) was added 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (50 mg). The resultant mixture was stirred at 90° C. over 3 h and diluted with EtOAc (15 mL). The organic solution was treated with 1 M TBAF in THF (0.4 mL) over 30 min. The organic solution was washed with water and brine, then dried over anhydrous Na2SO4. Concentration under reduced pressure and purification by silica gel column chromatography afforded the title product (I-786, 18 mg) as light tan solids. MASS: m/z: [M+1]+ : 421.


Step 5: 2-((2R,3S)-1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidin-2-yl)acetic acid (I-785). To a solution of I-786 (44.8 mg) in MeOH (0.8 mL), THF (0.6 mL) and water (0.2 mL) was added LiOH·H2O (18 mg). The solution was stirred at room temperature over 3 h. After removal of the organic solvents under reduced pressure, the resultant slurry was mixed with AcOH (0.025 mL) and water (1.5 mL) to afford a suspended solution. The solid was isolated via a centrifuge and rinsed with water (0.5 mL×2). The wet cake was lyophilized to afford the title product (30 mg) as tan solids. MASS: m/z: [M+1]+ : 407.


Example 114: Synthesis of ethyl (2-((2R,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidin-2-yl)acetyl)glycinate (I-787) and (2-((2R,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidin-2-yl)acetyl)glycine (I-788)



embedded image


Step 1: Ethyl (2-((2R,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidin-2-yl)acetyl)glycinate (I-787). To a solution of 2-((2R,3S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidin-2-yl)acetic acid (I-785, 20 mg) in DMF (0.7 mL) were added EDC HCl (19 mg), HOBt (13 mg) and TEA (20 mg). The resultant solution was stirred at room temperature over 24 h. After the dilution with EtOAc (15 mL), the organic layer was washed with sat. NaHCO3, brine and dried over anhydrous Na2SO4. Concentration under reduced pressure and silica gel column chromatography, eluting with a gradient between hexane and EtOAc, afforded the title product (16 mg). MASS: m/z: [M+1]+ : 492.


Step 2: (2-((2R,3S)-1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidin-2-yl)acetyl)glycine (I-788). To a solution of I-787 (8 mg) in MeOH (0.6 mL) and water (0.2 mL) was added LiOH·H2O (8 mg). The resultant solution was stirred at room temperature over 2 h. After removal of MeOH and acidification of aqueous part with 1M HOAc (0.08 mL), the formed solids were isolated via a centrifuge and lyophilized to afford the titled product (6 mg) as tan solids. MASS: m/z: [M+1]+ : 464.


Example 115: Synthesis of (3aR,6aR)-4-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)hexahydro-2H-furo[3,2-b]pyrrol-2-one (I-774) and 2-((2R,3R)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidin-2-yl)acetic acid (I-781)



embedded image


Step 1: Benzyl (2R,3S)-3-((tert-butyldimethylsilyl)oxy)-2-(2-methoxy-2-oxoethyl)pyrrolidine-1-carboxylate. To a solution of methyl 2-((2R,3S)-3-((tert-butyldimethylsilyl)oxy)pyrrolidin-2-yl)acetate (280 mg) in EtOAc (8 mL) and water (3 mL) were added benzyl carbonochloridate (CBZ-Cl, 0.2 mL) and K2CO3 (200 mg). The reaction mixture was stirred at room temperature over 4 h. After the reaction mixture was diluted with EtOAc (25 mL), the organic layer was washed with water and brine, then dried over anhydrous Na2SO4. Removal of the organic solvents afforded the title product (300 mg) as oil. MASS: m/z: [M+1]+: 408.


Step 2: Benzyl (2R,3S)-3-hydroxy-2-(2-methoxy-2-oxoethyl)pyrrolidine-1-carboxylate. To a solution of benzyl (2R,3S)-3-((tert-butyldimethylsilyl)oxy)-2-(2-methoxy-2-oxoethyl)pyrrolidine-1-carboxylate (300 mg) in MeOH (5 mL) were added water (0.2 mL) and conc. HCl (0.2 mL). After the resultant solution was stirred overnight, it was diluted with EtOAc (40 mL). The organic layer was washed with water and brine, then dried over anhydrous Na2SO4. Removal of the organic solvent and purification by a silica gel column afforded the title product (143 mg) as oil. MASS: m/z: [M+1]+ 294.


Step 3: 2-((2R,3S)-1-((Benzyloxy)carbonyl)-3-hydroxypyrrolidin-2-yl)acetic acid. To a solution of benzyl (2R,3S)-3-hydroxy-2-(2-methoxy-2-oxoethyl)pyrrolidine-1-carboxylate (143 mg) in MeOH (5 mL) and water (2 mL) was added LiOH·H2O (62 mg). After the resultant solution was stirred over 3 h, 12 M HCl (0.014 mL) was added to adjust the pH to 2. After the reaction mixture was extracted with EtOAc (15 mL×3), the combined organic layers were washed with brine and dried over anhydrous Na2SO4. Removal of the organic solvents afforded the title product (98 mg) as colorless solids. MASS: m/z: [M+1]+ 280.


Step 4: Benzyl (3aR,6aR)-2-oxohexahydro-4H-furo[3,2-b]pyrrole-4-carboxylate. To a solution of 2-((2R,3S)-1-((benzyloxy)carbonyl)-3-hydroxypyrrolidin-2-yl)acetic acid (97 mg) in THF (8 mL) were added PPh3 (137 mg) and DEAD (0.24 mL, 40 wt % in toluene). After being stirred over 4 h, the reaction mixture was diluted with EtOAc. The organic layer was washed with water and brine, then dried over anhydrous Na2SO4. After removal of the organic solvents, the residuals were purified by a silica gel column, eluting with EtOAc/Hex, to afford the title product (75 mg).


Step 5: (3aR,6aR)-Hexahydro-2H-furo[3,2-b]pyrrol-2-one. To a solution of benzyl (3aR,6aR)-2-oxohexahydro-4H-furo[3,2-b]pyrrole-4-carboxylate (75 mg) in MeOH (4 mL) were added AcOH (0.1 mL) and 5% Pd/C (20 mg). The resultant suspension was degassed (vacuum-purge) with hydrogen three times and stirred under hydrogen overnight. Filtration and concentration afforded the title product. MASS: m/z: [M+1]+ 128.


Step 6: (3aR,6aR)-4-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)hexahydro-2H-furo[3,2-b]pyrrol-2-one (I-774) and 2-((2R,3R)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-hydroxypyrrolidin-2-yl)acetic acid (I-781). Following the last two steps of the preparation procedures for I-786 and I-785, both title compounds were prepared. I-774, MASS: m/z: [M+1]+ 389. I-781, MASS: m/z: [M+1]+ 407.


Example 116: Synthesis of 1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)hexahydro-6H-furo[3,4-b]pyrrol-6-one (I-775) and 1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-(hydroxymethyl)pyrrolidine-2-carboxylic acid (I-782)



embedded image


Following the last two steps of the preparation procedures of I-786 and I-785, I-775 was prepared from hexahydro-6H-furo[3,4-b]pyrrol-6-one and following hydrolysis afforded I-782. I-775, MASS: m/z: [M+1]+ 389. I-782, MASS: m/z: [M+1]+ 407.


Example 117: Synthesis of(S)-1-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl)-1H-pyrazole-5-carboxylic acid (I-727)



embedded image


Step 1: Methyl (S)-1-((1-(tert-butoxycarbonyl)pyrrolidin-2-yl)methyl)-1H-pyrazole-5-carboxylate. To a solution tert-butyl (S)-2-((tosyloxy)methyl)pyrrolidine-1-carboxylate (274 mg) in DMF (1.5 mL) were added methyl 1H-pyrazole-3-carboxylate (145.8 mg) and Cs2CO3 (377 mg). After the resultant mixture was stirred overnight, it was diluted with EtOAC. The organic layer was washed with water and brine, dried over anhydrous Na2SO4. Removal of solvents under reduced pressure and a silica gel column purification, eluting with a gradient between hexane and EtOAc/DCM (1:3), afforded the title product.


Step 2: (S)-1-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl)-1H-pyrazole-5-carboxylic acid (I-727). The title product was prepared by the deprotection of methyl (S)-1-((1-(tert-butoxycarbonyl)pyrrolidin-2-yl)methyl)-1H-pyrazole-5-carboxylate with TFA/DCM, coupling with 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline, and hydrolysis with LiOH which were identical to the last three steps described above to prepare I-664. MASS: m/z: [M+1]+ 457.


The following compounds are prepared essentially by the same methods described above for I-727.


















MS


Example
Starting Material
Structure
[M + 1]+



















I-728


embedded image




embedded image




embedded image


457





I-732


embedded image




embedded image




embedded image


456





I-731


embedded image




embedded image




embedded image


471





I-729




embedded image


471





I-730


embedded image




embedded image




embedded image


471









Example 118: Synthesis of 2-((R)-2-((S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)-2-hydroxyethoxy)acetic acid (I-726)



embedded image


embedded image


Step 1: tert-Butyl (S)-2-((R)-cyano(hydroxy)methyl)pyrrolidine-1-carboxylate and tert-butyl (S)-2-((S)-cyano(hydroxy)methyl)pyrrolidine-1-carboxylate. To a solution of tert-butyl (S)-2-formylpyrrolidine-1-carboxylate (1.92 g) in DCM (20 mL) was added TEA (2 mL) and acetone cyanohydrin (0.98 g). The resultant solution was stirred at room temperature over 40 h. After removal of DCM under reduced pressure, the residue was diluted with EtOAc (30 mL). The organic layer was washed with diluted HCl (0.5 N) and brine, then dried over anhydrous Na2SO4. The reaction mixture was purified with a silica gel column, eluting with a gradient of hexane and Hex/EtOAc/DCM (3/1/6), to afford tert-butyl (S)-2-((R)-cyano(hydroxy)methyl)pyrrolidine-1-carboxylate (1.4 g) as oil and tert-butyl (S)-2-((S)-cyano(hydroxy)methyl)pyrrolidine-1-carboxylate (0.98 g) as solids.


Step 2-4: tert-Butyl (S)-2-((R)-1-hydroxy-2-methoxy-2-oxoethyl)pyrrolidine-1-carboxylate. To a solution of tert-butyl (S)-2-((R)-cyano(hydroxy)methyl)pyrrolidine-1-carboxylate (0.68 g) in dioxane (8 mL) was added conc. HCl (8 mL). The resultant solution was heated at reflux over 4 h. The solution was concentrated under reduced pressure to dryness. The residuals were dissolved in MeOH (10 mL) and treated with SOCl2 (0.1 mL) overnight. After removal of MeOH under reduced pressure, the residuals were suspended in DCM (10 mL) and water (4 mL) in the presence of (Boc)2O (1.29 g) and Na2CO3 (954 mg) overnight. After adding DCM (15 mL), the combined DCM layers were isolated and washed with brine, then dried over anhydrous Na2SO4. A silica gel column purification afforded the title compound (0.64 g) as oil. MASS: m/z: [M+Na]+282.


Step 5-6: tert-Butyl (2S)-2-((1R)-2-hydroxy-1-((tetrahydro-2H-pyran-2-yl)oxy)ethyl)pyrrolidine-1-carboxylate. To a solution of tert-butyl (S)-2-((R)-1-hydroxy-2-methoxy-2-oxoethyl)pyrrolidine-1-carboxylate (440 mg) in DCM (6 mL) were added 3,4-dihydro-2H-pyran (336 mg) and PPTS (100 mg). The solution was stirred over 6 h. After the removal of the solvents under reduced pressure, the residuals were suspended in THF (6 mL) and treated with LiBH4 (96 mg). After the resultant suspension was stirred overnight, it was slowly quenched by adding water. After the aqueous work-up with EtOAc (25 mL), the isolated organic layer was washed with brine and dried over anhydrous Na2SO4. A silica gel column purification afforded the title product (224 mg). MASS: m/z: [M+Na]+338.


Step 7: tert-Butyl (2S)-2-((1R)-2-(2-(tert-butoxy)-2-oxoethoxy)-1-((tetrahydro-2H-pyran-2-yl)oxy)ethyl)pyrrolidine-1-carboxylate. To an ice-chilled solution of tert-butyl (2S)-2-((1R)-2-hydroxy-1-((tetrahydro-2H-pyran-2-yl)oxy)ethyl)pyrrolidine-1-carboxylate (101 mg) in DMF (3 mL) was slowly added NaH (40 mg, 60% wt suspension in mineral oil). After the resultant solution was stirred over 20 min at 0° C., tert-butyl 2-bromoacetate (187 mg) was added. The reaction mixture was stirred at room temperature overnight and then quenched with water (0.5 mL). Aqueous workup with EtOAc and a silica gel column purification gave the title product (86 mg) as oil. MASS: m/z: [M+Na]+452.


Step 8-9: 2-((R)-2-((S)-1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)-2-hydroxyethoxy)acetic acid (I-726). After tert-butyl (2S)-2-((1R)-2-(2-(tert-butoxy)-2-oxoethoxy)-1-((tetrahydro-2H-pyran-2-yl)oxy)ethyl)pyrrolidine-1-carboxylate (86 mg) in DCM (0.2 mL) was treated with TFA (0.2 mL) overnight, the organic solvents were removed under reduced pressure. Additional co-evaporation with toluene/ACN helped the removal of excess of TFA. The residuals in DMSO (0.4 mL) were mixed with 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (48 mg) and Na2CO3 (32 mg). The resultant suspension was heated at 90° C. for 5 h. The reaction mixture was treated with AcOH (0.03 mL) and suspended in water. Aqueous work-up with EtOAc and a silica gel column purification with a gradient eluting between EtOAc and DCM/MeOH/AcOH (9/0/0.1) afforded the desired product (14.2 mg) as tan solids. MASS: m/z: [M+1]+ : 451.


Example 119: Synthesis of 2-((S)-2-((S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)-2-hydroxyethoxy)acetic acid (I-725)



embedded image


Following the same methods described above to prepare I-726, I-725 was prepared from tert-butyl (S)-2-((S)-cyano(hydroxy)methyl)pyrrolidine-1-carboxylate. I-725: MASS: m/z: [M+1]+: 451.


Example 120: Synthesis of 3-((R)-2-((S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)-2-hydroxyethoxy)propanoic acid (I-724)



embedded image


Following the same methods described above to prepare I-664, I-724 was prepared from tert-butyl (2S)-2-((1R)-2-hydroxy-1-((tetrahydro-2H-pyran-2-yl)oxy)ethyl)pyrrolidine-1-carboxylate. I-724: MASS: m/z: [M+1]f: 465.


Example 121: Synthesis of (S)-6-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)hexanoic acid (I-675)



embedded image


Step 1: tert-Butyl (S)-2-((3-(tert-butoxy)-3-oxopropoxy)methyl)pyrrolidine-1-carboxylate. At 0° C., NaH (80 mg, 60% in mineral oil, 2.0 mmol) was added to a solution of tert-butyl (S)-2-(hydroxymethyl)pyrrolidine-1-carboxylate (201 mg, 1 mmol) in THF (5 mL). After stirring for 30 min at r.t., a solution of methyl 6-bromohexanoate (0.6 mL) in THF (1 mL) was added at 0° C. The mixture was stirred for 2 h at r.t. before quenching by H2O (1 mL) at 0° C. The organic phase was concentrated and purified by silica gel column to afford the title product (75 mg) as colorless oil. MS: [M+1]+ 330.


Step 2: Methyl (S)-6-(pyrrolidin-2-ylmethoxy)hexanoate. To a solution of tert-butyl (S)-2-((3-(tert-butoxy)-3-oxopropoxy)methyl)pyrrolidine-1-carboxylate (75 mg, 0.23 mmol) in DCM (0.8 mL) was added TFA (0.8 mL) at 0° C. After stirring 1 h at r.t., the reaction mixture was concentrated to colorless oil (75 mg) under reduced pressure. The crude was used for the next step without further purification. MS: [M+1]+ : 230.


Step 3: Methyl (S)-6-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)hexanoate. To a solution of methyl (S)-6-(pyrrolidin-2-ylmethoxy)hexanoate (75 mg, crude from last step, about 0.2 mmol) and 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (100 mg, 0.33 mmol) in DMSO (1 mL) was added DIPEA (0.1 mL). The mixture was stirred at 80° C. overnight. After cooling to r.t., H2O (10 mL) was added. The mixture was acidified to pH.=1 by 1 N HCl and the precipitate was collected by filtration. Purification by silica gel column afforded the title product (100 mg) as colorless oil. MS: [M+1]+ 491.


Step 4: (S)-6-((1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)hexanoic acid. To a solution of methyl (S)-6-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)hexanoate (100 mg, 0.2 mmol) in THF (2 mL) was added LiOH (aq., 1 M. 0.4 mL) at 0° C. The mixture was stirred at 50° C. overnight. After cooling to r.t., H2O (3 mL) and 1 N HCl (0.35 mL) was added. The precipitate was collected by filtration to afford the title product (80 mg) as white solid. MS: [M+1]+ 477.


The following compounds are prepared essentially by the same method described above to prepare I-675.


















MS


Example
Starting Material
Structure
[M + 1]+



















I-676


embedded image




embedded image




embedded image


463





I-723


embedded image




embedded image




embedded image


461









Example 122: Synthesis of tert-Butyl (S)-2-(2-hydroxyethyl)pyrrolidine-1-carboxylate



embedded image


At 0° C., LAH (1.2 eq.) was added to a solution of (S)-2-(1-(tert-butoxycarbonyl)pyrrolidin-2-yl)acetic acid (4 g, 17 mmol) in THF (75 mL). The mixture was stirred at r.t. for 1 h. The reaction was quenched by H2O (3 mL), 15% NaOH (3 mL) and H2O (9 mL) respectively at 0° C. After filtration, the filtrate was purified by silica gel column to afford the title product (2.3 g) as colorless oil. MS: [M+1]+216.


Example 123: Synthesis of tert-Butyl (S)-2-(3-hydroxypropyl)pyrrolidine-1-carboxylate



embedded image


Step 1: tert-Butyl (S,E)-2-(3-methoxy-3-oxoprop-1-en-1-yl)pyrrolidine-1-carboxylate. To a solution of tert-butyl (S)-2-(hydroxymethyl)pyrrolidine-1-carboxylate (398 mg, 2 mmol) in THF (8 mL) was added Ph3P═CHCOOMe (700 mg, 2.2 mmol). The mixture was stirred at 50° C. overnight. After cooling to r.t., the mixture was concentrated by vacuum and purified by silica gel chromatography to afford the title product (430 mg) as colorless oil. MS: [M+1]+ 256.


Step 2: tert-Butyl (S)-2-(3-methoxy-3-oxopropyl)pyrrolidine-1-carboxylate. To a solution of tert-butyl (S,E)-2-(3-methoxy-3-oxoprop-1-en-1-yl)pyrrolidine-1-carboxylate (800 mg, 3.1 mmol) in MeOH (10 mL) was added Pd/C (10%, 40 mg). The mixture was stirred under H2 (1 atm) over night. After filtration, the filtrate was concentrated and afford the title product (800 mg) as colorless oil which was used directly without purification. MS: [M+1]+ 258.


Step 3: tert-Butyl (S)-2-(3-hydroxypropyl)pyrrolidine-1-carboxylate. The procedure was essentially the same as described above for the preparation of tert-butyl (S)-2-(2-hydroxyethyl)pyrrolidine-1-carboxylate. MS: [M+1]+ 230.


Example 124: Synthesis of tert-Butyl (R)-2-(4-methoxy-4-oxobutyl)pyrrolidine-1-carboxylate



embedded image


Step 1: tert-Butyl (S)-2-(2-oxoethyl)pyrrolidine-1-carboxylate. To a solution of tert-butyl (S)-2-(2-hydroxyethyl)pyrrolidine-1-carboxylate (340 mg, 1.5 mmol) in DCM (6 mL) and pyridine (1 mL) was added DMP (800 mg) at 0° C. The mixture was stirred at r.t. for 2 h and quenched by Na2S2O3/NaHCO3 (sat., 10 mL). The organic phase was concentrated by vacuum and purified by silica gel chromatography to afford the title product (280 mg) as colorless oil. MS: [M+1]+ 214.


Step 2 and step 3 were essentially the same procedure as described above to prepare tert-butyl (S)-2-(3-hydroxypropyl)pyrrolidine-1-carboxylate preparation (Step 1 and Step 2).


Example 125: Synthesis of tert-Butyl (R)-2-(5-methoxy-5-oxopentyl)pyrrolidine-1-carboxylate



embedded image


tert-butyl (R)-2-(5-methoxy-5-oxopentyl)pyrrolidine-1-carboxylate was prepared essentially by the same method described above to prepare tert-butyl (R)-2-(4-methoxy-4-oxobutyl)pyrrolidine-1-carboxylate.


Example 126: Synthesis of Ethyl (2S,3S,4R)-3-hydroxy-4-methylpyrrolidine-2-carboxylate and ethyl (2S,3S, 4R)-4-hydroxy-3-methylpyrrolidine-2-carboxylate



embedded image


The two intermediates were prepared according the method described in WO 2011/091407, the content of which in herein incorporated by reference.


Example 127: Synthesis of methyl (S)-3-(((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl)amino)propanoate (I-753) and dimethyl 3,3′-(((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl)azanediyl)(S)-dipropionate (I-757)



embedded image


embedded image


Step 1: tert-Butyl (S)-2-(azidomethyl)pyrrolidine-1-carboxylate. To a solution of tert-butyl (S)-2-(hydroxymethyl)pyrrolidine-1-carboxylate (400 mg, 2 mmol), Ph3P (800 mg, 3 mmol) and NaN3 (200 mg, 3 mmol) in THF (10 mL) was added DEAD (0.47 mL, 3 mmol). The reaction was stirred over night at r.t. After evaporation, the crude was purified by silica gel chromatography to afford the title product (280 mg) as colorless oil. MS: [M+1]+ 227.


Step 2 and step 3 were essentially the same as the procedure described above to prepare I-675 (Step 2 and Step 3).


Step 4: (S)-(1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methanamine. To a solution of (S)-2-(2-(azidomethyl)pyrrolidin-1-yl)-7,8-dichloro-4-(1H-imidazol-1-yl)quinoline (190 mg, 0.5 mmol) in THF (3 mL) and H2O (1 mL) was added Ph3P (262 mg, 1 mmol). The mixture was stirred over night at r.t. After evaporation, the crude was purified by silica gel chromatography to afford the title product (160 mg) as white solid. MS: [M+1]+ 362.


Step 5: Methyl (S)-3-(((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl)amino)propanoate (I-753) and dimethyl 3,3′-(((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl)azanediyl)(S)-dipropionate (I-757). To a solution of (S)-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methanamine (160 mg) and methyl 3-bromopropanolate (50 mg, 0.44 mmol) was added K2CO3 (60 mg). The mixture was stirred at 50° C. overnight. After filtration, the crude was purified by silica gel chromatography to afford the titled compounds I-753 (45 mg, MS: [M+1]+ 448) and I-757 (2.5 mg, MS: [M+1]+ 534) as white solid.


Example 128: Synthesis of (S)-3-(((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl)amino)propanoic acid (I-756)



embedded image


The procedure was essentially the same as the preparation described above for I-675 (Step 4). MS: [M+1]+ 434.


Example 129: Synthesis of methyl (S)-3-(((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl)(methyl)amino)propanoate (I-755) and (S)-3-(((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl)(methyl)amino)propanoic acid (I-754)



embedded image


Step 1: Methyl (S)-3-(((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl)(methyl)amino)propanoate (I-755). To a solution of methyl (S)-3-(((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl)amino)propanoate (19 mg, 0.05 mmol), paraformaldehyde (6 mg, 0.2 mmol), HOAc (0.025 mL) in MeOH (1 mL) was added NaBH3CN (13 mg, 0.2 mmol). The mixture was stirred at r.t. over night. After evaporation, the crude was purified by silica gel chromatography to afford the title product (15 mg). MS: [M+1]+ 462.


Step 2: (S)-3-(((1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methyl)(methyl)amino)propanoic acid (I-754). The procedure was essentially the same that described above for the preparation of I-675 (Step 4). MS: [M+1]+ 448.


The following compounds are prepared essentially by the same method described above to prepare I-123.


















MS


Example
Starting Material
Structure
[M + 1]+



















I-776


embedded image




embedded image




embedded image


391





I-687


embedded image




embedded image




embedded image


417





I-680


embedded image




embedded image




embedded image


419





I-679


embedded image




embedded image




embedded image


405





I-682


embedded image




embedded image




embedded image


419





I-685


embedded image




embedded image




embedded image


417





I-686


embedded image




embedded image




embedded image


431





I-681


embedded image




embedded image




embedded image


433





I-683


embedded image




embedded image




embedded image


433





I-684


embedded image




embedded image




embedded image


433





I-678


embedded image




embedded image




embedded image


435





I-779


embedded image




embedded image




embedded image


435





I-780


embedded image




embedded image




embedded image


407





I-778


embedded image




embedded image




embedded image


407





I-690


embedded image




embedded image




embedded image


393





I-739


embedded image




embedded image




embedded image


433





I-691


embedded image




embedded image




embedded image


407





I-758


embedded image




embedded image




embedded image


408





I-759




embedded image


394









Example 130: Synthesis of (S)-3-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)benzoic acid (I-735)



embedded image


Step 1: tert-Butyl (S)-2-((3-(methoxycarbonyl)phenoxy)methyl)pyrrolidine-1-carboxylate. To a stirred mixture of Boc-prolinol (300 mg, 1.49 mmol), methyl-3-hydroxybenzoate (240 mg, 1.45 mmol) and triphenylphosphine (391 mg, 1.49 mmol) in THF (10 mL) was added dropwise diethyl azodicarboxylate (261 mg, 0.65 mL, 1.49 mmol (40% in toluene)). The resulting mixture was heated at 75° C. for 3 h. After cooling, the mixture was concentrated in vacuo. The residue was dissolved in EtOAc and washed successively with 1N NaOH, water and then brine. The organics were dried (Na2SO4) then concentrated. The residue was purified by silica gel chromatography using 0-40% ethyl acetate/hexanes as eluent to afford the title compound. MS: [M+1]+ 280.1 (−t-Bu).


Step 2: Methyl (S)-3-(pyrrolidin-2-ylmethoxy)benzoate. To a flask containing tert-butyl (S)-2-((3-(methoxycarbonyl)phenoxy)methyl)pyrrolidine-1-carboxylate (200 mg, 0.546 mmol) was added dichloromethane (5 mL). Trifluoroacetic acid (1 mL) was added and the reaction allowed to stir at rt for 16 h. The volatiles were removed to afford the title compound which was taken onward without further purification. MS: [M+1]+ 236.1.


Step 3: Methyl (S)-3-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)benzoate. To a vial containing methyl (S)-3-(pyrrolidin-2-ylmethoxy)benzoate (70 mg, 0.3 mmol) was added DMSO (0.15 mL) and DIPEA (0.10 mL). Next 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (30 mg, 0.1 mmol) was added and the mixture heated to 90° C. for 16 h. The reaction was cooled to rt, then water was added and the organics extracted into 10% MeOH/CH2Cl2. The combined organic layers were dried (Na2SO4) then concentrated. The residue was purified by silica gel chromatography using 0-5% MeOH/CH2Cl2 with 0.5% HOAc as eluent to afford the title compound. MS: [M+1]+ 497.1. 108571 Step 4: (S)-3-((1-(7,8-Dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)benzoic acid. To a vial containing methyl (S)-3-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)benzoate (20 mg, 0.040 mmol) THF (2 mL) and water (0.5 mL) was added lithium hydroxide monohydrate (5 mg, 0.12 mmol) and the reaction allowed to stir at rt for 16 h. The volatiles were removed and the residue taken up in water then acidified with 1M HCl until pH˜7. The resulting solids were filtered off and vac-dried to afford the title compound. MS: [M+1]+ 483.1.


The following compounds were prepared essentially by the same methods described above to prepare I-735:


















MS


Example
Starting Material
Structure
[M + 1]+



















I-733


embedded image




embedded image




embedded image


483.1





I-737


embedded image




embedded image




embedded image


483.1





I-736


embedded image




embedded image




embedded image


497.1





I-738


embedded image




embedded image




embedded image


497.1





I-734


embedded image




embedded image




embedded image


497.1





I-842


embedded image




embedded image




embedded image


457





I-843


embedded image




embedded image




embedded image


471





I-846


embedded image




embedded image




embedded image


471









Example 131: Synthesis of methyl (S)-2-(2-(pyrrolidin-2-ylmethoxy)phenyl)acetate



embedded image


Step 1: tert-Butyl (S)-2-((2-(2-methoxy-2-oxoethyl)phenoxy)methyl)pyrrolidine-1-carboxylate. To a flask containing tert-butyl (S)-2-((tosyloxy)methyl)pyrrolidine-1-carboxylate (355 mg, 1.00 mmol), methyl 2-(2-hydroxyphenyl)acetate (166 mg, 1.00 mmol) and DMF (10 mL) was added potassium tert-butoxide (112 mg, 1.00 mmol) and the reaction was heated at 55° C. for 16 h. The reaction was cooled to rt, quenched by the slow addition of water then diluted with ethyl acetate. The organic layer was separated and washed further with 5% NaOH, water, and brine then dried (Na2SO4) and concentrated. The resulting residue purified by silica gel chromatography using 0-35% EtOAc/hexanes as eluent to afford to title compound. (MS: [M+1]+ 250.1 (−Boc).


Step 2: Methyl (S)-2-(2-(pyrrolidin-2-ylmethoxy)phenyl)acetate. To a vial containing tert-butyl (S)-2-((2-(2-methoxy-2-oxoethyl)phenoxy)methyl)pyrrolidine-1-carboxylate (25 mg, 0.061 mmol) was added dichloromethane (2 mL) and trifluoroacetic acid (0.5 mL). The reaction allowed to stir at rt for 16 h. The volatiles were removed to afford the title compound used to prepare I-735. MS: [M+1]+ 250.1.


Example 132: Synthesis of 3-(2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)ethoxy)benzoic acid (I-885)



embedded image


Step 1: Methyl 3-(2-((tert-butoxycarbonyl)amino)ethoxy)benzoate: To a flask containing methyl 3-hydroxybenzoate (152 mg, 1.0 mmol) and DMF (5 mL) was added potassium carbonate (152 mg, 1.1 mol). The contents were stirred at rt for 10 min then tert-butyl (2-bromoethyl)carbamate (246 mg, 1.1 mmol) was added. The reaction was heated to 65° C. for 16 h. Upon cooling, water (10 mL) was added and the organics were extracted into ethyl acetate (2×10 mL), dried (Na2SO4) and concentrated. The residue was purified by silica gel chromatography to afford 175 mg of the titled compound.


Step 2: Methyl 3-(2-aminoethoxy)benzoate TFA salt: To a flask containing methyl 3-(2-((tert-butoxycarbonyl)amino)ethoxy)benzoate (120 mg, 0.4 mmol) and dichloromethane (5 mL) was added trifluoroacetic acid (1 mL). The reaction was allowed to stir at rt for 16 h. The volatiles were concentrated off to afford crude titled compound which was taken on directly to the next step.


Step 3: Methyl 3-(2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)ethoxy)benzoate: To a vial containing 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (30 mg, 0.1 mmol) and DMSO was added methyl 3-(2-aminoethoxy)benzoate TFA salt (0.4 mmol) and DIPEA. The reaction was heated to 90° C. for 16 h then cooled to rt. Water (5 mL) was added and the organics extracted into ethyl acetate (2×5 mL), dried (Na2SO4) and concentrated. The residue was purified by silica gel chromatography to afford 27 mg of the titled compound.


Step 4: 3-(2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)ethoxy)benzoic acid: To a vial containing methyl 3-(2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)ethoxy)benzoate (20 mg, 0.44 mmol) in THF/H2O (4 mL/1 mL) was added lithium hydroxide monohydrate (7 mg, 0.175 mmol) and the reaction allowed to stir at rt for 16 h. The volatiles were removed and then residue acidified with 1M HCl. The resulting solids were filtered and dried to afford the titled compound.


The following compounds are prepared essentially by the same method described above to prepare I-885.


















MS


Example
Starting Material
Structure
[M + 1]+



















I-886


embedded image




embedded image




embedded image


443





I-889


embedded image




embedded image




embedded image


417









Example 133: Synthesis of 4-((S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)-2-hydroxybutanoic acid (I-817)



embedded image


Step 1: tert-butyl (2S)-2-(3-cyano-3-hydroxypropyl)pyrrolidine-1-carboxylate. To a solution of tert-butyl (S)-2-(3-oxopropyl)pyrrolidine-1-carboxylate (114 mg, 0.5 mmol) in DCM (1 mL) was added Et3N (0.1 mL) and acetone cyanohydrin (0.1 mL, 1.2 mmol). The mixture was stirred at r.t. over night. The solution was concentrated by vacuum and purified by silica gel column to afford the title product (120 mg) as colorless oil. MS: [M+1]+ 255.


Step 2: 2-hydroxy-4-((S)-pyrrolidin-2-yl)butanoic acid. To a solution of tert-butyl (2S)-2-(3-cyano-3-hydroxypropyl)pyrrolidine-1-carboxylate (60 mg, 0.23 mmol) in dioxane (0.8 mL) was added HCl (con., 0.8 mL). The mixture was stirred at 100° C. overnight. After cooling to r.t., dioxane was removed by evaporation. The residue was washed by EtOAc (0.5 mL×2). The aqueous phase was collected and evaporated to give a crude product which was used directly in the next step without purification. MS: [M+1]+ 174.


Step 3: 4-((S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)-2-hydroxybutanoic acid (I-817). The title compound was prepared essentially by the same methods as for I-664. MS: [M+1]+ 435.


Synthesis of (S)-4-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)but-2-ynoic acid (I-819)



embedded image


Step 1: tert-butyl (S)-2-(prop-2-yn-1-yl)pyrrolidine-1-carboxylate. To a solution of tert-butyl (S)-2-(2-oxoethyl)pyrrolidine-1-carboxylate (457 mg, 2.15 mmol) in MeOH (4.5 mL) was added K2CO3 (594 mg, 4.3 mmol) and diazole (480 mg, 3.22 mmol). The mixture was stirred at r.t. over night. The mixture was concentrated by vacuum and purified by silica gel column to afford the title product (330 mg) as colorless oil. MS: [M+1]+ 210.


Step 2: tert-butyl (S)-2-(4-methoxy-4-oxobut-2-yn-1-yl)pyrrolidine-1-carboxylate. To a solution of tert-butyl (S)-2-(prop-2-yn-1-yl)pyrrolidine-1-carboxylate (330 mg, 1.58 mmol) in THF (3.5 mL) was added n-BuLi (1.1 mL, 1.6 M in hexane) at −78° C. After 30 min, a solution of methyl chloroformate (0.15 mL) in THF (1 mL) was added dropwise. The mixture was warmed to r.t. and stirred over night. The reaction was then quenched by NH4Cl (sat., 0.5 mL). The mixture was concentrated by vacuum and purified by silica gel column to afford the title product (200 mg) as colorless oil. MS: [M+1]+ 268.


(S)-4-(1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)but-2-ynoic acid (I-819) is prepared essentially by the same methods as for I-664 from tert-butyl (S)-2-(4-methoxy-4-oxobut-2-yn-1-yl)pyrrolidine-1-carboxylate. MS: [M+1]+ 415.


Example 134: Synthesis of (S)-3-((1-(7-chloro-4-(1H-imidazol-1-yl)-8-phenylquinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoic acid (I-863)



embedded image


A mixture of (S)-3-((1-(8-bromo-7-chloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoic acid (14 mg, 0.03 mmol), Na2CO3 (10 mg, 0.14 mmol), dioxane (1 mL) and H2O (0.5 mL) was purged by N2 for 10 min. Tetrakis (1 mg, 0.001 mmol) was added. The mixture was stirred at 80° C. for 3 h. The mixture was concentrated by vacuum and purified by silica gel column to afford I-863 (6 mg) as white solid. MS: [M+1]+ 477.


Example 135: Synthesis of 3-(((S)-1-(7,8-dichloro-4-((R)-3-hydroxypyrrolidin-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoic acid (I-821)



embedded image


embedded image


Step 1: tert-butyl (S)-2-((3-(tert-butoxy)-3-oxopropoxy)methyl)pyrrolidine-1-carboxylate. The procedure was the same as that in the synthesis of I-665.


Step 2: (S)-3-(pyrrolidin-2-ylmethoxy)propanoic acid. The procedure was the same as that in the synthesis of I-665.


Step 3: methyl (S)-3-(pyrrolidin-2-ylmethoxy)propanoate. The procedure was the same as that in the synthesis of I-665.


Step 4: methyl (S)-3-((1-(4,7,8-trichloroquinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoate. The procedure was the same as that in the synthesis of I-665.


Step 5: methyl 3-(((S)-1-(7,8-dichloro-4-((R)-3-hydroxypyrrolidin-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoate. To a vial were added methyl (S)-3-((1-(4,7,8-trichloroquinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoate (21 mg, 0.050 mmol), (R)-pyrrolidin-3-ol (22 mg, 0.25 mmol), Na2CO3 (5.3 mg, 0.050 mmol) and DMSO (0.5 ml). The reaction mixture was stirred at 60° C. over-night. After simple work-up, the crude was purified by silica gel chromatography to afford the title product (9.2 mg). (MS: [M+1]+ 454)


Step 6: 3-(((S)-1-(7,8-dichloro-4-((R)-3-hydroxypyrrolidin-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoic acid. The procedure was the same as that in the synthesis of I-665. (MS: [M+1]+454)


Example 136: Synthesis of (S)-3-((1-(7,8-dichloro-4-(4-methyl-1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoic acid (I-825)



embedded image


Step 1: (S)-(1-(4,7,8-trichloroquinolin-2-yl)pyrrolidin-2-yl)methanol. The procedure was the same as that in the synthesis of I-287.


Step 2: (S)-(1-(7,8-dichloro-4-(4-methyl-1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methanol. The procedure was the same as that in the synthesis of I-821.


Step 3: tert-butyl (S)-3-((1-(7,8-dichloro-4-(4-methyl-1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoate. The procedure was the same as that in the synthesis of I-665.


Step 4: (S)-3-((1-(7,8-dichloro-4-(4-methyl-1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoic acid (I-825). To a vial were added tert-butyl (S)-3-((1-(7,8-dichloro-4-(4-methyl-1H-imidazol-1-yl)quinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoate (13 mg, 0.0257 mmol) and HCl in 1,4-dioxane (4 N, 0.3 ml). The mixture was stirred at room temperature for 3 hours. All volatiles were removed under reduced pressure. After lyophilization, the tilte compound was obtained as HCl salt (11 mg). (MS: [M+1]+ 449)


Example 137: Synthesis of (±)3-(((2S,3R)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-(methoxycarbonyl)pyrrolidin-3-yl)methoxy)propanoic acid (I-813)



embedded image


Step 1: (±)(3aR,6aS)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)hexahydro-6H-furo[3,4-b]pyrrol-6-one. The procedure was the same as that in the synthesis of I-665.


Step 2: (±)methyl (2S,3R)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-3-(hydroxymethyl)pyrrolidine-2-carboxylate. The crude product in step 1 was treated with TEA (20 μl, 0.143 mmol) and methanol (0.5 ml) for 18 hours. After removing all volatiles, the crude was used in next step.


Step 3: (±)methyl (2S,3R)-3-((3-(tert-butoxy)-3-oxopropoxy)methyl)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)pyrrolidine-2-carboxylate. The procedure was the same as that in the synthesis of I-665.


Step 4: (±)3-(((2S,3R)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-2-(methoxycarbonyl)pyrrolidin-3-yl)methoxy)propanoic acid. The procedure was the same as that in the synthesis of I-665. The tilte compound was obtained as TFA salt (17 mg). (MS: [M+1]+ 493)


Example 138: Synthesis of(S)-3-((1-(4-(6-aminopyridin-3-yl)-7,8-dichloroquinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoic acid (I-822)



embedded image


Step 1: methyl (S)-3-((1-(4-(6-aminopyridin-3-yl)-7,8-dichloroquinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoate. To a flask were added methyl (S)-3-((1-(4,7,8-trichloroquinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoate (104 mg, 0.25 mmol), 1,4-dioxane (8.0 ml), 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2-amine (110 mg, 0.50 mmol), NaHCO3 (62 mg, 0.75 mmol), Pd(dppf)Cl2—CH2Cl2 (41 mg, 0.050 mmol) and H2O (1.0 ml). After degassed with N2 for 4 times, the reaction mixture was stirred at 100° C. for 6 hours. The reaction mixture was diluted with ethyl acetate, washed with H2O, brine and dried over Na2SO4. After concentration the crude was purified by silica gel column to afford the title product (90 mg).


Step 2: (S)-3-((1-(4-(6-aminopyridin-3-yl)-7,8-dichloroquinolin-2-yl)pyrrolidin-2-yl)methoxy)propanoic acid. The procedure was the same as that in the synthesis of I-636. The tilte compound was obtained as free acid. (MS: [M+1]+ 461)


The following compounds are prepared essentially by the same methods as for I-822.


















MS


Example
Starting Material
Structure
[M + 1]+



















I-823


embedded image




embedded image




embedded image


435





I-824


embedded image




embedded image




embedded image


435





I-826


embedded image




embedded image




embedded image


449









Example 139: Synthesis of 3-(2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)oxy)ethoxy)propanoic acid (I-828)



embedded image


Step 1: 2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)oxy)ethan-1-ol. The procedure was the same as that in the synthesis of I-277.


Step 2: tert-butyl 3-(2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)oxy)ethoxy)propanoate. The procedure was the same as that in the synthesis of I-665.


Step 3: 3-(2-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)oxy)ethoxy)propanoic acid. The procedure was the same as that in the synthesis of I-825. The title compound was obtained as HCl salt. (MS: [M+1]+ 396)


Example 140: Synthesis of 3-((3-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)benzyl)oxy)propanoic acid (I-840)



embedded image


Step 1: (3-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)phenyl)methanol. The procedure was the same as that in the synthesis of I-822.


Step 2: tert-butyl 3-((3-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)benzyl)oxy)propanoate. The procedure was the same as that in the synthesis of I-665.


Step 3: 3-((3-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)benzyl)oxy)propanoic acid. The procedure was the same as that in the synthesis of I-825. The title compound was obtained as HCl salt. (MS: [M+1]+ 442)


The following compounds are prepared essentially by the same methods as for I-840.


















MS


Example
Starting Material
Structure
[M + 1]+



















I-841


embedded image




embedded image




embedded image


442





I-877


embedded image




embedded image




embedded image


456





I-878


embedded image




embedded image




embedded image


476





I-879


embedded image




embedded image




embedded image


476





I-880


embedded image




embedded image




embedded image


456





I-884


embedded image




embedded image




embedded image


472









Example 141: Synthesis of 5-chloro-2-(3-((7,8-dichloro-4-(1H-imidazol-1-yl) quinolin-2-yl)(isopropyl)amino) propoxy) benzoic acid (I-848)



embedded image


Step 1: 3-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(isopropyl)amino)propan-1-ol. To a mixture of 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (1.074 g), 3-(isopropylamino)propan-1-ol (0.53 g) and Na2CO3 (0.96 g) was added DMSO (3.4 mL). The resultant suspended solution was heated at 105° C. overnight. After cooling down to room temperature, the reaction mixture was diluted with water (5 mL) and extracted with EtOAc (20 mL). The isolated organic layer was washed with brine and dried over anhydrous Na2SO4. After concentration under reduced pressure, the residues were purified by silica gel column chromatography, eluting with a gradient of hexane and EtOAc (5% MeOH), to afford the tile compound (654 mg) as light brown solid. MS: [M+1]+ 379.


Step 2: 3-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl(isopropyl)amino)propyl 4-methylbenzenesulfonate. To a suspension of 3-((7,8-dichloro-4-(1H-imidazol-1-yl) quinolin-2-yl) (isopropyl)amino)propan-1-ol (654 mg), DMAP (45 mg) and TsCl (228 mg) in DCM (10 mL) was added TEA (0.3 mL). After the reaction mixture was stirred over 3 hours, it was diluted with DCM (20 mL). The resultant organic solution was washed with water (10 mL) and brine (10 mL) and dried over anhydrous Na2SO4. Concentration under reduced pressure afforded the crude title compound (610 mg), which was used in the next step without further purification. MS: [M+1]+ 533.


Step 3: methyl 5-chloro-2-(3-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(isopropyl)amino)propoxy)benzoate. To a solution of 3-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl(isopropyl)amino)propyl 4-methylbenzenesulfonate (32 mg) and methyl 5-chloro-2-hydroxybenzoate (13.5 mg) in anhydrous DMF (0.5 mL) was added anhydrous K2CO3 (10 mg). After the reaction mixture was stirred over 3 h at 55° C., it was diluted with water (4 mL) and EtOAc (15 mL). The organic layer was isolated, washed with brine (10 mL) and dried over anhydrous Na2SO4. Concentration under reduced pressure and purification by silicagel column chromatography, eluting with a gradient of hexane and EtOAc (5% MeOH), afforded the tile compound (24 mg). MS: [M+1]+ 547.


Step 4: 5-chloro-2-(3-((7,8-dichloro-4-(1H-imidazol-1-yl) quinolin-2-yl) (isopropyl)amino) propoxy) benzoic acid (I-848). To a suspension of methyl 5-chloro-2-(3-((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)(isopropyl)amino)propoxy)benzoate (24 mg) in MeOH (0.4 mL) and water (0.1 mL) was added LiOH·H2O (10.2 mg). After stirring over 1 h, THF (0.2 mL) was added. A clear solution was afforded after the reaction mixture was stirred overnight. The reaction progress was monitored by LC/MS. After concentration under reduced pressure removed the volatiles, the residues were suspended in water (0.2 mL) and treated with HOAc (0.015 mL). After sonicating over 5 mm, the solid was collected by centrifuge and lyophilized to afford the tile compound (13 mg). MS: [M+1]+ 533.


The following compounds were prepared essentially by the same methods as described above for I-848.


















MS


Example
Starting Material
Structure
[M + 1]+



















I-849


embedded image




embedded image




embedded image


529





I-850


embedded image




embedded image




embedded image


517





I-851


embedded image




embedded image




embedded image


500





I-852


embedded image




embedded image




embedded image


499





I-847


embedded image




embedded image




embedded image


505









Example 142: Synthesis of (S)-3-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-oxopyrrolidin-2-yl)methoxy)propanoic acid (I-812)



embedded image


Step 1: (S)-1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-(hydroxymethyl)pyrrolidin-2-one. To a mixture of 2,7,8-trichloro-4-(1H-imidazol-1-yl)quinoline (200 mg), bis(dibenzylideneacetone)palladium(0) (20 mg), Rac-BINAP (14 mg), (S)-5-(hydroxymethyl)pyrrolidin-2-one (92 mg) and Cs2CO3 (441 mg) were added anhydrous dioxane (6 mL) under N2. The resultant mixtures were degassed three cycles via vacuum and purging with N2. The resultant mixture was heated and stirred at 100° C. overnight under N2. After the aqueous work-up with EtOAc, the isolated organic layer was dried over anhydrous. Na2SO4. After concentration under reduced pressure, the residues were purified by silica gel column chromatography, eluting with a gradient of hexane and EtOAc (5% MeOH), to afford the title compound (28 mg). MS: [M+1]+ 377.


Step 2: tert-butyl (S)-3-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-oxopyrrolidin-2-yl)methoxy)propanoate. Following the same preparation procedure as described in step 3 of I-825, the tile compound was prepared. MS: [M+1]+ 505.


Step 3: (S)-3-((1-(7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)-5-oxopyrrolidin-2-yl)methoxy)propanoic acid (I-812). Following the same preparation procedure as described in step 4 of I-825, the tile compound was prepared. MS: [M+1]+ 449.


Example 143: Synthesis of methyl 2-(3-(((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)methyl)-5-fluorophenyl)acetate (I-890) and 2-(3-(((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)methyl)-5-fluorophenyl)acetic acid (I-891)



embedded image


Step 1: methyl 2-(3-fluoro-5-methylphenyl)acetate. To a solution of 2-(3-fluoro-5-methylphenyl)acetic acid (336 mg, 2 mmol) in MeOH (4 mL) and DMF (0.01 mL) was added SOCl2 (0.22 mL, 3 mmol) dropwise at 0° C. The solution was warmed to r.t. and stirred for 2 h. After evaporation, the reaction was quenched by NaHCO3 (sat., 2 mL) and extracted by EtOAc (5 mL×2). The organic phase was collected and evaporated to give crude product which was used directly for next step without purification.


Step 2: methyl 2-(3-(bromomethyl)-5-fluorophenyl)acetate. To a solution of crude from step 1 in CCl4 (5 mL) was added NBS (392 mg, 2.2 mmol) and benzoyl peroxide (10 mg). The mixture was stirred at 75° C. overnight. After cooling down to r.t., the solution was concentrated and purified by silica gel column to afford the title product (150 mg) as colorless oil.


Step 3: methyl 2-(3-(azidomethyl)-5-fluorophenyl)acetate. To a solution of methyl 2-(3-(bromomethyl)-5-fluorophenyl)acetate (50 mg, 0.19 mmol) in DMF (1 mL) was added NaN3 (19 mg, 0.29 mmol). The mixture was stirred at 75° C. overnight. After cooling down to r.t., the mixture was diluted by H2O (2 mL) and extracted by EtOAc (3 mL×2). The organic phase was collected and evaporated to give a crude product which was used directly for next step without purification.


Step 4: methyl 2-(3-(aminomethyl)-5-fluorophenyl)acetate. The Staudinger reaction is performed essentially the same as for I-757.


Step 5: methyl 2-(3-(((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)methyl)-5-fluorophenyl)acetate (I-890). I-890 is prepared essentially by the same methods as for I-664. MS: [M−1]457.


Step 6: 2-(3-(((7,8-dichloro-4-(1H-imidazol-1-yl)quinolin-2-yl)amino)methyl)-5-fluorophenyl)acetic acid (I-891). I-891 is prepared essentially by the same methods as for I-714. MS: [M−1]443.


Example 144: Biology

Expression and Purification of Recombinant cGAS Protein: cDNA encoding full-length or amino acids 147-520 of human cGAS was inserted into a modified pET28a vector containing an in-frame His6-SUMO tag. The E. coli strain BL21/pLys harboring the plasmid was induced with 0.5 mM IPTG at 18° C. overnight. His6-SUMO tag was removed by a SUMO protease digestion following purification of the His6-SUMO-cGAS as described previously (Sun et al, 2013, Science 339, 786).


In vitro inhibition assay of cGAS activity: A 60 μL mixture containing 20 mM Tirs-Cl, pH 7.5, 5 mM MgCl2, 0.2 mg/mL BSA, 0.01 mg/mL Herring testis DNA, 6.6 μM ATP, 0.1 mM GTP, 1.5 μg/mL of recombinant human cGAS (aa147-522)] and serial dilutions of a test compound in DMSO was added to a 96-well plate and incubated at 37° C. for 20 minutes. At the end of reaction, 40 μL of KinaseGlo Max (Promega) was added and chemiluminescence was measured with a luminometer. Inhibitory effect of a compound is evaluated by plotting percentage of ATP consumption against logarithm of compound concentrations. IC50 value was calculated using GraphPad Prism 8 (GraphPad Software, Inc.).


The results of the cGAS inhibition assay for compounds of the disclosure are presented in Table 2 and Table 3. The letter codes for cGAS IC50 include: A (<2 μM); B (2-30 μM); C (>30 μM).









TABLE 2







cGAS Inhibition Results










I-#
IC50







I-1
B



I-2
B



I-3
C



I-4
B



I-5
B



I-6
A



I-7
B



I-8
B



I-9
C



 I-10
B



 I-11
B



 I-12
C



 I-13
C



 I-14
C



 I-15
A



 I-16
C



 I-17
B



 I-18
B



 I-19
A



 I-20
A



 I-21
A



 I-22
A



 I-23
A



 I-24
A



 I-25
C



 I-26
A



 I-27
C



 I-28
A



 I-29
A



 I-30
B



 I-31
C



 I-32
A



 I-33
B



 I-34
C



 I-35
B



 I-36
C



 I-37
C



 I-38
C



 I-39
A



 I-40
C



 I-41
A



 I-42
A



 I-43
A



 I-44
C



 I-45
B



 I-46
A



 I-47
A



 I-48
A



 I-49
C



 I-50
A



 I-51
C



 I-52
C



 I-53
B



 I-54
B



 I-55
A



 I-56
B



 I-57
B



 I-58
C



 I-59
C



 I-60
C



 I-61
C



 I-62
C



 I-63
C



 I-64
C



 I-65
C



 I-66
C



 I-67
B



 I-68
C



 I-69
C



 I-70
C



 I-71
C



 I-72
C



 I-73
C



 I-74
C



 I-75
C



 I-76
B



 I-77
C



 I-78
C



 I-79
A



 I-80
A



 I-81
A



 I-82
A



 I-83
B



 I-84
B



 I-85
A



 I-86
A



 I-87
B



 I-88
B



 I-89
B



 I-90
C



 I-91
A



 I-92
A



 I-93
A



 I-94
A



 I-95
B



 I-96
A



 I-97
B



 I-98
B



 I-99
B



 I-100
A



 I-101
A



 I-102
B



 I-103
B



 I-104
B



 I-105
B



 I-106
B



 I-107
C



 I-108
C



 I-109
C



 I-110
A



 I-111
A



 I-112
A



 I-113
A



 I-114
B



 I-115
A



 I-116
A



 I-117
B



 I-118
A



 I-119
B



 I-120
A



 I-121
B



 I-122
B



 I-123
B



 I-124
A



 I-125
A



 I-126
B



 I-127
B



 I-128
B



 I-130
B



 I-131
B



 I-132
B



 I-133
A



 I-134
B



 I-135
C



 I-136
B



 I-137
B



 I-138
A



 I-139
A



 I-140
B



 I-141
A



 I-142
A



 I-143
A



 I-144
A



 I-145
A



 I-147
A



 I-148
A



 I-149
A



 I-150
A



 I-151
A



 I-152
B



 I-153
B



 I-154
A



 I-155
B



 I-156
A



 I-157
B



 I-158
A



 I-159
A



 I-160
A



 I-161
C



 I-162
C



 I-163
C



 I-164
B



 I-165
A



 I-166
B



 I-167
A



 I-168
A



 I-169
A



 I-170
A



 I-171
C



 I-172
B



 I-173
B



 I-174
A



 I-175
A



 I-176
B



 I-177
B



 I-178
B



 I-179
B



 I-180
B



 I-181
C



 I-182
B



 I-183
A



 I-184
A



 I-185
B



 I-186
A



 I-187
B



 I-188
B



 I-189
B



 I-190
A



 I-191
A



 I-192
A



 I-193
B



 I-194
A



 I-195
A



 I-196
A



 I-197
A



 I-198
A



 I-199
B



 I-200
A



 I-201
A



 I-202
A



 I-203
A



 I-204
A



 I-205
A



 I-206
B



 I-207
A



 I-208
B



 I-209
C



 I-210
B



 I-211
B



 I-212
B



 I-213
B



 I-214
B



 I-215
B



 I-216
B



 I-217
C



 I-218
A



 I-219
A



 I-220
B



 I-221
A



 I-222
A



 I-223
A



 I-224
A



 I-225
A



 I-226
B



 I-227
C



 I-228
C



 I-229
C



 I-230
B



 I-231
B



 I-232
B



 I-234
A



 I-235
A



 I-236
B



 I-237
B



 I-238
A



 I-239
A



 I-240
A



 I-241
A



 I-242
A



 I-243
A



 I-244
A



 I-245
A



 I-246
B



 I-247
B



 I-248
A



 I-249
A



 I-250
A



 I-251
C



 I-252
B



 I-253
A



 I-254
A



 I-255
A



 I-256
A



 I-257
A



 I-258
A



 I-259
C



 I-260
A



 I-261
C



 I-262
A



 I-263
C



 I-264
A



 I-265
C



 I-266
C



 I-267
C



 I-268
C



 I-269
C



 I-270
A



 I-271
B



 I-272
B



 I-273
B



 I-274
B



 I-275
C



 I-276
A



 I-277
B



 I-278
B



 I-279
C



 I-280
C



 I-281
B



 I-282
B



 I-283
C



 I-284
C



 I-285
A



 I-286
A



 I-287
A



 I-288
A



 I-289
B



 I-290
B



 I-291
B



 I-292
B



 I-293
B



 I-294
C



 I-295
C



 I-296
C



 I-297
C



 I-298
C



 I-299
C



 I-300
C



 I-301
C



 I-302
C



 I-303
C



 I-304
C



 I-305
C



 I-306
C



 I-307
C



 I-308
C



 I-309
C



 I-310
C



 I-311
C



 I-312
C



 I-313
C



 I-314
C



 I-315
C



 I-316
C



 I-317
C



 I-318
C



 I-319
C



 I-320
A



 I-321
A



 I-322
A



 I-323
A



 I-324
A



 I-325
A



 I-326
A



 I-327
A



 I-328
A



 I-329
A



 I-330
A



 I-331
B



 I-332
A



 I-333
C



 I-334
C



 I-335
A



 I-336
B



 I-337
A



 I-338
A



 I-339
A



 I-340
A



 I-341
A



 I-342
A



 I-343
B



 I-344
A



 I-345
A



 I-346
A



 I-347
A



 I-348
C



 I-349
A



 I-350
A



 I-351
A



 I-352
B



 I-353
C



 I-354
C



 I-355
B



 I-356
C



 I-357
B



 I-358
B



 I-359
B



 I-360
A



 I-361
B



 I-362
C



 I-363
B



 I-364
B



 I-365
C



 I-366
B



 I-367
C



 I-368
B



 I-369
A



 I-370
A



 I-371
A



 I-372
A



 I-373
A



 I-374
A



 I-375
B



 I-376
C



 I-377
C



 I-378
A



 I-379
B



 I-380
B



 I-381
C



 I-382
B



 I-383
C



 I-384
B



 I-385
B



 I-386
A



 I-387
A



 I-388
B



 I-389
A



 I-390
B



 I-391
A



 I-392
B



 I-393
C



 I-394
C



 I-395
B



 I-396
A



 I-397
A



 I-398
B



 I-399
B



 I-400
B



 I-401
C



 I-402
B



 I-403
C



 I-404
C



 I-405
B



 I-406
B



 I-407
C



 I-408
C



 I-409
C



 I-410
C



 I-411
C



 I-412
B



 I-413
C



 I-414
C



 I-415
C



 I-416
C



 I-417
C



 I-418
C



 I-419
C



 I-420
C



 I-421
B



 I-422
C



 I-423
A



 I-424
B



 I-425
C



 I-426
C



 I-427
A



 I-428
A



 I-429
A



 I-430
A



 I-431
A



 I-432
B



 I-433
A



 I-434
B



 I-435
A



 I-436
B



 I-437
A



 I-438
B



 I-439
B



 I-443
A



 I-444
A



 I-445
B



 I-446
B



 I-447
C



 I-448
A



 I-449
A



 I-450
B



 I-451
B



 I-452
B



 I-453
C



 I-454
A

















TABLE 3







cGAS Inhibition Results










I-#
IC50







I-455
A



I-456
C



I-457
B



I-458
B



I-459
B



I-460
B



I-461
A



I-462
B



I-463
B



I-464
A



I-465
A



I-466
A



I-467
B



I-468
B



I-469
A



I-470
A



I-471
B



I-472
B



I-473
A



I-474
B



I-475
B



I-476
B



I-477
A



I-478
A



I-479
A



I-480
A



I-481
A



I-482
A



I-483
B



I-484
B



I-485
A



I-486
A



I-487
A



I-488
A



I-489
A



I-490
A



I-491
A



I-492
B



I-493
A



I-494
A



I-495
A



I-496
B



I-497
B



I-498
B



I-499
A



I-500
B



I-501
A



I-502
B



I-503
A



I-504
A



I-505
B



I-506
B



I-507
A



I-508
A



I-509
A



I-510
A



I-511
A



I-512
A



I-513
A



I-514
A



I-515
A



I-516
A



I-517
A



I-518
A



I-519
A



I-520
A



I-521
A



I-522
B



I-524
A



I-525
A



I-526
A



I-527
A



I-528
C



I-529
A



I-530
A



I-531
A



I-532
B



I-533
A



I-534
B



I-535
B



I-536
A



I-537
B



I-538
A



I-539
A



I-540
A



I-541
A



I-542
B



I-543
B



I-544
B



I-545
A



I-546
B



I-547
B



I-548
A



I-549
A



I-550
B



I-551
C



I-552
A



I-553
A



I-554
B



I-555
A



I-556
A



I-557
A



I-558
A



I-559
A



I-560
A



I-561
A



I-562
A



I-563
A



I-564
A



I-565
A



I-566
A



I-567
A



I-568
B



I-569
A



I-570
B



I-571
A



I-572
A



I-573
B



I-574
B



I-575
B



I-576
A



I-577
B



I-578
B



I-579
C



I-580
C



I-581
B



I-582
A



I-583
B



I-584
B



I-585
B



I-586
B



I-587
B



I-588
A



I-589
B



I-590
B



I-591
B



I-592
B



I-593
B



I-594
B



I-595
B



I-596
B



I-597
B



I-598
B



I-599
B



I-600
B



I-601
B



I-602
B



I-603
A



I-604
B



I-605
C



I-606
B



I-607
A



I-608
B



I-609
C



I-610
A



I-611
C



I-612
B



I-613
A



I-614
B



I-615
B



I-616
A



I-617
A



I-618
A



I-619
B



I-620
B



I-621
B



I-622
A



I-623
B



I-624
A



I-625
A



I-626
A



I-627
A



I-628
A



I-629
A



I-630
A



I-631
A



I-632
A



I-633
A



I-634
A



I-635
A



I-636
A



I-637
A



I-638
B



I-639
A



I-640
A



I-641
A



I-642
B



I-643
A



I-644
B



I-645
B



I-646
B



I-647
B



I-648
B



I-649
B



I-650
B



I-651
A



I-652
B



I-653
A



I-654
A



I-655
A



I-656
C



I-657
A



I-658
A



I-659
A



I-660
B



I-661
A



I-662
B



I-663
A



I-664
A



I-665
A



I-666
A



I-667
A



I-668
A



I-669
B



I-670
A



I-671
A



I-672
A



I-673
B



I-674
A



I-675
A



I-676
A



I-677
A



I-678
A



I-679
A



I-680
B



I-681
B



I-682
A



I-683
A



I-684
A



I-685
A



I-686
B



I-687
B



I-688
A



I-689
A



I-690
A



I-691
A



I-692
A



I-693
A



I-694
A



I-695
B



I-696
A



I-697
A



I-698
A



I-699
A



I-700
A



I-701
A



I-702
A



I-703
A



I-704
A



I-705
A



I-706
A



I-707
A



I-708
A



I-709
A



I-710
A



I-711
A



I-712
A



I-713
A



I-714
A



I-715
A



I-716
A



I-717
A



I-718
B



I-719
A



I-720
B



I-721
B



I-722
A



I-723
A



I-724
A



I-725
A



I-726
A



I-727
A



I-728
A



I-729
A



I-730
A



I-731
A



I-732
A



I-733
A



I-734
A



I-735
A



I-736
A



I-737
A



I-738
A



I-739
A



I-740
A



I-741
A



I-742
A



I-743
A



I-744
A



I-745
A



I-746
A



I-747
A



I-748
A



I-749
A



I-750
A



I-751
A



I-752
A



I-753
C



I-754
B



I-755
C



I-756
B



I-757
B



I-758
B



I-759
A



I-760
A



I-761
A



I-762
A



I-763
A



I-764
A



I-765
B



I-766
A



I-767
B



I-768
B



I-769
B



I-770
A



I-771
B



I-772
B



I-773
A



I-774
A



I-775
A



I-776
A



I-778
B



I-779
B



I-780
B



I-781
A



I-782
A



I-783
A



I-784
A



I-785
A



I-786
A



I-787
A



I-788
A



I-789
A



I-790
B



I-791
B



I-792
A



I-793
A



I-794
A



I-795
A



I-796
A



I-797
A



I-798
A



I-799
A



I-800
A



I-801
A



I-802
A



I-803
A



I-804
A



I-805
A



I-806
A



I-807
A



I-808
A



I-809
A



I-810
A



I-811
A



I-812
A



I-813
A



I-814
A



I-815
A



I-816
A



I-817
A



I-818
B



I-819
A



I-820
A



I-821
C



I-822
C



I-823
C



I-824
C



I-825
C



I-826
C



I-827
C



I-828
B



I-829
A



I-830
A



I-831
A



I-832
A



I-833
A



I-834
A



I-835
A



I-836
A



I-837
A



I-838
A



I-839
A



I-840
A



I-841
A



I-842
A



I-843
A



I-844
A



I-845
A



I-846
A



I-847
B



I-848
B



I-849
B



I-850
B



I-851
B



I-852
B



I-853
A



I-854
A



I-855
B



I-856
A



I-857
A



I-858
C



I-859
B



I-860
A



I-861
C



I-862
A



I-863
C



I-864
A



I-865
A



I-866
A



I-867
A



I-868
A



I-869
B



I-870
B



I-871
B



I-872
B



I-873
A



I-874
A



I-875
A



I-876
A



I-877
A



I-878
A



I-879
A



I-880
A



I-881
A



I-882
A



I-883
A



I-884
A



I-885
A



I-886
A



I-887
A



I-888
A



I-889
A



I-890
B



I-891
A










Cellular assay to measure cGAS activity: Reporter THP1 cell line harboring a gene encoding Gaussia Luciferase under the control of 5 tandem repeats of interferon-stimulated response elements (ISRE) fused to an ISG54 minimal promoter was used to test inhibition of cGAS activity by synthetic compounds in human cells. These cells were plated on 96-well plates at 0.3×106/well and incubated with serial dilutions of compounds or DMSO for 5 min, followed by transfection of 2 μg/mL of ISD (Interferon Stimulatory DNA, a 45 bp DNA oligo) or mock transfected using lipofectamine 2000 (Life Technology) method, according to manufacturer's instructions. 16 hours later, 15 μL of the media from each well was transferred to a new plate, 50 μL of solution containing 50 mM Hepes-NaOH, pH 6.5, 50 mM NaCl, 10 mM EDTA, 1 μM of coeleanterazine was added to each well and luminescence was measured immediately. Fold increase in luminescence compared to mock transfection was plotted against concentrations of each compound, and IC50 is calculated using Graphpad. To evaluate the specificity of a compound, the same procedure was performed except that cells were transfected with 2 μg/mL poly(I:C) or infected with Sendai Virus (SeV) at 50 Unit/mL, which are known to activate the RIG-I-MAVS pathway. A specific inhibitory compound should inhibit interferon induction by DNA but have minimal effect on poly(I:C) or Sendai virus induced interferon reporter gene expression.


The results of the cellular assay for compounds of the disclosure are presented in Table 4. The letter codes for cGAS IC50 include: A (<2 μM); B (2-10 μM); C (>10 μM).









TABLE 4







Cellular Assay Results










I-#
IC50







 I-15
C



 I-24
B



 I-39
B



 I-86
B



 I-92
C



 I-116
B



 I-139
B



 I-149
C



 I-200
A



 I-322
C



 I-428
B



 I-429
C



 I-435
B



 I-443
B



 I-448
C



 I-461
C



 I-467
B



 I-469
A



 I-470
B



 I-471
B



 I-473
A



 I-477
B



 I-478
B



 I-487
B



 I-507
B



 I-516
C



 I-532
B



 I-539
B



 I-556
A



 I-575
B



 I-576
A



 I-577
B



 I-578
B



 I-582
A



 I-583
A



 I-584
B



 I-585
A



 I-586
A



 I-591
B



 I-592
B



 I-593
B



 I-594
B



 I-595
B



 I-596
A



 I-597
C



 I-598
A



 I-599
A



 I-600
A



 I-601
A



 I-642
A



 I-645
A



 I-649
B



 I-650
B



 I-663
A



 I-664
A



 I-665
A



 I-666
A



 I-667
A



 I-668
A



 I-669
A



 I-670
A



 I-671
A



 I-672
A



 I-673
A



 I-674
B



 I-676
A



 I-677
A



 I-678
B



 I-679
B



 I-680
B



 I-681
A



 I-682
A



 I-683
A



 I-684
A



 I-685
A



 I-686
A



 I-687
C



 I-688
A



 I-689
C



 I-690
A



 I-691
A



 I-692
A



 I-693
C



 I-694
A



 I-695
B



 I-696
A



 I-698
C



 I-699
A



 I-700
B



 I-701
B



 I-702
B



 I-703
C



 I-704
A



 I-705
A



 I-706
A



 I-707
A



 I-708
A



 I-709
A



 I-710
B



 I-711
A



 I-712
A



 I-713
A



 I-714
B



 I-715
C



 I-716
C



 I-717
A



 I-718
A



 I-719
A



 I-720
A



 I-722
B



 I-723
B



 I-724
B



 I-725
C



 I-726
C



 I-728
B



 I-730
B



 I-732
A



 I-733
A



 I-734
A



 I-735
B



 I-736
A



 I-739
A



 I-740
C



 I-741
B



 I-742
C



 I-743
A



 I-744
A



 I-745
A



 I-747
A



 I-748
C



 I-749
A



 I-750
A



 I-751
B



 I-752
C



 I-753
C



 I-755
C



 I-756
C



 I-758
C



 I-759
C



 I-760
C



 I-761
C



 I-762
C



 I-764
C



 I-765
B



 I-773
A



 I-774
B



 I-775
B



 I-776
C



 I-778
C



 I-779
C



 I-780
C



 I-781
C



 I-782
C



 I-784
C



 I-785
C



 I-786
B



 I-787
B



 I-788
C



 I-789
A



 I-791
C



 I-792
A



 I-793
A



 I-794
A



 I-795
A



 I-796
A



 I-797
A



 I-798
A



 I-799
C



 I-800
A



 I-801
A



 I-802
A



 I-803
A



 I-804
A



 I-805
A



 I-806
B



 I-807
A



 I-808
A



 I-809
A



 I-810
A



 I-811
A



 I-813
C



 I-814
A



 I-815
B



 I-816
A



 I-817
C



 I-820
A



 I-828
C



 I-829
B



 I-830
A



 I-831
B



 I-832
A



 I-833
A



 I-834
A



 I-835
B



 I-836
A



 I-837
A



 I-838
C



 I-839
C



 I-840
A



 I-841
C



 I-842
B



 I-843
B



 I-844
A



 I-845
A



 I-846
A



 I-853
A



 I-854
C



 I-855
C



 I-856
B



 I-857
A



 I-860
C



 I-862
A



 I-864
C



 I-865
A



 I-866
B



 I-867
A



 I-868
B



 I-872
A



 I-875
A



 I-876
A



 I-877
A



 I-878
A



 I-879
A



 I-880
B



 I-882
C



 I-883
B



 I-884
A



 I-885
B



 I-886
A



 I-887
B



 I-888
B



 I-889
C



 I-890
A



 I-891
A










All publications, patents, patent applications and other documents cited in this application, including U.S. Provisional Appl. Nos. 63/074,446, 63/124,713, and 63/196,146, are hereby incorporated by reference in their entireties for all purposes to the same extent as if each individual publication, patent, patent application or other document were individually indicated to be incorporated by reference for all purposes.


While various specific embodiments have been illustrated and described, it will be appreciated that various changes can be made without departing from the spirit and scope of the claimed invention(s).

Claims
  • 1. A compound, wherein the compound is of formula I*:
  • 2. (canceled)
  • 3. The compound of claim 1, wherein R1 is optionally substituted imidazole.
  • 4. The compound of claim 1, wherein R1 is
  • 5. The compound of claim 1, wherein R1 is optionally substituted pyrrole.
  • 6. The compound of claim 1, wherein R1 is pyrazole.
  • 7. The compound of claim 1, wherein R1 is
  • 8. The compound of claim 1, wherein R1 is optionally substituted furan.
  • 9. The compound of claim 1, wherein R1 is optionally substituted pyridine.
  • 10. (canceled)
  • 11. The compound of claim 1, wherein at least one occurrence of R3 is halogen.
  • 12. The compound of claim 11, wherein at least one occurrence of R3 is chloro.
  • 13-14. (canceled)
  • 15. The compound of claim 1, wherein R2 is —NRR5.
  • 16. The compound of claim 1, wherein R2 is —OR.
  • 17. (canceled)
  • 18. The compound of claim 15, wherein R5 is —(CH2)2-4OH, —(CH2)2-4O(CH2)1-5CO2H, —(CH2)2-4O(CH2)1-5CO2C1-4alkyl, —(CH2)0-3CH(CH2OH)2, —(CH2)2-4OC1-4alkyl, —(CH2)1-4CO2H, —(CH2)1-4CO2C1-4alkyl, —(CH2)1-4CONH2, —(CH2)1-4CONHC1-4alkyl, —(CH2)1-4CON(C1-4alkyl)2, —(CH2)1-4C(O)NRa(CH2)1-4CO2H, —(CH2)1-4C(O)NRa(CH2)1-4CONH2, —(CH2)1-4C(O)NRa(CH2)1-4CONHC1-4alkyl, —(CH2)1-4C(O)NRa(CH2)1-4CON(C1-4alkyl)2, —(CH2)0-4NRaC(O)(CH2)1-5CO2H, —(CH2)0-4NRaC(O)(CH2)1-5CO2C1-4alkyl, —(CH2)2-4SO3H, —(CH2)2-4SO3C1-4alkyl, —(CH2)2-4SO2NH2, —(CH2)2-4SO2NHC1-4alkyl, —(CH2)2-4SO2N(C1-4alkyl)2, —(CH2)2-4OSO2NH2, —(CH2)2-4NRaSO2C1-4alkyl, —(CH2)2-4NRaSO3H, —(CH2)1-4OP(OH)2, —(CH2)1-4P(O)(OH)2, —(CH2)1-4P(O)(OH)(OC1-4alkyl), or —(CH2)2-4OP(O)(H)OH.
  • 19-20. (canceled)
  • 21. The compound of claim 1, wherein R2 is substituted phenyl.
  • 22. The compound of claim 1, wherein R2 is
  • 23-67. (canceled)
  • 68. The compound of claim 1, wherein each R6 is independently fluoro, —CN, methyl, —CF3, —CO2H, —NH2, —OH, —OC1-4alkyl, —C3-6cycloalkyl, ═CH2, ═O, tetrazolyl, imidazoyl, thiophenyl, 1,2,4-oxadiazol-3(2H)-onyl, morpholinyl, phenyl, —(CH2)1-4OC1-4alkyl, —(CH2)0-4O(CH2)1-5CO2H, —(CH2)0-4O(CH2)1-5CO2C1-4alkyl, —(CH2)1-4O(CH2)1-4CHFCO2H, —(CH2)1-4O(CH2)1-4CHFCO2C1-4alkyl, —(CH2)1-4OCH(CH3)(CH2)1-4CO2H, —(CH2)1-4OCH(CH3)(CH2)1-4CO2C1-4alkyl, —(CH2)1-4O(CH2)1-4CH(CH3)CO2H, —(CH2)1-4O(CH2)1-4CH(CH3)CO2C1-4alkyl, —CHOH(CH2)1-4O(CH2)1-5CO2H, —CHOH(CH2)1-4O(CH2)1-5CO2C1-4alkyl, —CHCF3(CH2)1-4O(CH2)1-5CO2H, —CHCF3(CH2)1-4O—(CH2)1-5CO2C1-4alkyl, —(CH2)1-4O(CH2)1-5CONRa2, —(CH2)1-4O(CH2)1-5CONRaOH, —(CH2)1-4O(CH2)1-5CONRaCN, —(CH2)1-4O(CH2)1-5P(O)OH2, —CH(OH)CF3, —COH(CF3)2, —(CH2)1-5CO2H, —(CH2)1-5CO2C1-4alkyl, —(CH2)0-4CHCHCO2H, —(CH2)0-4CHCHCO2C1-4alkyl, —CO2C1-4alkyl, —CO2C1-4haloalkyl, —OCOC1-4alkyl, —O(CH2)1-4CO2H, —O(CH2)1-4CO2C1-4alkyl, —OCONRa(CH2)0-4CO2H, —OCONRa(CH2)0-4CO2C1-4alkyl, —OCO—(N-morpholine), —CONH2, —CONHOH, —CONHOC1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4CO2H, —(CH2)0-4C(O)NRa(CH2)1-4CO2C1-4alkyl, —(CH2)0-4C(O)NRa(CH2)1-4(pyrrolid-2-one), —(CH2)0-4C(O)NRa(CH2)1-4(piperidin-2-one), —(CH2)0-4C(O)NRa(CH2)1-4C(OH)(CF3)2, —(CH2)0-4C(O)NRa(CH2)1-4CONH2, —(CH2)0-4CONRaSO2C1-4haloalkyl, —(CH2)0-4CO(N-pyrrolidine)-(CH2)0-4CO2H, —(CH2)0-4CO(N-pyrrolidine)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4(N-pyrazole)-(CH2)0-4CO2H, —(CH2)0-4CO(N-pyrazole)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4(N-pyrrole)-(CH2)0-4CO2H, —(CH2)0-4CO(N-pyrrole)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4—O(phenyl)-(CH2)0-4CO2H, —(CH2)0-4—O(phenyl)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4—O(cycloalkane)-(CH2)0-4CO2H, —(CH2)0-4—O(cycloalkane)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CO(N-pyrrolidine)-(CH2)0-4SO2NH2, —(CH2)0-4CO(N-pyrrolidine)-(tetrazole), —(CH2)0-4CO(N-piperdine)-(CH2)0-4CO2H, —(CH2)0-4CO(N-piperdine)-(CH2)0-4CO2C1-4alkyl, —(CH2)0-4CO(N-morpholine)-(CH2)0-4CO2H, —CONRa(bicyclo[1.1.1]pentane)-(CH2)0-4CO2H, —NH2, —(CH2)0-4NRa(CH2)1-4CO2H, —(CH2)0-4NRa(CH2)1-4CO2C1-4alkyl, —(CH2)0-4N[(CH2)1-4CO2C1-4alkyl]2, —(CH2)0-4NRaCO(isoxazole)-OH, —(CH2)0-4NRaSO2(benzene)-CO2H, —(CH2)0-4NRaSO2(CH2)1-4CO2H, —(CH2)0-4NRaSO2(CH2)1-4CO2C1-4alkyl, —(CH2)0-4NRaSO2(CH2)1-4(imidazolidine-2,4-dione), or —(CH2)0-4OP(O)(H)OH.
  • 69. (canceled)
  • 70. The compound of claim 1, wherein the compound is selected from:
  • 71. The compound of claim 1, wherein the compound is selected from:
  • 72. The compound of claim 1, wherein the compound is selected from:
  • 73. The compound of claim 1, wherein the compound is selected from:
  • 74. The compound of claim 1, wherein the compound is selected from:
  • 75-76. (canceled)
  • 77. A pharmaceutical composition comprising the compound claim 1.
  • 78. A method of antagonizing cyclic GMP-AMP synthase (cGAS) in a patient in need thereof, comprising administering an effective amount of the compound of claim 1.
  • 79. A method of treating an inflammatory, allergic, autoimmune, ocular inflammation, or neurodegenerative disease in a patient in need thereof, comprising administering an effective amount of the compound of claim 1.
  • 80-82. (canceled)
Provisional Applications (3)
Number Date Country
63196146 Jun 2021 US
63124713 Dec 2020 US
63074446 Sep 2020 US
Continuations (1)
Number Date Country
Parent 17446951 Sep 2021 US
Child 18334325 US