WO2021072330A1 - Galnac-tgfbr1 inhibitor conjugates for the treatment of liver diseases - Google Patents
Galnac-tgfbr1 inhibitor conjugates for the treatment of liver diseases Download PDFInfo
- Publication number
- WO2021072330A1 WO2021072330A1 PCT/US2020/055159 US2020055159W WO2021072330A1 WO 2021072330 A1 WO2021072330 A1 WO 2021072330A1 US 2020055159 W US2020055159 W US 2020055159W WO 2021072330 A1 WO2021072330 A1 WO 2021072330A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- alkyl
- independently selected
- occurrence
- salt
- optionally substituted
- Prior art date
Links
- 0 CCCNCC(C)(CN*C)CNC(C)C Chemical compound CCCNCC(C)(CN*C)CNC(C)C 0.000 description 11
- SOPDDXWAPBIEHX-QQZUBYLESA-N CCOC(C/C(/C(/N)=C/C=C\C(C)=C)=N\N(CCC1)C1=O)=O Chemical compound CCOC(C/C(/C(/N)=C/C=C\C(C)=C)=N\N(CCC1)C1=O)=O SOPDDXWAPBIEHX-QQZUBYLESA-N 0.000 description 1
- VYIRUPFDNYGERY-UHFFFAOYSA-N Cc1cccc(-c(nc2CNCCc3c(cccc4)c4ccc3)c(-c3c[n]4ncnc4cc3)[n]2N)n1 Chemical compound Cc1cccc(-c(nc2CNCCc3c(cccc4)c4ccc3)c(-c3c[n]4ncnc4cc3)[n]2N)n1 VYIRUPFDNYGERY-UHFFFAOYSA-N 0.000 description 1
- GJZPOUOCAKPIIE-UHFFFAOYSA-N Cc1cccc(-c2c(-c3cc4ncn[n]4cc3)[nH]c(CN3CC4C=CC=CC4CC3)n2)n1 Chemical compound Cc1cccc(-c2c(-c3cc4ncn[n]4cc3)[nH]c(CN3CC4C=CC=CC4CC3)n2)n1 GJZPOUOCAKPIIE-UHFFFAOYSA-N 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/16—Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/54—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
- A61K47/549—Sugars, nucleosides, nucleotides or nucleic acids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/56—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
- A61K47/61—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule the organic macromolecular compound being a polysaccharide or a derivative thereof
Definitions
- Fibrosis is the formation of excess fibrous connective tissue or scar tissue in an organ or tissue in a reparative or reactive process. Fibrosis can occur in many tissues within the body, typically as a result of inflammation or damage, which include the lungs, liver, heart, and brain. Scar tissue blocks arteries, immobilizes joints and damages internal organs, wreaking havoc on the body's ability to maintain vital functions. Every year, millions of people are hospitalized due to the damaging effects of fibrosis. However, current therapeutics for treating fibrotic diseases are lacking or have drawbacks.
- the present disclosure provides a conjugate of Formula (I): wherein Region G comprises at least one GalNAc moiety; Region L 3 is a connector that connects Region G to Region Inh; Region Inh comprises a TGF ⁇ R1 inhibitor; z is 1, 2, 3, 4, or 5; or a pharmaceutically acceptable salt thereof.
- the present disclosure relates to a pharmaceutical composition comprising a conjugate of Formula (I) as described herein and a pharmaceutically acceptable carrier.
- the present disclosure relates to a method for treating a disease mediated by TGF ⁇ R1 activity in subject in need thereof, such as liver cancer or liver fibrosis, comprising administering to the subject an effective amount of a conjugate of Formula (I) or a pharmaceutical composition comprising a conjugate of Formula (I) as described herein.
- the present disclosure provides a method for enhancing an immune response (e.g., an anti-cancer immune response) in a subject comprising administering to the subject an effective amount of a conjugate of Formula (I) or a pharmaceutical composition comprising a conjugate of Formula (I) as described herein.
- Region Inh comprises a TGF ⁇ R1 inhibitor
- Z is the residual portion of a released, cleavable linker or comprises a non-cleavable linker and a residual portion of a degraded Region G; or a pharmaceutically acceptable salt thereof.
- FIG. 1A-1C illustrate the activity of GalNAc ⁇ TGF ⁇ R1 inhibitor conjugates in a HEK293 cell reporter assay (ASGR-positive and ASGR-negative cells: Fig. 1A, Conjugate 3.1; Fig. 1B, Conjugate 3.2; Fig. 1C, Conjugate 3.3).
- ASGR-positive and ASGR-negative cells Fig. 1A, Conjugate 3.1; Fig. 1B, Conjugate 3.2; Fig. 1C, Conjugate 3.3.
- Activin receptor-like kinase 5 (ALK5), which is also commonly known as transforming growth factor beta receptor 1 (TGF ⁇ R1), is a serine/threonine kinase transmembrane receptor. It is a part of the TGF ⁇ signaling pathway and is involved in signal transduction from the cell surface to the cytoplasm. The TGF ⁇ signaling pathway regulates gene expression of genes involved in cellular processes such as differentiation, apoptosis, wound healing, and cell growth.
- TGF ⁇ R2 type II TGF ⁇ receptor
- Smad2 Mothers Against Decapentaplegic homolog 2
- Smad3 Mothers Against Decapentaplegic homolog 3
- ALK5 inhibitors and conjugates thereof may be useful for treatment and/or prevention, e.g., vaccination, of cancer, autoimmune diseases, inflammation, fibrosis, sepsis, allergy, asthma, graft rejection, graft-versus-host disease, immunodeficiencies, and infectious diseases.
- an ALK5 inhibitor has an IC 50 value of between 0.1 nM and 1000 nM, between 0.1 nm and 100 nM, or between 0.1 nM and 80 nM in an ALK5 enzyme inhibition assay and/or in a TGF ⁇ R1 reporter assay.
- Exemplary ALK5 enzyme inhibition and TGF ⁇ R1 reporter assays are as set forth in the example section.
- any concentration range, percentage range, ratio range, or integer range is to be understood to include the value of any integer within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an integer), unless otherwise indicated.
- any number range recited herein relating to any physical feature, such as polymer subunits, size or thickness are to be understood to include any integer within the recited range, unless otherwise indicated. It should be understood that the terms “a” and “an” as used herein refer to “one or more" of the enumerated components. The use of the alternative (e.g., "or”) should be understood to mean either one, both, or any combination thereof of the alternatives.
- the terms “include” and “comprise” are used synonymously.
- the phrase “at least one of” when followed by a list of items or elements refers to an open ended set of one or more of the elements in the list, which may, but does not necessarily, include more than one of the elements.
- a “TGF ⁇ R1 inhibitor” refers to a compound that reduces, minimizes, or inactivates activin receptor-linke kinase 5 (ALK5) activity (e.g., directly inhibiting serine/threonine kinase activity or indirectly inhibiting downstream TGF ⁇ -dependent signaling activity, such as SBE-mediated responsiveness to TGF ⁇ and SMAD proteins) by about 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% as compared to untreated ALK5.
- ALK5 activin receptor-linke kinase 5
- a “conjugate” refers to a GalNAc moiety covalently attached to at least one TGF ⁇ R1 inhibitor via a linker.
- a “liver cell” refers to any cell type associated with normal liver tissue.
- a liver cell can be a canalicular cell, a Kupffer cell, a hepatocyte, sinusoidal endothelial cell, or a stellate cell.
- an “immune cell” refers to a T cell, B cell, NK cell, NKT cell, or an antigen presenting cell.
- an immune cell is a T cell, B cell, NK cell, or NKT cell. In some embodiments, an immune cell is an antigen presenting cell. In some embodiments, an immune cell is not an antigen presenting cell.
- salt or “pharmaceutically acceptable salt” refers to salts derived from a variety of organic and inorganic counter ions well known in the art. Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids. Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
- Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like.
- Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
- Inorganic bases from which salts can be derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like.
- Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, specifically such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine.
- the pharmaceutically acceptable base addition salt is chosen from ammonium, potassium, sodium, calcium, and magnesium salts.
- C x-y when used in conjunction with a chemical moiety, such as alkyl, alkenyl, or alkynyl is meant to include groups that contain from x to y carbons in the chain.
- C 1-6 alkyl refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups that contain from 1 to 6 carbons.
- ⁇ C x-y alkylene ⁇ refers to a substituted or unsubstituted alkylene chain with from x to y carbons in the alkylene chain.
- ⁇ C 1-6 alkylene ⁇ may be selected from methylene, ethylene, propylene, butylene, pentylene, and hexylene, any one of which is optionally substituted.
- C x-y alkenyl and “C x-y alkynyl” refer to substituted or unsubstituted unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond, respectively.
- ⁇ C x-y alkenylene ⁇ refers to a substituted or unsubstituted alkenylene chain with from x to y carbons in the alkenylene chain.
- ⁇ C 2-6 alkenylene ⁇ may be selected from ethenylene, propenylene, butenylene, pentenylene, and hexenylene, any one of which is optionally substituted.
- An alkenylene chain may have one double bond or more than one double bond in the alkenylene chain.
- ⁇ C x-y alkynylene ⁇ refers to a substituted or unsubstituted alkynylene chain with from x to y carbons in the alkenylene chain.
- ⁇ C 2-6 alkenylene ⁇ may be selected from ethynylene, propynylene, butynylene, pentynylene, and hexynylene, any one of which is optionally substituted.
- An alkynylene chain may have one triple bond or more than one triple bond in the alkynylene chain.
- Alkyl refers to a monovalent hydrocarbon consisting solely of carbon and hydrogen, containing no unsaturation, and preferably having from one to twelve carbon atoms, for example, methyl, ethyl, propyl, isopropyl, butyl, and the like.
- an alkyl comprises one to five carbon atoms (i.e., C 1 -C 5 alkyl). In other embodiments, an alkyl comprises one to four carbon atoms (i.e., C 1 -C 4 alkyl). In other embodiments, an alkyl comprises one to three carbon atoms (i.e., C 1 -C 3 alkyl). In other embodiments, an alkyl comprises one to two carbon atoms (i.e., C 1 -C 2 alkyl). In other embodiments, an alkyl comprises one carbon atom (i.e., C 1 alkyl or methyl).
- an alkyl comprises five to eight carbon atoms (i.e., C 5 - C 8 alkyl). In other embodiments, an alkyl comprises two to five carbon atoms (i.e., C 2 -C 5 alkyl). In other embodiments, an alkyl comprises three to five carbon atoms (i.e., C 3 -C 5 alkyl). Unless stated otherwise specifically in the specification, an alkyl chain is optionally substituted by one or more substituents such as those substituents described herein.
- Alkylene refers to a divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing no unsaturation, and preferably having from one to twelve carbon atoms, for example, methylene, ethylene, propylene, butylene, and the like.
- the alkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond.
- the points of attachment of the alkylene chain to the rest of the molecule and to the radical group are through the terminal carbons respectively.
- an alkylene comprises one to five carbon atoms (i.e., C 1 -C 5 alkylene).
- an alkylene comprises one to four carbon atoms (i.e., C 1 -C 4 alkylene). In other embodiments, an alkylene comprises one to three carbon atoms (i.e., C 1 -C 3 alkylene). In other embodiments, an alkylene comprises one to two carbon atoms (i.e., C 1 -C 2 alkylene). In other embodiments, an alkylene comprises one carbon atom (i.e., C 1 alkylene). In other embodiments, an alkylene comprises five to eight carbon atoms (i.e., C 5 -C 8 alkylene). In other embodiments, an alkylene comprises two to five carbon atoms (i.e., C 2 -C 5 alkylene).
- an alkylene comprises three to five carbon atoms (i.e., C 3 -C 5 alkylene).
- an alkylene chain is optionally substituted by one or more substituents such as those substituents described herein.
- Alkenyl refers to a monovalent hydrocarbon chain consisting solely of carbon and hydrogen, containing at least one carbon-carbon double bond, and preferably having from two to twelve carbon atoms. The alkenyl chain is attached to the rest of the molecule through a single bond.
- an alkenyl comprises two to five carbon atoms (i.e., C 2 -C 5 alkenyl).
- an alkenyl comprises two to four carbon atoms (i.e., C 2 -C 4 alkenyl). In other embodiments, an alkenyl comprises two to three carbon atoms (i.e., C 2 -C 3 alkenyl). In other embodiments, an alkenyl comprises two carbon atom (i.e., C 2 alkenyl). In other embodiments, an alkenyl comprises five to eight carbon atoms (i.e., C 5 -C 8 alkenyl). In other embodiments, an alkenyl comprises three to five carbon atoms (i.e., C 3 -C 5 alkenyl).
- alkenyl chain is optionally substituted by one or more substituents such as those substituents described herein.
- “Alkenylene” refers to a divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one carbon-carbon double bond, and preferably having from two to twelve carbon atoms.
- the alkenylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. The points of attachment of the alkenylene chain to the rest of the molecule and to the radical group are through the terminal carbons respectively.
- an alkenylene comprises two to five carbon atoms (i.e., C 2 -C 5 alkenylene). In other embodiments, an alkenylene comprises two to four carbon atoms (i.e., C 2 -C 4 alkenylene). In other embodiments, an alkenylene comprises two to three carbon atoms (i.e., C 2 -C 3 alkenylene). In other embodiments, an alkenylene comprises two carbon atom (i.e., C 2 alkenylene). In other embodiments, an alkenylene comprises five to eight carbon atoms (i.e., C 5 -C 8 alkenylene).
- an alkenylene comprises three to five carbon atoms (i.e., C 3 -C 5 alkenylene).
- an alkenylene chain is optionally substituted by one or more substituents such as those substituents described herein.
- Alkynyl refers to a monovalent hydrocarbon chain consisting solely of carbon and hydrogen, containing at least one carbon-carbon triple bond, and preferably having from two to twelve carbon atoms. The alkynyl chain is attached to the rest of the molecule through a single bond.
- an alkynyl comprises two to five carbon atoms (i.e., C 2 -C 5 alkynyl).
- an alkynyl comprises two to four carbon atoms (i.e., C 2 -C 4 alkynyl). In other embodiments, an alkynyl comprises two to three carbon atoms (i.e., C 2 -C 3 alkynyl). In other embodiments, an alkynyl comprises two carbon atom (i.e., C 2 alkynyl). In other embodiments, an alkynyl comprises five to eight carbon atoms (i.e., C 5 -C 8 alkynyl). In other embodiments, an alkynyl comprises three to five carbon atoms (i.e., C 3 -C 5 alkynyl).
- alkynyl chain is optionally substituted by one or more substituents such as those substituents described herein.
- “Alkynylene” refers to a divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one carbon-carbon triple bond, and preferably having from two to twelve carbon atoms.
- the alkynylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. The points of attachment of the alkynylene chain to the rest of the molecule and to the radical group are through the terminal carbons respectively.
- an alkynylene comprises two to five carbon atoms (i.e., C 2 -C 5 alkynylene). In other embodiments, an alkynylene comprises two to four carbon atoms (i.e., C 2 -C 4 alkynylene). In other embodiments, an alkynylene comprises two to three carbon atoms (i.e., C 2 -C 3 alkynylene). In other embodiments, an alkynylene comprises two carbon atom (i.e., C 2 alkynylene). In other embodiments, an alkynylene comprises five to eight carbon atoms (i.e., C 5 -C 8 alkynylene).
- an alkynylene comprises three to five carbon atoms (i.e., C 3 -C 5 alkynylene).
- an alkynylene chain is optionally substituted by one or more substituents such as those substituents described herein.
- “Heteroalkyl” refers to a monovalent hydrocarbon chain including at least one heteroatom in the chain, containing no unsaturation, and preferably having from one to twelve carbon atoms and from one to 6 heteroatoms, e.g., ⁇ O ⁇ , ⁇ NH ⁇ , ⁇ S ⁇ . The heteroalkyl is attached to the rest of the molecule through a single bond.
- a heteroalkyl comprises one to five carbon atoms and from one to three heteroatoms. In other embodiments, a heteroalkyl comprises one to four carbon atoms and from one to three heteroatoms. In other embodiments, a heteroalkyl comprises one to three carbon atoms and from one to two heteroatoms. In other embodiments, a heteroalkyl comprises one to two carbon atoms and from one to two heteroatoms. In other embodiments, a heteroalkyl comprises one carbon atom and from one to two heteroatoms. In other embodiments, a heteroalkyl comprises five to eight carbon atoms and from one to four heteroatoms.
- a heteroalkyl comprises two to five carbon atoms and from one to three heteroatoms. In other embodiments, a heteroalkyl comprises three to five carbon atoms and from one to three heteroatoms. Unless stated otherwise specifically in the specification, a heteroalkyl chain is optionally substituted by one or more substituents such as those substituents described herein. “Heteroalkylene” refers to a divalent hydrocarbon chain including at least one heteroatom in the chain, containing no unsaturation, and preferably having from one to twelve carbon atoms and from one to 6 heteroatoms, e.g., ⁇ O ⁇ , ⁇ NH ⁇ , ⁇ S ⁇ .
- the heteroalkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond.
- the points of attachment of the heteroalkylene chain to the rest of the molecule and to the radical group are through the terminal atoms of the chain.
- a heteroalkylene comprises one to five carbon atoms and from one to three heteroatoms.
- a heteroalkylene comprises one to four carbon atoms and from one to three heteroatoms.
- a heteroalkylene comprises one to three carbon atoms and from one to two heteroatoms.
- a heteroalkylene comprises one to two carbon atoms and from one to two heteroatoms.
- a heteroalkylene comprises one carbon atom and from one to two heteroatoms. In other embodiments, a heteroalkylene comprises five to eight carbon atoms and from one to four heteroatoms. In other embodiments, a heteroalkylene comprises two to five carbon atoms and from one to three heteroatoms. In other embodiments, a heteroalkylene comprises three to five carbon atoms and from one to three heteroatoms. Unless stated otherwise specifically in the specification, a heteroalkylene chain is optionally substituted by one or more substituents such as those substituents described herein.
- Carbocycle refers to a saturated, unsaturated or aromatic ring in which each atom of the ring is carbon.
- Carbocycle includes 3- to 10- membered monocyclic rings, 6- to 12-membered bicyclic rings, and 6- to 12-membered bridged rings.
- Each ring of a bicyclic carbocycle may be selected from saturated, unsaturated, and aromatic rings.
- an aromatic ring e.g., phenyl
- a bicyclic carbocycle includes any combination of saturated, unsaturated and aromatic bicyclic rings, as valence permits.
- a bicyclic carbocycle includes any combination of ring sizes such as 4-5 fused ring systems, 5-5 fused ring systems, 5-6 fused ring systems, 6-6 fused ring systems, 5-7 fused ring systems, 6-7 fused ring systems, 5-8 fused ring systems, and 6-8 fused ring systems.
- Exemplary carbocycles include cyclopentyl, cyclohexyl, cyclohexenyl, adamantyl, phenyl, indanyl, and naphthyl.
- the term “unsaturated carbocycle” refers to carbocycles with at least one degree of unsaturation and excluding aromatic carbocycles. Examples of unsaturated carbocycles include cyclohexadiene, cyclohexene, and cyclopentene.
- aryl refers to an aromatic monocyclic or aromatic multicyclic hydrocarbon ring system. The aromatic monocyclic or aromatic multicyclic hydrocarbon ring system contains only hydrogen and carbon and from five to eighteen carbon atoms, where at least one of the rings in the ring system is aromatic, i.e., it contains a cyclic, delocalized (4n+2) ⁇ -electron system in accordance with the Hückel theory.
- aryl groups include, but are not limited to, groups such as benzene, fluorene, indane, indene, tetralin and naphthalene.
- cycloalkyl refers to a saturated ring in which each atom of the ring is carbon. Cycloalkyl may include monocyclic and polycyclic rings such as 3- to 10-membered monocyclic rings, 6- to 12-membered bicyclic rings, and 6- to 12-membered bridged rings. In certain embodiments, a cycloalkyl comprises three to ten carbon atoms. In other embodiments, a cycloalkyl comprises five to seven carbon atoms.
- the cycloalkyl may be attached to the rest of the molecule by a single bond.
- monocyclic cycloalkyls include, e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
- Polycyclic cycloalkyl radicals include, for example, adamantyl, norbornyl (i.e., bicyclo[2.2.1]heptanyl), decalinyl, 7,7 dimethyl bicyclo[2.2.1]heptanyl, and the like.
- halo or, alternatively, “halogen” or “halide,” means fluoro, chloro, bromo or iodo. In some embodiments, halo is fluoro, chloro, or bromo.
- haloalkyl refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, for example, trifluoromethyl, dichloromethyl, bromomethyl, 2,2,2-trifluoroethyl, 1-chloromethyl-2-fluoroethyl, and the like.
- heterocycle or “heterocyclyl” as used herein refers to a saturated, unsaturated or aromatic ring comprising one or more heteroatoms.
- heteroatoms include N, O, Si, P, B, and S atoms.
- Heterocycles include 3- to 10-membered monocyclic rings, 6- to 12- membered bicyclic rings, and 6- to 12-membered bridged rings.
- a bicyclic heterocycle includes any combination of saturated, unsaturated and aromatic bicyclic rings, as valence permits.
- an aromatic ring e.g., pyridyl
- a bicyclic heterocycle includes any combination of ring sizes such as 4-5 fused ring systems, 5-5 fused ring systems, 5-6 fused ring systems, 6-6 fused ring systems, 5-7 fused ring systems, 6-7 fused ring systems, 5-8 fused ring systems, and 6-8 fused ring systems.
- the term “unsaturated heterocycle” refers to heterocycles with at least one degree of unsaturation and excluding aromatic heterocycles. Examples of unsaturated heterocycles include dihydropyrrole, dihydrofuran, oxazoline, pyrazoline, and dihydropyridine.
- heteroaryl includes aromatic single ring structures, preferably 5- to 7- membered rings, more preferably 5- to 6-membered rings, whose ring structures include at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms.
- heteroaryl also includes polycyclic ring systems having two or more rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heteroaromatic, e.g., the other rings can be aromatic or non-aromatic carbocyclic, or heterocyclic.
- Heteroaryl groups include, for example, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, pyrazole, pyridine, pyrazine, pyridazine, and pyrimidine, and the like.
- the term “heterocycloalkyl” refers to a saturated ring with carbon atoms and at least one heteroatom. Exemplary heteroatoms include N, O, Si, P, B, and S atoms.
- Heterocycloalkyl may include monocyclic and polycyclic rings such as 3- to 10-membered monocyclic rings, 6- to 12- membered bicyclic rings, and 6- to 12-membered bridged rings.
- heteroatoms in the heterocycloalkyl radical are optionally oxidized.
- One or more nitrogen atoms, if present, are optionally quaternized.
- the heterocycloalkyl is attached to the rest of the molecule through any atom of the heterocycloalkyl, valence permitting, such as any carbon or nitrogen atoms of the heterocycloalkyl.
- heterocycloalkyl radicals include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thio
- substituted refers to moieties having substituents replacing a hydrogen on one or more carbons or substitutable heteroatoms, e.g., ⁇ NH ⁇ , of the structure. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, i.e., a compound which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc.
- substituted refers to moieties having substituents replacing two hydrogen atoms on the same carbon atom, such as substituting the two hydrogen atoms on a single carbon with an oxo, imino or thioxo group.
- substituted is contemplated to include all permissible substituents of organic compounds.
- the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds.
- the permissible substituents can be one or more and the same or different for appropriate organic compounds.
- the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms.
- Protecting group refers to a moiety, except alkyl groups, that when attached to a reactive group in a molecule masks, reduces or prevents that reactivity. Examples of protecting groups can be found in T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3.sup.rd edition, John Wiley & Sons, New York, 1999, and Harrison and Harrison et al., Compendium of Synthetic Organic Methods, Vols. 1-8 (John Wiley and Sons, 1971-1996), which are incorporated herein by reference in their entirety.
- Representative amino or amine protecting groups include, formyl, acyl groups (such as acetyl, trifluoroacetyl, and benzoyl), benzyl, alkoxycarbonyl (such as benzyloxycarbonyl (CBZ), and tert-butoxycarbonyl (Boc)), trimethyl silyl (TMS), 2-trimethylsilyl-ethanesulfonyl (SES), trityl and substituted trityl groups, allyloxycarbonyl, 9-fluorenylmethyloxycarbonyl (FMOC), nitro-veratryloxycarbonyl (NVOC), sulfonyl, and the like.
- acyl groups such as acetyl, trifluoroacetyl, and benzoyl
- benzyl alkoxycarbonyl (such as benzyloxycarbonyl (CBZ), and tert-butoxycarbonyl (Boc)
- TMS tri
- tautomer refers to a molecule wherein a proton shift from one atom of a molecule to another atom of the same molecule is possible.
- T he compounds presented herein, in certain embodiments, exist as tautomers. In circumstances where tautomerization is possible, a chemical equilibrium of the tautomers will exist. The exact ratio of the tautomers depends on several factors, including physical state, temperature, solvent, and pH. Some examples of tautomeric equilibrium include: .
- the compounds described herein may in some cases exist as diastereomers, enantiomers, or other stereoisomeric forms.
- the compounds presented herein include all diastereomeric, enantiomeric, and epimeric forms as well as the appropriate mixtures thereof. Separation of stereoisomers may be performed by chromatography or by forming diastereomers and separating by recrystallization, or chromatography, or any combination thereof. (Jean Jacques, Andre Collet, Samuel H. Wilen, “Enantiomers, Racemates and Resolutions”, John Wiley And Sons, Inc., 1981, herein incorporated by reference for this disclosure). Stereoisomers may also be obtained by stereoselective synthesis.
- the compounds disclosed herein, in some embodiments, are used in different enriched isotopic forms, e.g., enriched in the content of 2 H, 3 H, 14 and/or C.
- the compound is deuterated in at least one position.
- deuterated forms can be made by the procedure described in U.S. Patent Nos. 5,846,514 and 6,334,997.
- deuteration can improve the metabolic stability and or efficacy, thus increasing the duration of action of drugs.
- structures depicted herein are intended to include compounds which differ only in the presence of one or more isotopically enriched atoms.
- compounds having the present structures except for the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by 13 C- or 14 C-enriched carbon are within the scope of the present disclosure.
- the compounds of the present disclosure optionally contain unnatural proportions of atomic isotopes at one or more atoms that constitute such compounds.
- the compounds may be labeled with isotopes, such as for example, deuterium ( 2 H), tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C).
- isotopes such as for example, deuterium ( 2 H), tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C).
- Isotopic substitution with 2 H, 11 C, 13 C, 14 C, 15 C, 12 N, 13 N, 15 N, 16 N, 16 O, 17 O, 14 F, 15 F, 16 F, 17 F, 18 F, 33 S, 34 S, 35 S, 36 S, 35 Cl, 37 Cl, 79 Br, 81 Br, 125 I are all contemplated.
- the compounds disclosed herein have some or all of the 1 H atoms replaced with 2 H atoms.
- the methods of synthesis for deuterium-containing compounds are known in the art and include, by way of non-limiting example only, the following synthetic methods.
- Deuterium substituted compounds are synthesized using various methods such as described in: Dean, Dennis C.; Editor. Recent Advances in the Synthesis and Applications of Radiolabeled Compounds for Drug Discovery and Development. [In: Curr., Pharm. Des., 2000; 6(10)] 2000, 110 pp; George W.; Varma, Rajender S.
- parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
- phrases “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
- pharmaceutically acceptable excipient or “pharmaceutically acceptable carrier” as used herein means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
- materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide;
- a conjugate as described herein comprises a GalNAc moiety linked to a TGF ⁇ R1 inhibitor via a linker.
- the present disclosure provides a conjugate of Formula (I): (I) wherein Region G comprises at least one GalNAc moiety; Region L 3 is a connector that connects Region G to Region Inh; Region Inh comprises a TGF ⁇ R1 inhibitor; z is 1, 2, 3, 4, or 5; or a pharmaceutically acceptable salt thereof.
- Region G is as described herein.
- Region L 3 is as described herein.
- the TGF ⁇ R1 inhibitor is as described herein.
- z is 1. In some embodiments, z is 2. In some embodiments, z is 3.
- Region G/GalNAc Moiety A conjugate as described herein comprises Region G, which comprises at least one GalNAc moiety.
- Region G comprises a Display Element for display of the one or more GalNAc moieties. In some embodiments, Region G comprises 1, 2, or 3 GalNAc moieties. In some embodiments, Region G comprises two or three GalNAc moieties.
- SP is a spacer, wherein each SP is independently a heteroalkylene, heteroalkenylene, or heteroalkynylene
- n is 2 or 3. In some embodiments, n is 2. In some embodiments, n is 3. In some embodiments, when n is 1, the branched Display Element is absent.
- one or more SP units comprise an ethylene glycol or poly(ethylene glycol) region. In some embodiments, one or more SP units comprise an amide bond. In some embodiments, one or more SP units comprise an unsubstituted ethylene or propylene unit. In some embodiments, each SP comprises 5 to 20 components in the longest linear chain.
- each SP is ⁇ (CH 2 ) 2 ⁇ C(O) ⁇ NH ⁇ (CH 2 ) x ⁇ CH 2 ⁇ (where x is 1, 2, 3, 4, 5, or 6, or x is 1, 2, or 3, or x is 1, or x is 3, or x is 4, or x is 5); ⁇ CH 2 ⁇ (CH 2 ) a ⁇ CH 2 ⁇ NH ⁇ C(O) ⁇ (CH 2 ) b ⁇ , where a is 0, 1, 2, or 3 (or a is 1 or 2, or a is 1); and b is 1, 2, 3, 4, 5, or 6 (or b is 3, 4, or 5, or b is 5); ⁇ C(O) ⁇ NH ⁇ (CH 2 ) (a+2) ⁇ NH ⁇ C(O) ⁇ (CH 2 ) b ⁇ , where a is 0, 1, 2, or 3 (or a is 1) and b is 1, 2, 3, 4, 5, or 6 (or b is 3, 4, or 5, or b is 5); or ⁇ C(O) ⁇ NH ⁇ (CH 2 ) a
- the Display Element comprises a central atom unit and two or three Arm Elements (AE) attached to the central atom unit that connect DE to each SP.
- the central atom unit is methine (CH).
- the central atom unit is carbon (C).
- the central atom unit is nitrogen (N).
- the central atom unit i where the two nitrogen substituents and one carbon substituent are each an SP, and the other carbon substituent connects the central atom unit to the linker.
- each AE is independently ⁇ (CH 2 ) 1-3 O ⁇ , ⁇ (CH 2 ) 1-3 NR ⁇ , ⁇ NR ⁇ , ⁇ (CH 2 ) 1-3 ⁇ , or a bond, wherein each CH 2 is optionally substituted with one or more C 1-4 alkyl or fluoro substituents, and R is H or C 1-4 alkyl.
- DE is one of the following structures: . Representative structures of Formula (V) and representative Display Elements are described in U.S. Patent Nos. 8,450,467, 9,399,775, and 10,077,433, which are incorporated herein by reference.
- the Region G has the structure:
- Region G has the structure: where a is 0, 1, 2, or 3; and b is 1, 2, 3, 4, 5, or 6. In some embodiments, a is 1 or 2. In some embodiments, a is 1. In some embodiments, b is 3, 4, or 5. In some embodiments, b is 5. In some embodiments, Region G has the structure:
- each p is independently 2, 3, 4, 5, or 6. In some embodiments, each p is 2. In some embodiments, each p is 4. In some embodiments, each p is 6. TGF ⁇ R1 Inhibitors
- the conjugates described herein comprise a TGF ⁇ R1 inhibitor.
- the TGF ⁇ R1 inhibitor can provide a direct or indirect effect.
- the TGF ⁇ R1 inhibitor is not an siRNA, an antisense RNA, or other oligonucleotide.
- the TGF ⁇ R1 inhibitor is a non-naturally occurring small molecule.
- a “small molecule” is an organic compound with a molecular weight of less than 1500, or 1000, or 900, or 750, or 600, or 500 Daltons.
- a small molecule TGF ⁇ R1 inhibitor has an octanol-water partition coefficient (logP) in the range of 3 to 6, or from 4 to 5, or from 2 to 4. In some embodiments, a small molecule TGF ⁇ R1 inhibitor has a polar surface area of less than 200, or less than 150 ⁇ 2 . In some embodiments, a small molecule TGF ⁇ R1 inhibitor has not more than five, or not more than three, hydrogen bond donors, and not more than 10, or not more than three hydrogen bond acceptors. A small molecule TGF ⁇ R1 inhibitor is not a protein, a polysaccharide, or a nucleic acid.
- the TGF ⁇ R1 inhibitor is a compound of Formula (A-I): wherein one of M 1 and M 2 is and the other of M 1 and M 2 is selected from: R 1 and R 2 are, at each occurrence, independently selected from hydrogen, halogen, ⁇ OR 11 , ⁇ SR 11 , ⁇ N(R 11 ) 2 , ⁇ NO 2 , ⁇ CN, phenyl, and ⁇ C 1 -C 6 alkyl, wherein said ⁇ C 1 -C 6 alkyl is optionally substituted with one or more substituents independently selected from halogen, ⁇ OR 11 , ⁇ SR 11 , ⁇ S(O)R 10 , ⁇ S(O) 2 R 11 , ⁇ S(O) 2 N(R 11 ) 2 , ⁇ N(R 11 ) 2 , ⁇ C(O)R 10 , ⁇ C(O)N(R 11 ) 2 , ⁇ N(
- a compound represented by Formula (A-I) or a salt thereof wherein one of M 1 and M 2 is and the other of M 1 and M 2 is the remaining variables (e.g., R 1 -R 8 , R 10 , R 20 , R 40 , R 11 , R 21 , R 31 , R 41 , R 50 , R 51 , R 61 , Y, R X , R Y , Z 1 , Z 2 , Z 3 , Z 4 , n, m, s, and w) are as set forth in the first aspect.
- R 1 -R 8 , R 10 , R 20 , R 40 , R 11 , R 21 , R 31 , R 41 , R 50 , R 51 , R 61 , Y, R X , R Y , Z 1 , Z 2 , Z 3 , Z 4 , n, m, s, and w are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein one of M 1 and the other of M 1 and M 2 is the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) wherein one of one of M 1 and M 2 is and the other of M 1 and M 2 is ; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein one of one of M 1 and M 2 is and the other of M 1 and M 2 is ; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 and M 2 is as set forth in the fifth aspect and R 7 and R 8 are independently selected from hydrogen, halogen, C 1 -C 3 alkyl, -OH, OC 1 -C 3 alkyl, and -OC 1 -C 3 haloalkyl, or R 7 and R 8 join together with the atoms to which they are attached to form a C 5 -C 6 carbocycle or 5- or 6- membered heterocycle each of which is optionally substituted with one or more substituents independently selected from halogen, -OR 31 , -SR 31 , -N(R 31 ) 2 , and -C 1 -C 6 alkyl wherein said C 1 -C 6 alkyl is optionally substituted with one or more substituents independently selected from halogen, -OR 31 , -SR 31 , and -N(R 31 ) 2 ; and the remaining variables
- a seventh aspect disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M 1 and M 2 is as set forth in the fifth aspect and R 7 and R 8 are independently selected from hydrogen, halogen, C 1 -C 3 alkyl, -OH, OC 1 -C 3 alkyl, and -OC 1 -C 3 haloalkyl, or R 7 and R 8 join together with the atoms to which they are attached to form an unsubstituted C 5 -C 6 carbocycle or an unsubstituted 5- or 6- membered heterocycle; and the remaining variables are as set forth in the first aspect.
- R 7 and R 8 are independently selected from hydrogen, halogen, C 1 -C 3 alkyl, -OH, OC 1 -C 3 alkyl, and -OC 1 -C 3 haloalkyl, or R 7 and R 8 join together with the atoms to which they are attached to form an unsubstituted C 5 -C 6
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 and M 2 is as set forth in any one of aspects 5-7 wherein the 5- or 6- membered heterocycle of R 7 and R 8 is a 5- or 6- membered heterocycle contains one ring heteroatom selected from nitrogen contains one ring heteroatom selected from nitrogen; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 and M 2 is as set forth in any one of aspects 5-7 wherein R 7 and R 8 join together with the atoms to which they are attached to form a phenyl ring optionally substituted with one or more substituents independently selected from halogen, -OR 31 , -SR 31 , -N(R 31 ) 2 , and - C 1 -C 6 alkyl wherein said C 1 -C 6 alkyl is optionally substituted with one or more substituents independently selected from halogen, -OR 31 , -SR 31 , and -N(R 31 ) 2; and the remaining variables are as set forth in the first aspect.
- R 7 and R 8 join together with the atoms to which they are attached to form a phenyl ring optionally substituted with one or more substituents independently selected from halogen, -OR 31 , -SR 31 , -N(R 31 ) 2 , and
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 and M 2 is as set forth in any one of aspects 5-7 wherein R 7 and R 8 join together with the atoms to which they are attached to form an unsubstituted phenyl ring; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 and M 2 is as set forth in any one of aspects 5-7 wherein R 7 and R 8 are each hydrogen; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 and R 8 are as set forth in any one of aspects 1-11 and wherein m is 1 or 2 and R 3 is, at each occurrence, independently selected from halogen, -C 1 -C 3 alkyl, -C 1 -C 3 haloalkyl, -OH, OC 1 -C 3 alkyl, and -OC 1 -C 3 haloalkyl; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 and R 8 are as set forth in any one of aspects 1-11 and wherein m is 1 and R 3 is, at each occurrence, independently selected from halogen, -C 1 -C 3 alkyl, -C 1 -C 3 haloalkyl, -OH, and -OC 1 -C 3 alkyl; and the remaining variables are as set forth in the first aspect.
- a fourteenth aspect disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M 1 , M 2 , R 7 and R 8 are as set forth in any one of aspects 1-11 and wherein m is zero; and the remaining variables are as set forth in the first aspect.
- a fifteenth aspect disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein one of one of M 1 and M 2 is and the other of M 1 and M 2 is ; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 and M 2 is as set forth in aspect 15 wherein Z 1 , Z 2 , Z 3 , and Z 4 are -C(H); and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 and M 2 is as set forth in aspect 15 wherein Z 2 is N and Z 1 , Z 3 , and Z 4 are -C(H); and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 and M 2 are as set forth in aspect 15 wherein Z 1 is N and Z 2 , Z 3 , and Z 4 are -C(H); and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 2 , R 7 , R 8 , m, R 3 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1- 18 wherein M 1 is ; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , R 7 , R 8 , m, R 3 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1- 18 wherein M 2 is ; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, R 3 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-20 wherein R 5 is hydrogen, halogen, or C 1 -C 3 alkyl optionally substituted with halogen; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, R 3 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-20 wherein R 5 is hydrogen or C 1 -C 3 alkyl; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, R 3 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-20 and wherein R 5 is methyl; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein one of M 1 and M and the other of M 1 and M 2 is selected from: and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 is as set forth in aspect 24 and wherein M 2 is ; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-25 and wherein R 1 and R 2 are, at each occurrence, independently selected from hydrogen, halogen, -OR 11 , -SR 11 , -N(R 11 ) 2 , phenyl, and -C 1 -C 6 alkyl wherein said C 1 -C 6 alkyl is optionally substituted with one or more substituents independently selected from halogen, ⁇ OR 11 , -SR 11
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-25 and wherein R 1 and R 2 are independently selected at each occurrence from hydrogen, phenyl, and -C 1 -C 3 alkyl wherein said -C 1 -C 3 alkyl is optionally substituted with one or more substituents independently selected from halogen, -OR 11 , and -C(O)OR 11 ; and the remaining variables are as set forth in the first aspect.
- R 1 and R 2 are independently selected at each occurrence from hydrogen, phenyl, and -C 1 -C 3 alkyl wherein said -C 1 -C 3 alkyl is optionally substituted with one or more substituents independently selected from halogen, -OR 11 , and -C(O)OR 11 ; and
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-25 and wherein R 1 and R 2 are independently selected at each occurrence from hydrogen, -CH 3 , -CH 2 OH, CH 2 CO 2 CH 3 , and phenyl; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-25 and wherein R 1 and R 2 are each hydrogen; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, R 1 , R 2 , R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-29 and wherein s is zero; and the remaining variables are as set forth in the first aspect.
- a thirty-first aspect disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M 1 , M 2 , R 7 , R 8 , m, R 1 , R 2 , R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-29 and wherein s is one; and the remaining variables are as set forth in the first aspect.
- a thirty-second aspect disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M 1 , M 2 , R 7 , R 8 , m, R 1 , R 2 , R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-29 and wherein s is one and n is two or three; and the remaining variables are as set forth in the first aspect.
- a thirty-third aspect disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M 1 , M 2 , R 7 , R 8 , m, R 1 , R 2 , R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-29 and wherein s is zero or one and n is one; and the remaining variables are as set forth in the first aspect.
- M 1 , M 2 , R 7 , R 8 , m, R 1 , R 2 , R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-29 and wherein s is zero or one and n is one; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, R 1 , R 2 , R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-29 and wherein s is zero or one and n is two; and the remaining variables are as set forth in the first aspect.
- a thirty-fifth aspect disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M 1 , M 2 , R 7 , R 8 , m, R 1 , R 2 , R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-29 and wherein s is zero or one and n is three; and the remaining variables are as set forth in the first aspect.
- M 1 , M 2 , R 7 , R 8 , m, R 1 , R 2 , R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-29 and wherein s is zero or one and n is three; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, s, n R 1 , R 2 , R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-35 and wherein Y is selected from ⁇ O- and -N(R 9 )- and R 9 is, at each occurrence, independently selected from: hydrogen; and -C 1 -C 6 alkyl optionally substituted with one or more substituents independently selected from halogen, -OR 41 , -SR 41 , -S(O)R 40 , ⁇ S(O) 2 R 41 , -S(O) 2 N(R 41 ) 2 , -N(R 41 ) 2 , -C(O)R 40 , -C(O)N(R 41 ) 2 , -N
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, s, n R 1 , R 2 , R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-35 and wherein Y is selected from ⁇ O- and -N(R 9 )- and R 9 is, at each occurrence, independently selected from: hydrogen; and unsubstituted-C 1 -C 6 alkyl; and the remaining variables are as set forth in the first aspect.
- M 1 , M 2 , R 7 , R 8 , m, s, n R 1 , R 2 , R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-35 and wherein Y is selected from ⁇ O- and -N(R 9 )- and R 9 is, at each occurrence, independently
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, s, n R 1 , R 2 , R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-35 and wherein Y is selected from O, N(H), and N(Me); and unsubstituted ⁇ C 1 -C 6 alkyl; and the remaining variables are as set forth in the first aspect.
- M 1 , M 2 , R 7 , R 8 , m, s, n R 1 , R 2 , R 3 , R 5 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-35 and wherein Y is selected from O, N(H), and N(Me); and unsubstituted ⁇ C 1 -C 6 alkyl; and the remaining variables are as set forth in the first
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, s, n R 1 , R 2 , R 3 , R 5 , R 9 , Y, Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-38 and wherein w is zero; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, s, n R 1 , R 2 , R 3 , R 5 , R 9 , Y, Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-38 and wherein w is 1, 2, 3, 4, or 5; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, s, n R 1 , R 2 , R 3 , R 5 , R 9 , Y, Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-38 and wherein w is 1, 2, or 3; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, s, n R 1 , R 2 , R 3 , R 5 , R 9 , Y, Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-38 and wherein w is 1 or 2; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, s, n R 1 , R 2 , R 3 , R 5 , R 9 , Y, Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-38 having formula (A-IA), (A-IB), (A-IC), (A-ID) or (A-IE): or a pharmaceutically acceptable salt of any one of formula (A-IA), (A-IB), (A-IC), (A-ID) or (A-IE); and the remaining variables are as set forth in the first aspect.
- M 1 , M 2 , R 7 , R 8 , m, s, n R 1 , R 2 , R 3 , R 5 , R 9 , Y, Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-38 having formula (A
- a compound represented by Formula (A-I) wherein M 1 , M 2 , R 7 , R 8 , m, s, n R 1 , R 2 , R 3 , R 5 , R 9 , Y, Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-38 having formula (A-IC) or (A-ID) or a pharmaceutically acceptable salt of any one of formula (A-IC), or (A-ID); and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) wherein M 1 , M 2 , R 7 , R 8 , m, s, n R 1 , R 2 , R 3 , R 5 , R 9 , Y, Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-38 having formula (A-IF): wherein w is 1, 2, 3, or 4; or a pharmaceutically acceptable salt thereof); and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-IF) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, s, n, R 1 , R 2 , R 3 , R 5 , R 9 , Y, Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-38 and w is 2 or 3; and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-IF) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, s, n, R 1 , R 2 , R 3 , R 5 , R 9 , Y, Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-38 and w is 2; and the remaining variables are as set forth in the first aspect.
- R 6 does not comprise cyano.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, s, n, R 1 , R 2 , R 3 , R 5 , R 9 , R 21 , Y, Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-55 and is: , wherein represents the point of attachment to and the remaining variables are as set forth in the first aspect.
- a compound represented by Formula (A-I) or a salt thereof wherein M 1 , M 2 , R 7 , R 8 , m, R 3 , R 5 , R 9 , R 21 , Z 1 , Z 2 , Z 3 , and Z 4 are as set forth in any one of aspects 1-55 and wherein is: , wherein represents the point of attachment to and the remaining variables are as set forth in the first aspect.
- the TGF ⁇ R1 inhibitor is a compound from Table 14.
- the TGF ⁇ R1 inhibitor is a compound of Formula (B-I): wherein: M 1 and M 2 are independently selected from R 1 and R 2 are independently selected at each occurrence from: a halogen, -OR 10 , -SR 10 , -N(R 10 ) 2 , -C(O)R 10 , -C(O)N(R 10 ) 2 , -N(R 10 )C(O)R 10 , -C(O)OR 10 , ⁇ OC(O)R 10 , -S(O)R 10 , -S(O) 2 R 10 , -S(O) 2 N(R 10 ) 2 , -P(O)(OR 10 ) 2 , -OP(O)(OR 10 ) 2 , -NO 2 , and -CN; -C 1 -C 10 alkyl, -C 2 -C 10 alkenyl,
- M 1 and M 2 are and the other of M 1 and M 2 is .
- M 1 may be .
- the compound or salt is represented by Formula (B-Ia): (B-Ia); or a pharmaceutically acceptable salt thereof.
- M 2 is .
- the compound or salt is represented by Formula (B-Ib): (B-Ib); or a pharmaceutically acceptable salt thereof.
- M 1 is and M 2 is .
- M 1 is and M 2 is .
- the compound or salt is represented by Formula (B-Ic) or Formula (B-Id): (B-Ic) (B-Id) or a salt thereof.
- R 3 for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), and (B-Id) is selected from hydrogen and -C 1 -C 10 alkyl optionally substituted with one or more substituents independently selected from a halogen, -NO 2 , -CN, -OR 10 , -SR 10 , ⁇ N(R 10 ) 2 , - C(O)R 10 , -C(O)N(R 10 ) 2 , -N(R 10 )C(O)R 10 , -C(O)OR 10 , and -OC(O)R 10 .
- R 3 may be selected from hydrogen and -C 1 -C 10 alkyl optionally substituted with one or more substituents independently selected from a halogen, -NO 2 , -CN, -OR 10 , -SR 10 , and -N(R 10 ) 2 .
- R 3 is hydrogen.
- n is 0. In other embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), and (B-Id), n is selected from 1, 2, 3, and 4.
- each R 1 is independently selected from a halogen, -OR 10 , -SR 10 , -N(R 10 ) 2 , -C(O)R 10 , -C(O)N(R 10 ) 2 , ⁇ N(R 10 )C(O)R 10 , -C(O)OR 10 , -OC(O)R 10 , -S(O)R 10 , -S(O) 2 R 10 , -S(O) 2 N(R 10 ) 2 , -P(O)(OR 10 ) 2 , ⁇ OP(O)(OR 10 ) 2 , -NO 2 , and -CN; and -C 1 -C 10 alkyl, -C 2 -C 10 alkenyl, and -C 2 -C 10 al
- each R 1 is selected from a halogen, -OR 10 , -SR 10 , -N(R 10 ) 2 , -NO 2 , and -CN; and -C 1 -C 10 alkyl optionally substituted at each occurrence with one or more substituents independently selected from a halogen, -OR 10 , -SR 10 , -N(R 10 ) 2 , -C(O)R 10 , -NO 2 , and -CN.
- each R 1 is independently selected from a -C 1 -C 10 alkyl optionally substituted at each occurrence with one or more substituents independently selected from a halogen, -OR 10 , -SR 10 , - N(R 10 ) 2 , -C(O)R 10 , -NO 2 , and ⁇ CN.
- m is 0.
- each R 2 is independently selected from a halogen, -OR 10 , -SR 10 , -N(R 10 ) 2 , -C(O)R 10 , -C(O)N(R 10 ) 2 , ⁇ N(R 10 )C(O)R 10 , ⁇ C(O)OR 10 , ⁇ OC(O)R 10 , ⁇ S(O)R 10 , ⁇ S(O) 2 R 10 , ⁇ S(O) 2 N(R 10 ) 2 , ⁇ P(O)(OR 10 ) 2 , ⁇ OP(O
- each R 2 is independently selected from a halogen, ⁇ OR 10 , ⁇ SR 10 , ⁇ N(R 10 ) 2 , ⁇ NO 2 , and ⁇ CN; and ⁇ C 1 -C 10 alkyl optionally substituted at each occurrence with one or more substituents independently selected from a halogen, ⁇ OR 10 , ⁇ SR 10 , ⁇ N(R 10 ) 2 , ⁇ C(O)R 10 , ⁇ NO 2 , and ⁇ CN.
- each R 2 is independently selected from ⁇ OR 10 and ⁇ C 1 -C 10 alkyl optionally substituted with one or more substituents independently selected from a halogen, ⁇ OR 10 , ⁇ SR 10 , ⁇ N(R 10 ) 2 , ⁇ C(O)R 10 , ⁇ NO 2 , and ⁇ CN.
- m is 1 and R 2 is ⁇ CH 3 .
- M 2 may be .
- M 2 is a compound or salt of any of Formulas (B-I), (B- Ia), (B-Ib), (B-Ic), and (B-Id).
- the compound of Formula (B-I) is represented by Formula (B-Ie): (B-Ie); or a salt thereof.
- Q is selected from ⁇ (CR 10 2) p ⁇ , ⁇ (CR 10 2) q O(CR 10 2) q ⁇ , ⁇ (CR 10 2) q S(CR 10 2) q ⁇ , and ⁇ (CR 10 2) q NR 10 (CR 10 2) q ⁇ , where p is selected from 1, 2, 3, 4, and 5 and q is independently selected at each occurrence from 0, 1, 2, 3, 4, and 5.
- Q is selected from ⁇ (CR 10 2) p ⁇ .
- p may be 1 such that Q is ⁇ C(R 10 ) 2 ⁇ .
- Q may be selected from ⁇ (CR 10 2) q O(CR 10 2) q ⁇ , ⁇ (CR 10 2) q S(CR 10 2) q ⁇ , and ⁇ (CR 10 2) q NR 10 (CR 10 2) q ⁇ .
- Q is ⁇ CH 2 NH ⁇ .
- Q is selected from ⁇ (CR 10 2) q NR 10 (CR 10 2) q ⁇ .
- Q is selected from ⁇ (CR 10 2) q NR 10 ⁇ .
- Q is selected from ⁇ (CR 10 2) q NR 10 ⁇ .
- Q is ⁇ CR 10 2NR 10 (CR 10 2) 1-2 ⁇ .
- Q is ⁇ CH 2 NHCH 2 ⁇ or ⁇ CH 2 NHCH 2 CH 2 ⁇ .
- Q is ⁇ CH 2 NHCH 2 ⁇ .
- each R 10 is independently selected from hydrogen; and ⁇ C 1 -C 10 alkyl, ⁇ C 2 -C 10 alkenyl, and ⁇ C 2 -C 10 alkynyl, each of which is optionally substituted at each occurrence with one or more substituents independently selected from a halogen, ⁇ OH, ⁇ CN, ⁇ NO 2 , and ⁇ NH 2 .
- R 10 is hydrogen at each occurrence.
- each R 10 of Q is hydrogen at each occurence.
- T is selected from a saturated C 3 -C 7 carbocycle optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T may be a saturated C 3 carbocycle optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T may be selected from: , , , , and , each of which is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T is selected from , , , , , , and .
- T may be: .
- T is selected from a C 5-12 bicyclic carbocycle optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T is selected from a saturated C 5-12 bridged carbocycle optionally substituted with one or more substituents independently selected at each occurrence from R 13 , such as an optionally substituted C 5 bridged carbocycle.
- T may be selected from: , and , each of which is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T is selected from C 8-11 bicyclic carbocycle optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T is a fused bicyclic ring.
- T is selected from a C 8-11 fused bicyclic carbocycle. In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B- Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from:
- T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T may be selected from: , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T is , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T may be selected from: and .
- T may be selected from: and .
- T is selected from: , , , , , , , , , , , and .
- T is selected from naphthalene, 1,2,3,4-tetrahydronaphthalene, decahydronaphthalene, octahydro-1H-indene, 2,3-dihydro-1H-indene, 1H-indene, octahydropentalene, decahydro-1H-benzo[7]annulene, 7H-benzo[7]annulene, 4aH- benzo[7]annulene, 6,7,8,9-tetrahydro-5H-benzo[7]annulene, 2,3,4,5-tetrahydro-1H- benzo[7]annulene, 2,3,4,7-tetrahydro-1H-benzo[7]annulene, azu
- T is selected from a 4- to 12-membered heterocycle optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T is selected from:
- T is selected from: , , , , , , , , , , , , , , , , , , , , , , , , , , , and , any one of which is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T is selected from: , , , , , , , ,
- T is selected from a 7- to 12-membered bicyclic heterocycle optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T is selected from an 8- to 11-membered bicyclic heterocycle optionally substituted with one or more substituents independently selected at each occurrence from R 13 , such as an 8- to 11-membered bicyclic heteroaryl group.
- T is selected from: wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- ⁇ Q ⁇ T is selected from: wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- ⁇ Q ⁇ T is selected from In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T has one to four ring heteroatoms independently selected from N, O, S, and B. In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B- Ib), (B-Ic), (B-Id), and (B-Ie), T has at least one ring heteroatom that is boron.
- T may be selected from: wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T may be selected from , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T is represented by: , wherein dashed lines represent single or double bonds, valence permitting; k is selected from 0, 1, 2, and 3; and W, X, Y, and Z are independently selected from N(R 10 ) g and C(R 10 ) h , wherein g is selected from 0 and 1 and h is selected from 1 and 2, and wherein a straight line linked to a wavy line indicates connectivity to Q from any position, valence permitting, of the bicyclic heterocycle.
- T is represented by: .
- T is represented by: .
- T is selected from: , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T may be selected from: , , , , , , , , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T is selected from: , , , , , , , , , , , , , , , , , , , , , , , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T may be selected from: , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T may be selected from: , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T may be selected from: , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T may be selected from: , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T may be selected from: and .
- T may be selected from: .
- T may be selected from: , , , , , , , , and .
- T is selected from: , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , ,
- T is selected from: , , , , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T is selected from: , , , , , , , , , , , , , , , , , , , , , , , , , , and ,wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T is selected from: , , , , , , , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- T is selected from 1,2,3,4-tetrahydroquinoline, 1,2,3,4- tetrahydroisoquinoline, 1,2-dihydroquinoline, 1,2-dihydroisoquinoline, 1,2,3,4- tetrahydroquinazoline, decahydroquinoline, decahydroisoquinoline, quinoline, isoquinoline, quinoxaline, phthalazine, quinazoline, and cinnoline, wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 .
- each R 13 may be independently selected from a halogen, ⁇ OR 10 , ⁇ SR 10 , ⁇ N(R 10 ) 2 , ⁇ NO 2 , and ⁇ CN; and ⁇ C 1 -C 10 alkyl optionally substituted at each occurrence with one or more substituents independently selected from a halogen, ⁇ OR 10 , ⁇ SR 10 , ⁇ N(R 10 ) 2 , ⁇ C(O)R 10 , ⁇ NO 2 , and ⁇ CN.
- Q is selected from a bond, ⁇ (CR 10 2) p ⁇ , and ⁇ (CR 10 2) q NR 10 (CR 10 2) q ⁇ ;
- T is selected from an optionally substituted saturated C 3 -C 7 carbocycle, an optionally substituted C 5-12 bicyclic carbocycle, and an optionally substituted 4- to 12-membered heterocycle, wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 , wherein R 13 is independently selected at each occurrence from halogen, ⁇ OR 10 , ⁇ N(R 10 ) 2 , ⁇ C(O)R 10 , ⁇ C(O)OR 10 , ⁇ N(R 10 )C(O)R 10 , and ⁇ C 1 -C 3 alkyl optionally substituted with one or
- Q is selected from a bond, ⁇ (CR 10 2) p ⁇ , and ⁇ (CR 10 2) q NR 10 (CR 10 2) q ⁇ ;
- T is selected from an optionally substituted saturated C 3 -C 7 carbocycle, an optionally substituted C 5-12 bicyclic carbocycle, and an optionally substituted 4- to 12-membered heterocycle, wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 , wherein R 13 is independently selected at each occurrence from halogen, ⁇ OR 10 , ⁇ N(R 10 ) 2 , and ⁇ C 1 -C 3 alkyl optionally substituted with one or more substituents independently selected from a halogen, ⁇ OR 10 and ⁇ N(R 10 ) 2 ; and R 10 is as set forth
- Q is selected from a bond, ⁇ CH 2 ⁇ , ⁇ CH 2 NH ⁇ , ⁇ CH 2 NHCH 2 ⁇ , and ⁇ CH 2 NHCH 2 CH 2 ⁇ ; and T is selected from: , , , , , , , , , , , , , , , , , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 wherein R 13 is independently selected at each occurrence from halogen, ⁇ OR 10 , ⁇ N(R 10 ) 2 , ⁇ C(O)R 10 , ⁇ C(O)OR 10 , ⁇ N(R 10 )C(O)R 10 , and ⁇ C 1 -C 3 alkyl optionally substituted
- Q is selected from ⁇ CH 2 ⁇ , ⁇ CH 2 NH ⁇ , ⁇ CH 2 NHCH 2 ⁇ , and ⁇ CH 2 NHCH 2 CH 2 ⁇ ; and T is selected from: , , , , , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R 13 , wherein R 13 is independently selected at each occurrence from halogen, ⁇ OH, ⁇ NH 2 , and ⁇ C 1 -C 3 alkyl.
- the compound of Formula (B-I) is selected from:
- the compound of Formula (B-I) is selected from:
- the compound of Formula (B-I) is selected from the compounds in Table 15.
- TGF ⁇ R1 Inhibitors of Formula (C-I) is a compound of Formula (C-I): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein: L is ⁇ [CR′ 2 ]p ⁇ L’ ⁇ [CH 2 ]q ⁇ L’ is absent, ⁇ S ⁇ , ⁇ O ⁇ , or ⁇ NH ⁇ ; A is absent, carbocycle, or heterocycle; Q 3 is N or CR 3 ; Q 4 is N or CR 4 ; Q 5 is N or CR 5 ; Q 6 is N or CR 6 ; R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl; R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alk
- compounds are provided having the structure of Formula (C-I), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein L’ is absent. In other embodiments, L’ is ⁇ S ⁇ . In other embodiments, L’ is ⁇ O ⁇ . In still other embodiments, L’ is ⁇ NH ⁇ . In certain embodiments, compounds are provided having the structure of Formula (C-I), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein p is 1. In some embodiments, p is 2. In other embodiments, p is 3.
- compounds are provided having the structure of Formula (C-I), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein q is 0. In other embodiments, q is 1. In still other embodiments, q is 2. In further embodiments, q is 3.
- compounds are provided having the structure of Formula (C-II): (C-II) or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; Q 3 is N or CR 3 ; Q 4 is N or CR 4 ; Q 5 is N or CR 5 ; Q 6 is N or CR 6 ; R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10 ; R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy; R 3 is H, halo, C 1–3 alkyl, C 1–3 haloalkyl, or C 1–3 alkoxy; R 4 is H, halo, C 1–3 alkyl, C 1
- A is carbocycle or heterocycle
- R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10
- R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy
- R 6 is H, halo, C 1–3 alkyl, or C 1–3 alkoxy
- R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety
- R 9 is, at each occurrence, independently halo, ⁇ OR′, ⁇ SR′, ⁇ N(R′) 2 , ⁇ C(O)R′, ⁇ C(O)N(R′) 2 , ⁇ N(R
- A is carbocycle or heterocycle
- R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10
- R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy
- R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety
- R 9 is, at each occurrence, independently halo, ⁇ OR′, ⁇ SR′, ⁇ N(R′) 2 , ⁇ C(O)R′, ⁇ C(O)N(R′) 2 , ⁇ N(R′)C(O)R′ , ⁇ C(O)OR′, ⁇ OC(O)R
- A is carbocycle or heterocycle
- R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10
- R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy
- R 6 is H, halo, C 1–3 alkyl, or C 1–3 alkoxy
- R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety
- R 9 is, at each occurrence, independently halo, ⁇ OR′, ⁇ SR′, ⁇ N(R′) 2 , ⁇ C(O)R′, ⁇ C(O)N(R′) 2 , ⁇ N(R
- A is carbocycle or heterocycle
- Q 3 is N or CR 3
- Q 4 is N or CR 4
- Q 5 is N or CR 5
- Q 6 is N or CR 6
- R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10
- R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy
- R 3 is H, halo, C 1–3 alkyl, C 1–3 haloalkyl, or C 1–3 alkoxy
- R 4 is H, halo, C 1–3 alkyl, C 1–3 haloalkyl, or C 1–3 alkoxy
- R 5 is H, halo
- compounds having the structure of Formula (C-III-A): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10 ; R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy; R 6 is H, halo, C 1–3 alkyl, or C 1–3 alkoxy; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R 9 is, at each occurrence, independently halo, ⁇ OR′, ⁇ SR′, ⁇ N(R′) 2 , ⁇ C(O)R
- compounds are provided having the structure of Formula (C-III-B): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10 ; R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R 9 is, at each occurrence, independently halo, ⁇ OR′, ⁇ SR′, ⁇ N(R′)2, ⁇ C(O)R′, ⁇ C(O)N(R′) 2 , ⁇ N(R′)C(O)R
- compounds having the structure of Formula (C-III-C): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10 ; R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy; R 6 is H, halo, C 1–3 alkyl, or C 1–3 alkoxy; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R 9 is, at each occurrence, independently halo, ⁇ OR′, ⁇ SR′, ⁇ N(R′) 2 , ⁇ C(O)R
- A is carbocycle or heterocycle
- Q 3 is N or CR 3
- Q 4 is N or CR 4
- Q 5 is N or CR 5
- Q 6 is N or CR 6
- R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10
- R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy
- R 3 is H, halo, C 1–3 alkyl, C 1–3 haloalkyl, or C 1–3 alkoxy
- R 4 is H, halo, C 1–3 alkyl, C 1–3 haloalkyl, or C 1–3 alkoxy
- R 5 is H, halo
- compounds are provided having the structure of Formula (C-IV-A): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10 ; R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy; R 6 is H, halo, C 1–3 alkyl, or C 1–3 alkoxy; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R 9 is, at each occurrence, independently halo, ⁇ OR′, ⁇ SR′, ⁇ N(R′) 2 , ⁇ C(O)R
- compounds are provided having the structure of Formula (C-IV-B): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10 ; R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R 9 is, at each occurrence, independently halo, ⁇ OR′, ⁇ SR′, ⁇ N(R′)2, ⁇ C(O)R′, ⁇ C(O)N(R′) 2 , ⁇ N(R′)C(O)R
- compounds are provided having the structure of Formula (C-IV-C): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10 ; R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy; R 6 is H, halo, C 1–3 alkyl, or C 1–3 alkoxy; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R 9 is, at each occurrence, independently halo, ⁇ OR′, ⁇ SR′, ⁇ N(R′) 2 , ⁇ C(O)R
- A is carbocycle or heterocycle
- Q 3 is N or CR 3
- Q 4 is N or CR 4
- Q 5 is N or CR 5
- Q 6 is N or CR 6
- R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10
- R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy
- R 3 is H, halo, C 1–3 alkyl, C 1–3 haloalkyl, or C 1–3 alkoxy
- R 4 is H, halo, C 1–3 alkyl, C 1–3 haloalkyl, or C 1–3 alkoxy
- R 5 is H, halo
- compounds are provided having the structure of Formula (C-V-A): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10 ; R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy; R 6 is H, halo, C 1–3 alkyl, or C 1–3 alkoxy; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R 9 is, at each occurrence, independently halo, ⁇ OR′, ⁇ SR′, ⁇ N(R′) 2 , ⁇ C(O)R
- compounds are provided having the structure of Formula (C-V-B): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10 ; R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R 9 is, at each occurrence, independently halo, ⁇ OR′, ⁇ SR′, ⁇ N(R′)2, ⁇ C(O)R′, ⁇ C(O)N(R′) 2 , ⁇ N(R′)C(O)R
- compounds are provided having the structure of Formula (C-V-C): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10 ; R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy; R 6 is H, halo, C 1–3 alkyl, or C 1–3 alkoxy; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R 9 is, at each occurrence, independently halo, ⁇ OR′, ⁇ SR′, ⁇ N(R′) 2 , ⁇ C(O)R
- compounds having the structure of Formula (C-VI): (C-VI) or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: Q 3 is N or CR 3 ; Q 4 is N or CR 4 ; Q 5 is N or CR 5 ; Q 6 is N or CR 6 ; R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10 ; R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy; R 3 is H, halo, C 1–3 alkyl, C 1–3 haloalkyl, or C 1–3 alkoxy; R 4 is H, halo, C 1–3 alkyl, C 1–3 haloalkyl, or C
- compounds having the structure of Formula (C-VI-A): (C-VI-A) or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10 ; R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy; R 6 is H, halo, C 1–3 alkyl, or C 1–3 alkoxy; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R 9 is, at each occurrence, independently halo, ⁇ OR′, ⁇ SR′, ⁇ N(R′) 2 , ⁇ C(O)R′,
- compounds having the structure of Formula (C-VI-B): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10 ; R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R 9 is, at each occurrence, independently halo, ⁇ OR′, ⁇ SR′, ⁇ N(R′) 2 , ⁇ C(O)R′, ⁇ C(O)N(R′) 2 , ⁇ N(R′)C(O)R′ , ⁇ C(C(O)R′
- R 1 is hydrogen, C 1–3 alkyl, or C 1–3 haloalkyl, wherein R 1 is substituted with 0–3 R 10 ;
- R 2 is, at each occurrence, independently halo, C 1–3 alkyl, C 1-3 alkoxy, C 1–3 haloalkyl, or C 1-3 haloalkoxy;
- R 6 is H, halo, C 1–3 alkyl, or C 1–3 alkoxy;
- R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety;
- R 9 is, at each occurrence, independently halo, ⁇ OR′, ⁇ SR′, ⁇ N(R′) 2 , ⁇ C(O)R′, ⁇ C(O)N(R′) 2 , ⁇ N(R′)C(O)R′
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein R 1 is hydrogen.
- R 1 is unsubstituted C 1–3 alkyl.
- R 1 is methyl.
- R 1 is C 1–3 alkyl substituted with 1–3 R 10 .
- R 1 is ⁇ CH 2 OCH 3 .
- R 1 is unsubstituted C 1–3 haloalkyl.
- R 1 is ⁇ CF 3 .
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein m is 0. In other embodiments, m is 1–3. In certain embodiments, m is 1.
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein at least one R 2 is halo. In some embodiments, at least one R 2 is F.
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein A is heterocycle.
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein A is carbocycle.
- A is phenyl.
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein n is 0. In some embodiments, n is 1–5. In other embodiments, n is 1 or 2.
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein at least one R 9 is halo. In some embodiments, at least one R 9 is F or Cl.
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein at least one R 9 is ⁇ OR′.
- R′ of the at least one ⁇ OR′ is C 1–6 alkyl.
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein at least one R 9 is C 1–4 alkyl. In some embodiments, at least one R 9 is methyl.
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein at least one R 9 is ⁇ CN.
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein at least one R 9 is ⁇ C(O)N(R′) 2 . In some embodiments, at least one R 9 is ⁇ C(O)N(CH 3 ) 2 .
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein at least one R 9 is ⁇ N(R′) 2 . In some embodiments, at least one R 9 is ⁇ N(CH 3 ) 2 .
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein at least one R 9 is ⁇ NO 2 .
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein R 6 is H.
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein R 6 is halo. In some embodiments, R 6 is F or Cl.
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein R 6 is C 1–3 alkyl. In some embodiments, R 6 is methyl.
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein R 6 is C 1–3 alkoxy. In some embodiments, R 6 is ⁇ OCH 3 .
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein the reactive moiety capable of attachment to a linker or the reactive moiety capable of attachment to an antibody is ⁇ Y ⁇ (CR 11 R 12 ) p ⁇ NHR 13 , ⁇ Y ⁇ (CR 11 R 12 ) p ⁇ C(O)OH, or ⁇ Y ⁇ (CR 11 R 12 ) p ⁇ C(O)NHR 13 , wherein: Y is ⁇ O ⁇ , ⁇ CH 2 ⁇ , ⁇ OCH 2 CH 2 ⁇ , or ⁇ OCH 2 CH 2 NR 14 ⁇ ; R 11 is at each occurrence, independently, H, halogen, C 1–3 alkyl, or C 1–3 haloalkyl; R 12 is at each occurrence
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein R 7 is ⁇ Y ⁇ (CR 11 R 12 ) p ⁇ NHR 13 .
- Y is .
- Y is ⁇ CH 2 ⁇ .
- Y is ⁇ OCH 2 CH 2 ⁇ .
- Y is ⁇ OCH 2 CH 2 NR 14 ⁇ .
- compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein R 7 is:
- Representative compounds of Formula (C-I), and Formulas (C-II) through (C-VI) as applicable, include the compounds listed in Tables 16–20 below, as well as pharmaceutically acceptable salts thereof.
- TGF ⁇ R1 Inhibitors of Formula (D-I) is a compound of Formula (D-I): (D-I) or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: Q A is CR A or N; Q B is CR B ; Q 3 is N or CR 3 ; Q 4 is N or CR 4 ; Q 5 is N or CR 5 ; R A is H, halo, C 1-3 alkyl, or C 1-3 haloalkyl and R B is B R , together with the atoms to which they are attached, form a heterocyclic ring; R a and R b are each H, or R a and R b , together with the atoms to which they are attached, form a
- compounds are provided having the structure of Formula (D-II): (D-II) or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: Q 3 is N or CR 3 ; Q 4 is N or CR 4 ; Q 5 is N or CR 5 ; Q 6 is N or CR 6 ; R a and R b are each H, or R a and R b , together with the atoms to which they are attached, form a heterocyclic ring; R 1 is H, C 1-3 alkyl, or C 1-3 haloalkyl; R 2 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, or C 1-3 haloalkoxy; R 3 is H, halo, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, or C 1-3 haloalkoxy; R 3 is
- Q 6 is CR 6 when R 1 is methyl and R a and R b , together with the atoms to which they are attached, form a heterocyclic ring.
- compounds are provided having the structure of Formula (D-III): (D-III) or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: Q 6 is N or CR 6 ; R 1 is H, C 1-3 alkyl, or C 1-3 haloalkyl; R 2 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, or C 1-3 haloalkoxy; R 6 is H, halo, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, or C 1-3 haloalkoxy; wherein R 1 , R 2 , and R 6 are, at each occurrence, independently
- Q 6 is CR 6 when R 1 is methyl.
- compounds are provided having the structure of Formula (D-IV): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: Q 6 is N or CR 6 ; R 1 is H, C1-3 alkyl, or C1-3 haloalkyl; R 2 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, or C 1-3 haloalkoxy; R 6 is H, halo, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, or C 1-3 haloalkoxy; wherein R 1 , R 2 , and R 6 are each substituted with 0–3 R 10 ; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is H. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is C 1-3 alkyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is methyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, R 1 is ethyl or propyl.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, R 1 is C 1-3 haloalkyl.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is ⁇ CFH 2 , ⁇ CF 2 H, or ⁇ CF 3 .
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein m is 0.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein m is 1–3.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein Q 6 is N.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein Q 6 is CR 6 .
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 6 is H.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 6 is halo, C 1-3 alkyl, or C 1-3 haloalkyl.
- TGF ⁇ R1 inhibitor is a compound having the structure of Formula (D-V): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: R a and R b are each H, or R a and R b , together with the atoms to which they are attached, form a heterocyclic ring; Q A is CR A or N; Q 3 is N or CR 3 ; Q 4 is N or CR 4 ; Q 5 is N or CR 5 ; R A is H, halo, C 1-3 alkyl, or C 1-3 haloalkyl; R a and R b are each H, or R a and R b , together with the atoms to which they are attached, form a heterocyclic ring; ring B is carbocycle or heterocycle; R 1 is H, C 1-3 alkyl, or C 1-3 haloalkyl; R 2 is, at each
- TGF ⁇ R1 inhibitor is a compound having the structure of Formula (D-VI): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: Q A is CR A or N; R A is H, halo, C 1-3 alkyl, or C 1-3 haloalkyl; ring B is carbocycle or heterocycle; R 1 is H, C 1-3 alkyl, or C 1-3 haloalkyl; R 2 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, or C 1-3 haloalkoxy; wherein R 1 and R 2 are, at each occurrence, independently substituted with 0-3 R 10 ; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R 9 is, at each occurrence, independently substituted with 0-3 R
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is heterocycle. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is heteroaryl.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, isoxazolyl, oxazolyl, isothiazolyl, thiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, or pyrazinyl.
- compounds having the structure of Formula (D-VI-A): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: Q A is CR A or N; R A is H, halo, C 1-3 alkyl, or C 1-3 haloalkyl; R 1 is H, C 1-3 alkyl, or C 1-3 haloalkyl; R 2 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, or C 1-3 haloalkoxy; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; wherein R 1 and R 2 are, at each occurrence, independently substituted with 0-3 R 10 ; R 9 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 haloalky
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is carbocycle.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is aryl.
- compounds having the structure of Formula (D-VI-B): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: Q A is CR A or N; R A is H, halo, C 1-3 alkyl, or C 1-3 haloalkyl; R 1 is H, C 1-3 alkyl, or C 1-3 haloalkyl; R 2 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, or C 1-3 haloalkoxy; wherein R 1 and R 2 are, at each occurrence, independently substituted with 0-3 R 10 ; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R 9 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 haloalky
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein Q A is N. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein Q A is CR A . In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R A is H. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R A is halo.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R A is F.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R A is C 1-3 alkyl, or C 1-3 haloalkyl.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is H.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is C 1-3 alkyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is methyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is ethyl or propyl.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is C 1-3 haloalkyl.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is ⁇ CFH 2 , ⁇ CF 2 H, or ⁇ CF 3 .
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein m is 0.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein m is 1–3. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein n is 0. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein n is 1–2.
- TGF ⁇ R1 inhibitor is a compound having the structure of Formula (D-VII): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: Q A is CR A or N; R A is H, halo, C 1-3 alkyl, or C 1-3 haloalkyl; ring B is carbocycle or heterocycle; R 1 is H, C 1-3 alkyl, or C 1-3 haloalkyl; R 2 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, or C 1-3 haloalkoxy; wherein R 1 and R 2 are, at each occurrence, independently substituted with 0-3 R 10 ; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R 9 is, at each occurrence, independently substituted with 0-3
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: R 1 is not H or methyl when ring B is carbocycle; ring B is a five membered heteroaryl when R 1 is H and Q A is N; and R 7 is substituted with an acidic amino acid sidechain when R 1 is methyl and Q A is CR A .
- compounds having the structure of Formula (D-VIII): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: R A is H, halo, C 1-3 alkyl, or C 1-3 haloalkyl; ring B is carbocycle or heterocycle; R 1 is H, ethyl, propyl, or C 1-3 haloalkyl; R 2 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, or C 1-3 haloalkoxy; wherein R 1 and R 2 are, at each occurrence, independently substituted with 0-3 R 10 ; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R 9 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 haloalkyl;
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is heterocycle. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is heteroaryl.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, isoxazolyl, oxazolyl, isothiazolyl, thiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, or pyrazinyl.
- compounds are provided having the structure of Formula (D-VIII-A):
- R A is H, halo, C 1-3 alkyl, or C 1-3 haloalkyl
- R 1 is H, ethyl, propyl, or C 1-3 haloalkyl
- R 2 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, or C 1-3 haloalkoxy; wherein R 1 and R 2 are, at each occurrence, independently substituted with 0-3 R 10
- R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety
- R 9 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy
- R 10 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is carbocycle.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is aryl.
- compounds are provided having the structure of Formula (D-VIII-B):
- R A is H, halo, C 1-3 alkyl, or C 1-3 haloalkyl
- R 1 is ethyl, propyl, or C 1-3 haloalkyl
- R 2 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, or C 1-3 haloalkoxy; wherein R 1 and R 2 are, at each occurrence, independently substituted with 0-3 R 10
- R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety
- R 9 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy
- R 10 is, at each occurrence,
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R A is H. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R A is halo. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R A is F.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R A is C 1-3 alkyl, or C 1-3 haloalkyl.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is H.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is ethyl or propyl.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is C 1-3 haloalkyl.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is ⁇ CFH 2 , ⁇ CF 2 H, or ⁇ CF 3 .
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein m is 0.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein m is 1–3. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein n is 0. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein n is 1–2.
- compounds are provided having the structure of Formula (D-IX): or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: ring B is carbocycle or heterocycle; R 1 is H, C 1-3 alkyl, or C 1-3 haloalkyl; R 2 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, or C 1-3 haloalkoxy; wherein R 1 and R 2 are, at each occurrence, independently substituted with 0-3 R 10 ; R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R 9 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy; R 10 is, at each
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is heteroaryl.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is a 5-membered heteroaryl.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, isoxazolyl, oxazolyl, isothiazolyl, thiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, or pyrazinyl.
- compounds are provided having the structure of Formula (D-IX-A):
- R 1 is H, C 1-3 alkyl, or C 1-3 haloalkyl
- R 2 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, or C 1-3 haloalkoxy
- R 1 and R 2 are, at each occurrence, independently substituted with 0-3 R 10
- R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety
- R 9 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy
- R 10 is, at each occurrence, independently C1-3 alkoxy or C1-3 haloalkoxy
- m is 0–3
- n is
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is carbocycle.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is aryl.
- compounds are provided having the structure of Formula (D-IX-B):
- R 1 is ethyl, propyl, or C 1-3 haloalkyl
- R 2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C 1-3 haloalkoxy; wherein R 1 and R 2 are, at each occurrence, independently substituted with 0-3 R 10
- R 7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety
- R 9 is, at each occurrence, independently halo, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy
- R 10 is, at each occurrence, independently C 1-3 alkoxy or C 1-3 haloalkoxy
- m is 0–3
- n is 0–4.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is H. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is C1-3 alkyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is methyl.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is ethyl or propyl.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is C 1-3 haloalkyl.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 1 is ⁇ CFH 2 , ⁇ CF 2 H, or ⁇ CF 3 .
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein m is 0. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein m is 1–3. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein n is 0. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein n is 1–2.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 7 , the reactive moiety capable of attachment to a linker or the reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety, is ⁇ Y ⁇ (CR 11 R 12 ) p ⁇ NHR 13 , ⁇ Y ⁇ (CR 11 R 12 ) p ⁇ C(O)OR 13 , or ⁇ Y ⁇ (CR 11 R 12 ) p ⁇ C(O)NHR 13 , wherein: Y is ⁇ O ⁇ , ⁇ CH 2 ⁇ , ⁇ OCH 2 CH 2 ⁇ , ⁇ OCH 2 CH 2 NR 14 ⁇ , ⁇ C(O)NR 14 ⁇ ; ⁇ CH 2 C(O)NR 14 ⁇ , ⁇ CH 2 CH 2 C(O)NR 14 ⁇ , ⁇ C(O)O ⁇ ; ⁇ CH 2 C(O)O ⁇ , ⁇ CH 2 CH 2 C(O)O
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 15 is ⁇ C 1-3 alkyl ⁇ C(O)OR 13 ; In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 7 is ⁇ Y ⁇ (CR 11 R 12 ) p ⁇ NHR 13 . In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein Y is ⁇ O ⁇ .
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein Y is ⁇ CH 2 ⁇ . In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein Y is ⁇ OCH 2 CH 2 ⁇ . In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein Y is ⁇ OCH 2 CH 2 NR 14 ⁇ . In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 7 is:
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 7 , the reactive moiety capable of attachment to a linker or the reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety, comprises a substitutable nitrogen atom.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R 7 , the reactive moiety capable of attachment to a linker or the reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety, comprises a moiety that enhances cell permeability.
- compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein the compound has a pKa of about 3.5 to about 5.5.
- Representative compounds of Formula (D-I), and Formulas (D-II) through (D-IX) as applicable, include the compounds listed in Tables 21–24 below, as well as pharmaceutically acceptable salts thereof. To this end, representative compounds are identified herein by their respective “Compound Number”, which is sometimes abbreviated as “Compound No.”, “Cmpd No.” or “No.”
- Linker Region L 3 The conjugates of the present disclosure include a linker region, L 3 .
- Region L 3 covalently links Region G, which comprises the GalNAc moiety, to Region Inh, which comprises the TGF ⁇ R1 inhibitor.
- the covalent linkages can be formed by reaction between functional groups in synthetic precursors for each region.
- Linkers of the disclosure (L 3 ) may have from about 10 to about 500 atoms in a linker, such as from about 10 to about 400 atoms, such as about 10 to about 300 atoms in a linker. In certain embodiments, linkers of the disclosure have from about 30 to about 400 atoms, such as from about 30 to about 300 atoms in the linker.
- an inhibitor as described herein is covalently bound to a GalNAc moiety by way of a linker, also referred to herein as L 3 .
- L 3 as used herein, may be selected from any of the linker moieties discussed herein.
- a conjugate may be prepared by contacting an GalNAc moiety with a linker-compound described herein under conditions in which the linker- compound covalently links to the GalNAc moiety.
- One embodiment pertains to a method of making a conjugate by contacting a linker-compound with a unit comprising Region G under conditions in which the linker-compound covalently links to the GalNAc moiety.
- any one of the inhibitors described herein is covalently bound to a linker (L 3 ).
- the linker may be covalently bound to any position on the inhibitor, valence permitting.
- the linker may comprise a reactive moiety, e.g., a nucleophile or an an electrophile that can react to form a covalent bond.
- Linkers can be short, long, flexible, rigid, cleavable, non-cleavable, hydrophilic, hydrophobic, unbranched (e.g., where z is 1), or branched (e.g., where z is greater than 1).
- a linker can contain connector segments that have different characteristics, such as segments of flexibility and segments of rigidity, or segments that are cleavable and segments that are cleavable.
- a linker can contain multiple segments, such as one or more non-cleavable segments and one or more cleavable segments.
- the linkers may be polyvalent such that they covalently link more than one inhibitor to a single site on the GalNAc moiety, or monovalent such that covalently they link a single inhibitor to a single site on the GalNAc moiety.
- a linker can comprise alkylene, alkenylene, alkynylene, polyether, polyester, polyamide, polyamino acid, polypeptide, cleavable peptide, and/or para-aminobenzylcarbamate groups.
- the linker comprises a “non-cleavable” segment, such as a “non-cleavable linker,” that is chemically stable to extracellular environments, for example, chemically stable in the blood stream and in intracellular environments.
- the linker comprises a “cleavable” segment, such as a “cleavable linker,” that includes one or more linkages that are not stable, such as linkages that are designed to cleave and/or immolate or otherwise breakdown specifically or non-specifically in the blood stream and/or inside cells (i.e., in an intracellular environment).
- Linkers comprise one or more cleavable segments, one or more non-cleavable segments, or a combination thereof.
- a moiety, construct, or conjugate described herein includes the symbol , which indicates the point of attachment, e.g., the point of attachment of a chemical or functional moiety to the compound, the point of attachment of a linker to a compound of the disclosure, or the point of attachment of a linker to a GalNAc moiety.
- Sulfamide linkers may be used to link many compounds of the present invention to a GalNAc moiety. Sulfamide linkers are as described herein and e.g., U.S. Patent Publication Number 2019/0038765, the linkers of which are incorporated by reference herein. Cleavable linkers can be cleavable in vitro and in vivo.
- Cleavable linkers can include chemically or enzymatically unstable or degradable linkages. Cleavable linkers can rely on processes inside the cell to liberate a TGF ⁇ R1 inhibitor, such as reduction in the cytoplasm, exposure to acidic conditions in the lysosome, or cleavage by specific proteases or other enzymes within the cell.
- a cleavable linker can be sensitive to (i.e., cleavable by) enzymes at a specific site.
- a cleavable linker can be cleaved by enzymes such as proteases.
- Cleavable linkers can incorporate one or more chemical bonds that are either chemically or enzymatically cleavable while the remainder of the linker can be non-cleavable.
- L is a linker comprising a reactive moiety.
- ⁇ L is represented by the formula: .
- ⁇ L is represented by the formula: , wherein each R 30 is independently selected from optionally substituted C 1 -C 6 alkyl and optionally substituted phenyl, and RX is the reactive moiety.
- RX may comprise a leaving group.
- RX may be a maleimide.
- L may be further covalently bound to a GalNAc moiety.
- ⁇ L ⁇ is represented by the formula: , wherein RX * is a bond, a succinimide moiety, or a hydrolyzed succinimide moiety bound to a GalNAc moiety, wherein on RX* represents the point of attachment; and each R 30 is independently selected from optionally substituted C 1 -C 6 alkyl and optionally substituted phenyl.
- linker-payload comprising a compound of the present disclosure, or pharmaceutically acceptable isomer, tautomer, racemate, hydrate, solvate, isotope, or salt thereof, and linker L; L comprises a methylene carbamate unit.
- a linker can contain a chemically labile group such as hydrazone and/or disulfide groups. Linkers comprising chemically labile groups can exploit differential properties between the plasma and some cytoplasmic compartments.
- the intracellular conditions that can facilitate TGF ⁇ R1 inhibitor release for hydrazone containing linkers can be the acidic environment of endosomes and lysosomes, while the disulfide containing linkers can be reduced in the cytosol, which can contain high thiol concentrations, e.g., glutathione.
- the plasma stability of a linker containing a chemically labile group can be increased by introducing steric hindrance using substituents near the chemically labile group.
- Acid-labile groups such as hydrazone
- This pH dependent release mechanism can be associated with nonspecific release of the drug.
- the linker can be varied by chemical modification, e.g., substitution, allowing tuning to achieve more efficient release in the lysosome with a minimized loss in circulation.
- ⁇ L comprises a hydrazone moiety.
- L may be selected from: wherein M is selected from C 1 -C 6 alkyl, aryl, and ⁇ O ⁇ C 1 -C 6 alkyl.
- Hydrazone-containing linkers can contain additional cleavage sites, such as additional acid-labile cleavage sites and/or enzymatically labile cleavage sites.
- Conjugates including exemplary hydrazone-containing linkers can include, for example, the following structures:
- linker (Ia) the linker can comprise two cleavable groups– a disulfide and a hydrazone moiety.
- effective cleavage can require acidic pH or disulfide reduction and acidic pH.
- Linkers such as (Ib) and (Ic) can be effective with a single hydrazone cleavage site.
- Other acid-labile groups that can be included in linkers include cis-aconityl-containing linkers.
- cis-Aconityl chemistry can use a carboxylic acid juxtaposed to an amide bond to accelerate amide hydrolysis under acidic conditions.
- Cleavable linkers can also include a disulfide group.
- Disulfides can be thermodynamically stable at physiological pH and can be designed to release upon internalization inside cells, wherein the cytosol can provide a significantly more reducing environment compared to the extracellular environment. Scission of disulfide bonds can require the presence of a cytoplasmic thiol cofactor, such as (reduced) glutathione (GSH), such that disulfide-containing linkers can be reasonably stable in circulation, selectively releasing the TGF ⁇ R1 inhibitor in the cytosol.
- GSH cytoplasmic thiol cofactor
- the intracellular enzyme protein disulfide isomerase or similar enzymes capable of cleaving disulfide bonds, can also contribute to the preferential cleavage of disulfide bonds inside cells.
- GSH can be present in cells in the concentration range of 0.5-10 mM compared with a significantly lower concentration of GSH or cysteine, the most abundant low-molecular weight thiol, in circulation at approximately 5 ⁇ M.
- Tumor cells where irregular blood flow can lead to a hypoxic state, can result in enhanced activity of reductive enzymes and therefore even higher glutathione concentrations.
- the in vivo stability of a disulfide-containing linker can be enhanced by chemical modification of the linker, e.g., use of steric hindrance adjacent to the disulfide bond.
- L may be selected from: , wherein M is selected from C 1 -C 6 alkyl, aryl, and ⁇ O-C 1 -C 6 alkyl.
- exemplary cleavable linkers including disulfide moieties can include the following wherein R is independently selected at each occurrence from hydrogen or alkyl, for example. Increasing steric hindrance adjacent to the disulfide bond can increase the stability of the linker. Structures such as (Id) and (If) can show increased in vivo stability when one or more R groups is selected from a lower alkyl such as methyl.
- Another type of cleavable linker is specifically cleaved by an enzyme.
- the linker can be cleaved by a lysosomal enzyme.
- a lysosomal enzyme can be peptide-based or can include peptidic regions that can act as substrates for enzymes.
- Peptide-based linkers can be more stable in plasma and extracellular milieu than chemically labile linkers. Peptide bonds can have good serum stability, as lysosomal proteolytic enzymes can have very low activity in blood due to endogenous inhibitors and the unfavorably high pH value of blood compared to lysosomes. Release of a TGF ⁇ R1 inhibitor from a conjugate can occur due to the action of lysosomal proteases, e.g., cathepsin and/or plasmin.
- the linker can be cleavable by a lysosomal enzyme.
- the lysosomal enzyme can be, for example, cathepsin B, ⁇ -glucuronidase, or ⁇ -galactosidase.
- the cleavable peptide can be selected from tetrapeptides such as Gly-Phe-Leu-Gly, Ala- Leu-Ala-Leu or dipeptides such as Val-Cit, Val-Ala, and Phe-Lys. Dipeptides can have lower hydrophobicity compared to longer peptides.
- the cleavable linker comprises a cleavable peptide.
- the cleavable peptide is a dipeptide, tripeptide, or tetrapeptide.
- the cleavable peptide is Val ⁇ Cit; Cit ⁇ Val; Ala ⁇ Ala; Ala ⁇ Cit; Cit ⁇ Ala; Asn ⁇ Cit; Cit ⁇ Asn; Cit ⁇ Cit; Val ⁇ Glu; Glu ⁇ Val; Ser ⁇ Cit; Cit ⁇ Ser; Lys ⁇ Cit; Cit ⁇ Lys; Asp ⁇ Cit; Cit ⁇ Asp; Ala ⁇ Val; Val ⁇ Ala; Phe ⁇ Lys; Lys ⁇ Phe; Val ⁇ Lys; Lys ⁇ Val; Ala ⁇ Lys; Lys ⁇ Ala; Phe ⁇ Cit; Cit ⁇ Phe; Leu ⁇ Cit; Cit ⁇ Leu; Ile ⁇ Cit; Cit ⁇ Ile; Phe ⁇ Arg; Arg ⁇ Phe; Cit ⁇ Trp;
- the cleavable linker comprises a structure of formula: wherein ⁇ AA 1 ⁇ AA 2 ⁇ is the cleavable dipeptide and AA 1 and AA 2 are each an amino acid.
- the cleavable dipeptide is Val ⁇ Cit.
- Enzymatically cleavable linkers can include a self-immolative spacer to spatially separate the TGF ⁇ R1 inhibitor from the site of enzymatic cleavage. The direct attachment of a TGF ⁇ R1 inhibitor to a peptide linker can result in proteolytic release of an amino acid adduct of the TGF ⁇ R1 inhibitor, thereby impairing its activity.
- One self-immolative spacer can be a bifunctional para-aminobenzyl alcohol (PABA) group, which can link to the peptide through the amino group, forming an amide bond, while amine containing TGF ⁇ R1 inhibitors can be attached through carbamate functionalities to the benzylic hydroxyl group of the linker to form a para-aminobenzyl carbamate (PMBC) group.
- PABA para-aminobenzyl alcohol
- a cleavable linker comprises a valine-citrulline peptide or a valine-alanine peptide.
- a valine-citrulline or valine-alanine-containing linker can contain a succimide group.
- a valine-citrulline- or valine-alanine-containing linker can contain a para- aminobenzyl (PAB) group.
- the PAB group is a para-aminobenzyl alcohol (PABA) group or a para-aminobenzyl carbamate (PABC) group.
- a valine-citrulline- or valine-alanine-containing linker also includes a PAB group, such as a PABC group.
- a cleavable linker comprises a ⁇ (valine ⁇ citrulline) ⁇ (para ⁇ aminobenzyloxycarbonyl) group.
- a cleavable linker comprises a lysine with an N-terminal amine acetylated, and a valine-citrulline cleavage site.
- the cleavable linker comprises a succinimide group.
- the enzymatically cleavable linker can be a ß-glucuronic acid-based linker. Facile release of the TGF ⁇ R1 inhibitor can be realized through cleavage of the ß-glucuronide glycosidic bond by the lysosomal enzyme ß-glucuronidase. This enzyme can be abundantly present within lysosomes and can be overexpressed in some tumor types, while the enzyme activity outside cells can be low.
- ß-Glucuronic acid-based linkers can be used to circumvent the tendency of a conjugate to undergo aggregation due to the hydrophilic nature of ß-glucuronides.
- ß-glucuronic acid-based linkers can link the GalNAc moiety to a hydrophobic TGF ⁇ R1 inhibitor.
- cleavable ⁇ -glucuronic acid-based linkers useful for linking drugs such as auristatins, camptothecin and doxorubicin analogues, CBI minor-groove binders, and psymberin to antibodies have been described, and such linkers are useful for the present conjugates as well.
- ⁇ -glucuronic acid-based linkers may be used in the conjugates comprising a TGF ⁇ R1 inhibitor described herein.
- the enzymatically cleavable linker is a ⁇ -galactoside-based linker.
- ⁇ -Galactoside is present abundantly within lysosomes, while the enzyme activity outside cells is low.
- cleavable linkers may comprise a phenol and connection through the phenolic oxygen.
- One such linker employs diamino-ethane unit (e.g., “Space Link”) in conjunction with traditional “PABO”-based self-immolative groups to deliver a phenol.
- Inhibitors containing an aromatic or aliphatic hydroxyl group can be covalently bonded to a linker through the hydroxyl group using a methodology that relies on a methylene carbamate linkage, as described in WO 2015/095755.
- Cleavable linkers can include non-cleavable portions or segments, and/or cleavable segments or portions can be included in an otherwise non-cleavable linker to render it cleavable.
- polyethylene glycol (PEG) and related polymers can include cleavable groups in the polymer backbone.
- a polyethylene glycol or polymer linker can include one or more cleavable groups such as a disulfide, a hydrazone or a dipeptide.
- cleavable linkages such as a disulfide, a hydrazone or a dipeptide.
- Other degradable linkages that can be included in cleavable linkers include ester linkages formed by the reaction of PEG carboxylic acids or activated PEG carboxylic acids with alcohol groups on a TGF ⁇ R1 inhibitor, wherein such ester groups can hydrolyze under physiological conditions to release the TGF ⁇ R1 inhibitor.
- Hydrolytically degradable linkages can include, but are not limited to, carbonate linkages; imine linkages resulting from reaction of an amine and an aldehyde; phosphate ester linkages formed by reacting an alcohol with a phosphate group; acetal linkages that are the reaction product of an aldehyde and an alcohol; orthoester linkages that are the reaction product of a formate and an alcohol; and oligonucleotide linkages formed by a phosphoramidite group, including but not limited to, at the end of a polymer, and a 5’ hydroxyl group of an oligonucleotide.
- a linker can be a link created by a microbial transglutaminase, wherein the link can be created between an amine-containing moiety and a moiety engineered to contain glutamine as a result of the enzyme catalyzing a bond formation between the acyl group of a glutamine side chain and the primary amine of a lysine chain.
- a linker can contain a reactive primary amine.
- a linker can be a Sortase A linker.
- a Sortase A linker can be created by a Sortase A enzyme fusing an LPXTG (SEQ ID NO:1) recognition motif to an N-terminal GGG motif to regenerate a native amide bond.
- the linker created can therefore link a moiety attached to the LPXTG (SEQ ID NO:1) recognition motif with a moiety attached to the N-terminal GGG motif.
- a linker can be a link created between an unnatural amino acid on one moiety reacting with oxime bond that was formed by modifying a ketone group with an alkoxyamine on another moiety.
- a moiety can be a TGF ⁇ R1 inhibitor.
- a non-cleavable linker can be protease insensitive.
- a non-cleavable linker can contain a succinimide group.
- a non-cleavable linker can contain a caproyl group or a succinimidocaproyl group.
- a succinimidocaproyl linker can comprise N-succinimidomethylcyclohexane-1- carboxylate.
- a non-cleavable linker can be a combination of a succinimidocaproyl group and one or more ethylene glycol units.
- a non-cleavable linker can be a PEG4 linker or a succinimido-PEG4 linker.
- a non-cleavable linker can be a combination of a succinimidocaproyl linker containing a succinimide group and one or more ethylene glycol units.
- a non-cleavable linker can contain one or more succinimido groups linked to polyethylene glycol units in which the polyethylene glycol can allow for more linker flexibility or can be used lengthen the linker.
- a non-cleavable linker independently comprises a heteroalkylene comprising 3 to 15 components in the longest linear chain, wherein the components are selected from ⁇ CH 2 ⁇ , ⁇ CH(C 1-4 alkyl), ⁇ C(O) ⁇ , ⁇ O ⁇ , ⁇ NH ⁇ , and ⁇ N(C 1-4 alkyl).
- the non-cleavable linker comprises a polyethylene glycol region comprising two to six ethylene glycol units.
- a non-cleavable linker is ⁇ NH ⁇ C(O) ⁇ (CH 2 ) 2-5 ⁇ C(O) ⁇ , ⁇ NH ⁇ C(O) ⁇ (CH 2 ) 2-5 ⁇ C(O)NH ⁇ (CH 2 ) 2-7 ⁇ C(O) ⁇ , or ⁇ NH ⁇ C(O) ⁇ (CH 2 ) 2-5 ⁇ C(O)NH ⁇ (CH 2 ) 2-7 ⁇ NH ⁇ C(O) ⁇ (CH 2 ) 2-4 ⁇ C(O) ⁇ .
- a non-cleavable linker is ⁇ NH ⁇ C(O) ⁇ (CH 2 ) 3 ⁇ C(O) ⁇ .
- Attachment groups that are used to attach the linkers in a conjugate can be electrophilic in nature and include, for example, active esters such as NHS esters and HOBt esters, haloformates, acid halides, carboxylic acids, amines, alkyl halides, benzyl halides such as haloacetamides, maleimide groups, or activated disulfides.
- a linker can comprise a cleavable peptide, for example, a linker comprising structural formula (IVa), (IVb), (IVc), or (IVd): or a salt thereof, wherein: peptide represents a cleavable peptide (illustrated N ⁇ C, wherein peptide includes the amino and carboxy “termini”) as described herein; T represents a polymer comprising one or more ethylene glycol units or an alkylene chain, or combinations thereof; R a is selected from hydrogen, alkyl, sulfonate and methyl sulfonate; R y is hydrogen or C 1-4 alkyl ⁇ (O) r ⁇ (C 1-4 alkylene) s ⁇ G 1 or C 1-4 alkyl ⁇ (N) ⁇ [(C 1-4 alkylene) ⁇ G 1 ] 2 ; R z is C
- Exemplary polyvalent linkers that may be used to link inhibitors include Fleximer® linker technology that has the potential to enable high loading of conjugates with good physicochemical properties.
- the Fleximer® linker technology is based on incorporating drug molecules into a solubilizing poly-acetal backbone via a sequence of ester bonds.
- an aliphatic alcohol can be present or introduced into an inhibitor or salt thereof (e.g., an ALK5 inhibitor, as described herein).
- the alcohol moiety is then conjugated to an alanine moiety, which is then synthetically incorporated into the Fleximer® linker. Liposomal processing of the conjugate in vitro releases the parent alcohol-containing drug.
- Sulfamide linkers may also be used in the disclosed conjugates. Sulfamide linkers are as described herein and e.g., U.S. Patent Publication No. 2019/0038765, the linkers of which are incorporated by reference herein.
- Exemplary embodiments of linkers according to structural formula (IVa) that can be included in the conjugates described herein can include the structures illustrated below:
- cleavable linker can contain an enzymatically cleavable sugar moiety, for example, a linker comprising structural formula (Va), (Vb), (Vc), (Vd), or (Ve):
- linkers according to structural formula (IVa) that may be included in the conjugates described herein can include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the TGF ⁇ R1 inhibitor:
- linkers according to structural formula (IVb) that may be included in the conjugates described herein can include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the TGF ⁇ R1 inhibitor:
- linkers according to structural formula (IVc) that may be included in the conjugates described herein can include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the TGF ⁇ R1 inhibitor:
- linkers according to structural formula (IVd) that may be included in the conjugates described herein can include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the TGF ⁇ R1 inhibitor:
- linkers according to structural formula (IVe) that may be included in the conjugates described herein can include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the TGF ⁇ R1 inhibitor:
- the linker can be non-cleavable in vivo, for example, a linker according to the formulations below: or salts thereof, wherein: R a is selected from hydrogen, alkyl, sulfonate and methyl sulfonate; R x is a moiety that covalently links the linker to the rest of the conjugate, such as a bond; and represents the point of attachment of the linker to the rest of the conjugate.
- Exemplary embodiments of linkers according to structural formula (Va)-(Vf) that may be included in the conjugates described herein include the linkers illustrated: .
- Attachment groups that are used to attach the linkers to a TGF ⁇ R1 inhibitor can be electrophilic in nature and include, for example, maleimide groups, alkynes, alkynoates, allenes and allenoates, activated disulfides, active esters such as NHS esters and HOBt esters, haloformates, acid halides, alkyl, and benzyl halides such as haloacetamides.
- active esters such as NHS esters and HOBt esters
- haloformates acid halides, alkyl, and benzyl halides such as haloacetamides.
- Self-stabilizing linkers may also include aryl, e.g., phenyl, or heteroaryl, e.g., pyridine, groups optionally substituted with electron withdrawing groups such as those described herein. Examples of self-stabilizing linkers are provided in, e.g., U.S. Patent Publication No. US 2013/0309256, the linkers of which are incorporated by reference herein. It will be understood that a self-stabilizing linker useful in conjunction with immune-stimulatory compounds may be equivalently described as unsubstituted maleimide-including linkers, thio- substituted succinimide-including linkers, or hydrolyzed, ring-opened thio-substituted succinimide-including linkers. In certain embodiments, a linker comprises a stabilizing linker moiety selected from:
- the bottom structure may be referred to as (maleimido)- DPR-Val-Cit-PAB, where DPR refers to diaminopropinoic acid, Val refers to valine, Cit refers to citrulline, and PAB refers to para-aminobenzylcarbonyl, wherein represents the point of attachment of the linker to a nitrogen of the inhibitor.
- DPR refers to diaminopropinoic acid
- Val refers to valine
- Cit refers to citrulline
- PAB para-aminobenzylcarbonyl, wherein represents the point of attachment of the linker to a nitrogen of the inhibitor.
- An advantage of this methodology is the ability to synthesize homogenous DAR4 conjugates by full reduction of IgGs (to give 4 pairs of sulfhydryls from interchain disulfides) followed by reaction with 4 equivalents of the alkylating agent. Conjugates containing "bridged disulfides" are also claimed to have increased stability. Similarly, as depicted below, a maleimide derivative that is capable of bridging a pair of sulfhydryl groups has been developed.
- a linker can contain the following structural formulas (VIa), (VIb), or (VIc): or salts thereof, wherein: R q is H or–O-(CH 2 CH 2 O) 11 -CH 3 ; x is 0 or 1; y is 0 or 1; G 2 is -CH 2 CH 2 CH 2 SO 3 H or–CH 2 CH 2 O-(CH 2 CH 2 O) 11 -CH 3 ; R w is–O-CH 2 CH 2 SO 3 H or -NH(CO)-CH 2 CH 2 O-(CH 2 CH 2 O) 12 -CH 3 ; and * represents the point of attachment to the remainder of the linker.
- Exemplary embodiments of linkers according to structural formula (VIa) and (VIb) that can be included in the conjugates described herein can include the linkers illustrated:
- linkers according to structural formula (Vic) that can be included in the immune-stimulatory conjugates described herein can include the linkers illustrated below:
- ⁇ L 3 wherein represents the point of attachment of the linker to a nitrogen of the inhibitor.
- the reactive moiety may be selected, for example, from an electrophile, e.g., an ⁇ ⁇ -unsaturated carbonyl, such as a maleimide, and a leaving group.
- an electrophile e.g., an ⁇ ⁇ -unsaturated carbonyl, such as a maleimide, and a leaving group.
- ⁇ L can be represented by the formulas set forth in Table 2 below: TABLE 2
- such linkers can be, for example, represented by the Formulas set forth in Table 3 below: T
- –L 3 is represented by the formulas set forth in Table 4 below: TABLE 4 wherein represents attachment to a nitrogen of a TGF ⁇ R1 inhibitor and RX represents a reactive moiety.
- the reactive moiety may be selected from, for example, a leaving group.
- –L can be represented by the formulas set forth in Table 5 below:
- linkers can, for example, be represented by the Formulas set forth in Table 6 below wherein RX * is a bond to the GalNAc moiety, wherein on RX* represents the point of attachment to such residue: TABLE 6
- the linker selected for a particular conjugate may be influenced by a variety of factors, including the site of attachment to GalNAc moiety structural constraints of the drug pharmacophore, and the lipophilicity of the drug.
- the specific linker selected for a conjugate should seek to balance these different factors. For example, cytotoxic conjugates have been observed to effect killing of bystander antigen-negative cells present in the vicinity of the antigen-positive tumor cells. The mechanism of the bystander effect by cytotoxic conjugates has indicated that metabolic products formed during intracellular processing of the conjugates may play a role.
- a linker is selected to attenuate the bystander effect caused by cellular metabolites of the conjugate. In further embodiments, a linker is selected to increase the bystander effect.
- the properties of the linker, or linker-payload, may also impact aggregation of a conjugate under conditions of use and/or storage. Conjugates reported in the literature contain about 3-4 drug molecules per antibody molecule.
- DAR drug-to-antibody ratios
- Attempts to obtain higher drug-to-antibody ratios (“DAR”) often failed, particularly if both the drug and the linker were hydrophobic, due to aggregation of the conjugate.
- DARs higher than 3-4 could be beneficial as a means of increasing potency.
- a linker incorporates chemical moieties that reduce aggregation of the conjugates during storage and/or use.
- a linker may incorporate polar or hydrophilic groups such as charged groups or groups that become charged under physiological pH to reduce the aggregation of the conjugates.
- a linker may incorporate charged groups such as salts or groups that deprotonate, e.g., carboxylates, or protonate, e.g., amines, at physiological pH.
- aggregation of conjugates during storage or use is less than about 40% as determined by size-exclusion chromatography (SEC).
- the aggregation of the conjugates during storage or use is less than about 35%, such as less than about 30%, about 25%, about 20%, about 15%, about 10%, about 5%, about 4%, or even less, as determined by size-exclusion chromatography (SEC).
- SEC size-exclusion chromatography
- the reactive moiety may be selected, for example, from an electrophile, e.g., an ⁇ , ⁇ -unsaturated carbonyl, such as a maleimide, and a leaving group.
- an electrophile e.g., an ⁇ , ⁇ -unsaturated carbonyl, such as a maleimide, and a leaving group.
- ⁇ L 3 can be represented by the formulas set forth in Table 8 below: TABLE 8
- a particularly preferred pepide is val-ala or val-cit.
- attachment of the linker is to a nitrogen of a TGF ⁇ R1 inhibitor and ⁇ RX represents the reactive moiety for the point of attachment to the GalNAc moiety
- ⁇ RX represents the reactive moiety for the point of attachment to the GalNAc moiety
- the reactive moiety may be selected from, for example, a leaving group.
- – L 3 can be represented by the formulas set forth in Table 11 below:
- TABLE 11 L3 can be represented by the Formulas set forth in Table 12 below wherein RX * is a bond to the GalNAc moiety, wherein on RX* represents the point of attachment to such residue:
- TABLE 12 represents attachment to a nitrogen of a TGF ⁇ R1 inhibitor and ⁇ RX* represents the point of attachment to the GalNAc moiety.
- the linkers described herein, including those in the preceding paragraphs are attached to a compound of the present invention through the nitrogen of a secondary acyclic amine depicted in the structure of any one the compounds of the present invention.
- the linkers described herein, including those in the preceding paragraphs, are attached to a compound of the present invention at a nitrogen atom.
- the linker selected for a particular conjugate may be influenced by a variety of factors, including but not limited to, the site of attachment to the antibody construct (e.g., lys, cys or other amino acid residues), structural constraints of the drug pharmacophore and the lipophilicity of the drug.
- the specific linker selected for a conjugate should seek to balance these different factors for the specific GalNAc construct/drug combination.
- the conjugate comprises one of the following structures: , or a salt thereof.
- the conjugate comprises a structure as shown in Table 13: TABLE 13
- the conjugate comprises one of the following structures, where the wavy bond is the point of attachment to the rest of the conjugate:
- Region L 3 comprises a dipeptide. In some embodiments, the dipeptide is Val ⁇ Cit. In some embodiments, Region L 3 comprises a para-aminobenzyl moiety. In some embodiments, Region L 3 comprises a PABC moiety. In some embodiments, Region L 3 comprises ⁇ NHC(O) ⁇ (CH 2 ) p ⁇ C(O) ⁇ where p is 2, 3, 4, 5, or 6. In some embodiments, Region L 3 is ⁇ NHC(O) ⁇ (CH 2 ) p ⁇ C(O) ⁇ Val ⁇ Cit ⁇ PAB ⁇ O ⁇ C(O) ⁇ .
- a TGF ⁇ R1 inhibitor-linker compound can be synthesized by various methods known in the art.
- the linker selected for a particular conjugate may be influenced by a variety of factors, including but not limited to, the site of attachment to the GalNAc moiety, structural constraints of the drug pharmacophore, and the lipophilicity of the drug.
- the specific linker selected for a conjugate should seek to balance these different factors for the specific conjugate.
- the disclosure includes a TGF ⁇ R1 inhibitor of Formula (XX): [Inh] ⁇ Z (XX) wherein Region Inh comprises a TGF ⁇ R1 inhibitor (e.g., a compound of Formula (A-I), (B-I), or (C-1); and Z is the residual portion of a released, cleavable linker or comprises a non-cleavable linker and a residual portion of a degraded Region G; or a salt thereof.
- Z is a moiety that remains covalently attached to the rest of the inhibitor structure following cleavage (e.g., in vivo) of a cleavable linker of a conjugate as described herein.
- Z comprises an amino acid. In some embodiments, Z comprises valine or citrulline. In some embodiments, Z is ⁇ NH 2 . In some embodiments, Z is a moiety that remains covalently attached to the inhibitor following degradation (e.g., in vivo) of Region G of a conjugate as described herein. In some embodiments, Z comprises a degraded Region G and a non-cleavable linker as described herein. In some embodiments, the degradation is by enzyme- or acid-mediated degradation, such as hydrolysis. In some embodiments, a degraded Region G comprises one or more degraded GalNAc groups. Pharmaceutical Formulations The conjugates described herein are useful as pharmaceutical compositions for administration to a subject in need thereof.
- compositions can comprise the conjugates described herein and one or more pharmaceutically acceptable carriers, diluents, excipients, stabilizers, dispersing agents, suspending agents, and/or thickening agents.
- a pharmaceutical composition can comprise any conjugate described herein.
- a pharmaceutical composition can further comprise buffers, antibiotics, steroids, carbohydrates, drugs (e.g., chemotherapy drugs), radiation, polypeptides, chelators, adjuvants and/or preservatives.
- conjugates for which a is 1 comprise a Region Fc. These conjugates can have an average drug loading. The average drug loading is the average number of TGF ⁇ R1 inhibitor molecules per Region Fc.
- the average number of TGF ⁇ R1 inhibitors per Region Fc in a preparation may be characterized by conventional means such as mass spectroscopy, HIC, ELISA assay, and HPLC.
- the average drug loading is 1 to 8, or 1 to 6, or 1 to 4, or 1 to 3, or 1 to 2, or 2 to 4, or 2 to 3, TGF ⁇ R1 inhibitor molecules per Region Fc.
- Pharmaceutical compositions can be formulated using one or more physiologically- acceptable carriers comprising excipients and auxiliaries. Formulation can be modified depending upon the route of administration chosen.
- Pharmaceutical compositions comprising a conjugate as described herein can be manufactured, for example, by lyophilizing the conjugate, mixing, dissolving, emulsifying, encapsulating or entrapping the conjugate.
- the pharmaceutical compositions can also include the conjugates described herein in a free-base form or pharmaceutically-acceptable salt form.
- Methods for formulation of the pharmaceutical compositions can include formulating any of the conjugates described herein with one or more inert, pharmaceutically-acceptable excipients or carriers to form a solid, semi-solid, or liquid composition.
- Solid compositions can include, for example, powders, tablets, dispersible granules and capsules, and in some aspects, the solid compositions further contain nontoxic, auxiliary substances, for example wetting or emulsifying agents, pH buffering agents, and other pharmaceutically-acceptable additives.
- compositions described herein can be lyophilized or in powder form for re- constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use
- a suitable vehicle e.g., sterile pyrogen-free water
- Pharmaceutical compositions of the conjugates described herein can comprise at least a conjugate as an active ingredient, respectively.
- the active ingredients can be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization (e.g., hydroxymethylcellulose or gelatin microcapsules and poly- (methylmethacylate) microcapsules, respectively), in colloidal drug-delivery systems (e.g., liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
- colloidal drug-delivery systems e.g., liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsule
- compositions as described herein often further can comprise more than one active compound as necessary for the particular indication being treated.
- the active compounds can have complementary activities that do not adversely affect each other.
- the pharmaceutical composition can also comprise a cytotoxic agent, cytokine, growth- inhibitory agent, anti-hormonal agent, anti-angiogenic agent, and/or cardioprotectant.
- Such molecules can be present in combination in amounts that are effective for the purpose intended.
- the pharmaceutical compositions and formulations can be sterilized. Sterilization can be accomplished by filtration through sterile filtration.
- the pharmaceutical compositions described herein can be formulated for administration as an injection. Non-limiting examples of formulations for injection can include a sterile suspension, solution or emulsion in oily or aqueous vehicles.
- Suitable oily vehicles can include, but are not limited to, lipophilic solvents or vehicles such as fatty oils or synthetic fatty acid esters, or liposomes.
- Aqueous injection suspensions can contain substances which increase the viscosity of the suspension.
- the suspension can also contain suitable stabilizers.
- Injections can be formulated for bolus injection or continuous infusion.
- the pharmaceutical compositions described herein can be lyophilized or in powder form for reconstitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
- the conjugates can be formulated in a unit dosage injectable form (e.g., use letter solution, suspension, emulsion) in association with a pharmaceutically acceptable parenteral vehicle.
- Such vehicles can be inherently nontoxic, and non-therapeutic.
- a vehicle can be water, saline, Ringer’s solution, dextrose solution, and 5% human serum albumin.
- Nonaqueous vehicles such as fixed oils and ethyl oleate can also be used.
- Liposomes can be used as carriers.
- the vehicle can contain minor amounts of additives such as substances that enhance isotonicity and chemical stability (e.g., buffers and preservatives). Sustained-release preparations can also be prepared.
- sustained-release matrices can include polyesters, hydrogels (e.g., poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides, copolymers of L-glutamic acid and ⁇ ethyl-L-glutamate, non- degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPO TM (i.e., injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-( – )-3-hydroxybutyric acid.
- polyesters e.g., poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)
- polylactides e.g., poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)
- polylactides e.g., poly
- compositions described herein can be prepared for storage by mixing a conjugate with a pharmaceutically acceptable carrier, excipient, and/or a stabilizer.
- This formulation can be a lyophilized formulation or an aqueous solution.
- Acceptable carriers, excipients, and/or stabilizers can be nontoxic to recipients at the dosages and concentrations used.
- Acceptable carriers, excipients, and/or stabilizers can include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives, polypeptides; proteins, such as serum albumin or gelatin; hydrophilic polymers; amino acids; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes; and/or non- ionic surfactants or polyethylene glycol.
- buffers such as phosphate, citrate, and other organic acids
- antioxidants including ascorbic acid and methionine
- preservatives polypeptides
- proteins such as serum albumin or gelatin
- hydrophilic polymers amino acids
- conjugates, pharmaceutical compositions, and methods of the present disclosure can be useful for treating a subject such as, but not limited to, a mammal, a human, a non-human mammal, a domesticated animal (e.g., laboratory animals, household pets, or livestock), a non- domesticated animal (e.g., wildlife), a dog, a cat, a rodent, a mouse, a hamster, a cow, a bird, a chicken, a fish, a pig, a horse, a goat, a sheep, or a rabbit.
- conjugates, pharmaceutical compositions, and methods of the present disclosure are used for treating a human.
- conjugates, pharmaceutical compositions, and methods described herein can be useful as a therapeutic, for example a treatment that can be administered to a subject in need thereof.
- a therapeutic effect can be obtained in a subject by reduction, suppression, remission, or eradication of a disease state, including, but not limited to, a symptom thereof.
- a therapeutic effect in a subject having a disease or condition, or pre-disposed to have or is beginning to have the disease or condition can be obtained by a reduction, a suppression, a prevention, a remission, or an eradication of the condition or disease, or pre-condition or pre-disease state.
- therapeutically-effective amounts of the conjugates, or pharmaceutical compositions described herein can be administered to a subject in need thereof, often for treating and/or preventing a condition or progression thereof.
- a pharmaceutical composition can affect the physiology of the subject, such as the immune system, inflammatory response, or other physiologic affect.
- a therapeutically-effective amount can vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compounds used, and other factors. Treat and/or treating can refer to any indicia of success in the treatment or amelioration of the disease or condition.
- Treating can include, for example, reducing, delaying or alleviating the severity of one or more symptoms of the disease or condition, or it can include reducing the frequency with which symptoms of a disease, defect, disorder, or adverse condition, and the like, are experienced by a patient. Treat can be used herein to refer to a method that results in some level of treatment or amelioration of the disease or condition, and can contemplate a range of results directed to that end, including but not restricted to prevention of the condition entirely. Prevent, preventing and the like can refer to the prevention of the disease or condition, e.g., viral infection, in the patient.
- the disease or condition e.g., viral infection
- a therapeutically effective amount can be the amount of conjugates or pharmaceutical compositions or an active component thereof sufficient to provide a beneficial effect or to otherwise reduce a detrimental non-beneficial event to the individual to whom the composition is administered.
- a therapeutically effective dose can be a dose that produces one or more desired or desirable (e.g., beneficial) effects for which it is administered, such administration occurring one or more times over a given period of time. An exact dose can depend on the purpose of the treatment, and can be ascertainable by one skilled in the art using known techniques.
- the conjugates or pharmaceutical compositions described herein that can be used in therapy can be formulated and dosages established in a fashion consistent with good medical practice taking into account the disorder to be treated, the condition of the individual patient, the site of delivery of the conjugate, or pharmaceutical composition, the method of administration and other factors known to practitioners.
- the conjugates or pharmaceutical compositions can be prepared according to the description of preparation described herein.
- One of ordinary skill in the art would understand that the amount, duration and frequency of administration of a pharmaceutical composition or conjugate described herein to a subject in need thereof depends on several factors including, for example but not limited to, the health of the subject, the specific disease or condition of the patient, the grade or level of a specific disease or condition of the patient, the additional therapeutics the subject is being or has been administered, and the like.
- compositions described herein can be for administration to a subject in need thereof.
- administration of the conjugates, or pharmaceutical compositions can include routes of administration, non-limiting examples of administration routes include intravenous, intraarterial, subcutaneous, subdural, intramuscular, intracranial, intrasternal, intratumoral, or intraperitoneally.
- a pharmaceutical composition, or conjugate can be administered to a subject by additional routes of administration, for example, by inhalation, oral, dermal, intranasal, or intrathecal administration.
- Pharmaceutical compositions or conjugates of the present disclosure can be administered to a subject in need thereof in a first administration, and in one or more additional administrations.
- the one or more additional administrations can be administered to the subject in need thereof minutes, hours, days, weeks or months following the first administration. Any one of the additional administrations can be administered to the subject in need thereof less than 21 days, or less than 14 days, less than 10 days, less than 7 days, less than 4 days or less than 1 day after the first administration.
- the one or more administrations can occur more than once per day, more than once per week or more than once per month.
- the conjugates or pharmaceutical compositions can be administered to the subject in need thereof in cycles of 21 days, 14 days, 10 days, 7 days, 4 days or daily over a period of one to seven days.
- the disclosure relates to a method for treating a disease mediated by TGF ⁇ R1 activity, comprising administering an effective amount of a conjugate or a pharmaceutical composition as described herein to a subject in need thereof.
- the disease is cancer.
- the cancer is hepatocellular carcinoma (HCC).
- the cancer is metastatic liver cancer from colon, lung, breast, neuroendocrine, stomach or pancreatic cancer.
- the disease is liver fibrosis.
- the fibrosis is liver fibrosis.
- the fibrosis is cancer-associated.
- the fibrosis is not cancer- associated.
- the fibrosis is associated with scleroderma, systemic fibrosis, steatohepatitis, or non-alcoholic steatohepatitis (NASH) , chronic liver viral disease (such as HBV and HCV infection), autoimmune hepatitis, or primary biliary cholangitis.
- the disease is a liver metabolic disease, such as steatosis.
- treatment with conjugates or pharmaceutical compositions of the present disclosure reduces hepatocyte apoptosis or reduces altered lipid metabolism in subject consuming a fatty-diet.
- a method of treating a subject having liver cancer comprising administering to the subject an effective amount of a conjugate as described herein or a pharmaceutical composition comprising the conjugate.
- the presently described conjugates or pharmaceutical compositions can be used to decrease HCC tumor cell growth and invasiveness.
- the presently described conjugates or pharmaceutical compositions can be utilized to enhance liver-localized immune responses against liver cancer, such as HCC.
- the administering is in a regimen that comprises administering the conjugates or pharmaceutical compositions intravenously or subcutaneouly.
- the dose level and schedule are chosen to maintain a high ratio of liver exposure to systemic exposure.
- the dose and schedule are chosen to attain about an IC 90 , IC 80 , IC 50 or IC 30 of phospho-SMAD2/3 levels in hepatocytes as compared to non- conjugated treatment for at least 24 hours.
- the dose and schedule are chosen to attain about an IC 90 , IC 80 , IC 50 or IC 30 of non-hepatocyte liver cells compared to non-conjugate treatment for 24 hours.
- Increased dosages and reduced side-effects In certain embodiments, using a conjugate of this disclosure can allow administration of the conjugate at greater levels of the inhibitor in the form of the conjugate than the level of inhibitor alone.
- the conjugate can be administered at a level higher than the maximum tolerated dose for that inhibitor administered in the absence of the being conjugated to the antibody construct in the conjugate. In certain embodiments, administration of the conjugate can be associated with fewer side effects than when administered as the inhibitor alone.
- Diseases, Conditions and the like can be useful for the treatment of a plurality of diseases, conditions, preventing a disease or a condition in a subject or other therapeutic applications for subjects in need thereof.
- the conjugates, pharmaceutical compositions, and methods provided herein can be useful for treatment of liver diseases, such as liver fibrosis or liver cancer.
- the fibrosis is cancer-associated. In some aspects, the fibrosis is not cancer-associated.
- the fibrosis is associated with scleroderma, systemic fibrosis, steatohepatitis, or non-alcoholic steatohepatitis (NASH), chronic viral HBV and HCV infection, autoimmune hepatitis, or primary biliary cholangitis.
- NASH non-alcoholic steatohepatitis
- the disclosure further provides any conjugates disclosed herein for use in a method of treatment of the human or animal body by therapy. Therapy may be by any mechanism disclosed herein, such as inhbiting, reducing, or reducing progression of fibrosis or other liver damage.
- the disclosure further provides any conjugate disclosed herein for prevention or treatment of any condition disclosed herein, for example, fibrosis or cancer.
- the disclosure also provides any conjugate or pharmaceutical composition thereof disclosed herein for obtaining any clinical outcome disclosed herein for any condition disclosed herein, such as treating fibrosis or cancer.
- the disclosure also provides use of any conjugate disclosed herein in the manufacture of a medicament for preventing or treating any condition disclosed herein.
- EXAMPLES The following examples illustrate the various methods of making compounds described herein. The examples are included to further describe some embodiments of the present disclosure, and should not be used to limit the scope of the disclosure. The following synthetic schemes are provided for purposes of illustration, not limitation. It is understood that one skilled in the art may be able to make these compounds by similar methods or by combining other methods known to one skilled in the art.
- R 2 -substituted pyridine-2- carbaldehydes are reacted with aniline and diphenyl phosphite to give N,P-acetal (iv), which can be further coupled with R 1 substituted [1,2,4]triazolo[1,5-a]pyridine-6-carbaldehydes (iii) followed by hydrolysis in acidic condition to produce a monoketone (v).
- the monoketone (v) may be oxidized to a diketone (vi) with HBr in DMSO.
- Diketones (vi) can be condensed with 2,2- dimethoxyacetaldehyde in the presence of ammonium acetate to yield an acetal-protected imidazole (vii), which can be hydrolyzed in acidic condition to produce an imidazole-2- carbaldehyde (viii).
- the imidazole-2-carbaldehyde (viii) can be reductively aminated in the presence of an amine and a reducing agent such as sodium borohydride or sodium cyanoborohydride to yield a compound of Formula (A-I) or Formula (B-I), where substituents are selected as appropriate.
- the monoketone (iii) may be oxidized to a diketone (iv) with HBr in DMSO.
- Diketones (iv) can be condensed with aldehydes (v) in the presence of ammonium acetate to yield an imidazole (vi).
- the protecting group (PG) substituted nitrogen can then be depretoected under appropriate conditions to yield a compound of Formula (C-I).
- Scheme 3 for the Preparation of Exemplary ALK5 Inhibitors of Formula (D-I) In one method, compounds of Formula (I) are prepared according to Scheme 1.
- bromo-substituted pyrazoles (i) are reacted with an aryl- or heteroaryl-boronic acid (or ester) (iii) in the presence of an appropriate transition metal catalyst (e.g. palladium-based) to generate compounds of Formula (I).
- an appropriate transition metal catalyst e.g. palladium-based
- bromo-substituted pyrazoles can be converted to a boronic ester, or ester, (ii) via a borylation reaction.
- the resultant compound (ii) can then be coupled with an appropriate aryl- or heteroaryl-halide (or pseudohalide) (iv) to generate compounds of Formula (I).
- the pH of the reaction mixture was adjusted to 8 with saturated aqueous NaHCO 3 solution and extracted with CH 2 Cl 2 (2 ⁇ 10 mL). The combined organic extracts were washed with brine (10 mL) and dried over anhydrous Na 2 SO 4 , filtered, and evaporated. The residue was purified on silica gel (ISCO gold, 40 g; 0% to 20% CH 2 Cl 2 /MeOH over 15 min) to give the desired imidazole product, which was dissolved in 1 N HCl (20 mL) and heated at 70°C for 4 h. The reaction mixture was allowed to cool to 0°C and then it was neutralized with saturated aqueous NaHCO 3 solution.
- Step B Preparation of Compound 1
- acetic acid 79 mg, 1.3 mmol, 75 ⁇ L
- phenylmethanamine 106 mg, 0.99 mmol
- reaction mixture was quenched by addition of water (0.10 mL) at 0°C and was then concentrated under reduced pressure to give a residue that was purified by silica gel chromatography (0 ⁇ 10% MeOH in DCM) to afford 240 mg of 1-(5-([1,2,4]triazolo[1,5- a]pyridin-6-yl)-4-(6-methylpyridin-2-yl)-1H-imidazol-2-yl)-N-benzylmethanamine as an off- white solid.
- reaction mixture was cooled to 0 °C and methyl tert-butyl ether (300 mL) was added, desired solid was precipitated and then filtered to afford 6-bromoquinolin-3-ol (A1-c) (7.0 g, 31.2 mmol, 55.8% yield) as brown solid.
- the suspension was degassed under vacuum and purged with CO several times.
- the mixture was stirred under CO (50 psi) at 80°C for 15 hours.
- Water (70 mL) was added to the reaction mixture and the aqueous phase was extracted with ethyl acetate (20 mL x 3).
- the combined organic phase was washed with brine (50 mL x 2), dried with anhydrous Na 2 SO 4 , filtered and concentrated in vacuum.
- Water (20 mL) was added and the aqueous phase was extracted with ethyl acetate (10 mL x 3).
- the combined organic phase was washed with brine (20 mL x 2), dried with anhydrous Na 2 SO 4 , filtered and concentrated in vacuum.
- Step 6 Preparation of tert-butyl N-[2-[[6-[2-(6-methyl-2-pyridyl)-2-oxo-acetyl]-3- quinolyl]oxy]ethyl]carbamate (Compound A1)
- tert-butyl N-[2-[[6-[2-(6-methyl-2-pyridyl)-2-oxo-ethyl]-3-quinolyl]oxy] ethyl]carbamate (A1-f) 450 mg, 1.07 mmol, 1.0 equiv) in MeCN (15 mL) was added Pd/C (518 mg, 5% purity, 0.2 equiv) under N 2 .
- reaction mixture was poured into H 2 O (20 mL), and then the pH of the mixture was adjusted to ⁇ 8 by adding aq. NaHCO 3 , extracted with EtOAc (20 mL) for twice.
- the organic lawyers were washed with brine (20 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue.
- Step 3 Preparation of tert-butyl N-[2-[7-[2-(6-methyl-2-pyridyl)-2-oxo-acetyl] quinoxalin-2-yl]oxyethyl]carbamate (Compound A2)
- tert-butyl N-[2-[7-[2-(6-methyl-2-pyridyl)-2-oxo-ethyl]quinoxalin-2-yl] oxyethyl]carbamate (A2-d) (1.5 g, 3.55 mmol, 1.0 equiv) in MeCN (20 mL) was added Pd/C (0.5 g, 10% purity) at 20°C, the suspension was degassed under vacuum and purged with O 2 several times.
- Step 3 Preparation of tert-butyl N-(2-fluorophenyl)-N-(2-oxoethyl)carbamate (Compound C1.1-d) To a mixture of tert-butyl N-allyl-N-(2-fluorophenyl)carbamate (1.1-c) (10.3 g, 41.0 mmol, 1.0 equiv) in THF (100 mL) and H 2 O (100 mL) was added K 2 OsO 4 .
- reaction mixture was quenched by addition of H 2 O ⁇ 10 mL ⁇ at 20°C, and then extracted with EtOAc (10 mL). The combined organic layers were washed with brine (10 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue.
- reaction mixture was quenched by addition H 2 O (10 mL) at 20 °C, and then extracted with EtOAc (10 mL x 3). The combined organic layers were washed with brine (10 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue.
- Step 2 Preparation of tert-butyl N-[2-[7-[2-[2-(2-fluorophenyl)ethyl]-4-(6-methyl-2- pyridyl)-1H-imidazol-5-yl]quinoxalin-2-yl]oxyethyl]carbamate (Compound C2.1)
- Step 3 Preparation of tert-butyl N-[2-[7-[2-[(2-fluorophenoxy)methyl]-4-(6-methyl-2- pyridyl)-1H-imidazol-5-yl]quinoxalin-2-yl]oxyethyl]carbamate (Compound 3.1-d)
- a mixture of tert-butyl N-[2-[7-[2-(6-methyl-2-pyridyl)-2-oxo-acetyl]quinoxalin-2- yl]oxyethyl]carbamate (A2) 150 mg, 343 umol, 1.0 equiv
- 2-(2- fluorophenoxy)acetaldehyde (3.1-c) 79 mg, 515 umol, 1.5 equiv
- THF 3 mL
- reaction mixture was quenched by addition H 2 O (10 mL) at 20°C, and then extracted with EtOAc (10 mL x 3). The combined organic layers were washed with brine (20 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue.
- Step 4 Preparation of 2-[7-[2-[(2-fluorophenoxy)methyl]-4-(6-methyl-2-pyridyl)-1H- imidazol -5-yl]quinoxalin-2-yl]oxyethanamine (Compound C3.1)
- Step 1 Preparation of tert-butyl N-[2-[7- [2-(dimethoxymethyl)- 4-(6-methyl-2-pyridyl)- 1H-imidazol-5-yl]quinoxalin-2-yl]oxyethyl]carbamate (Compound C4.1-a) To a mixture of tert-butyl N-[2-[7-[2-(6-methyl-2-pyridyl)-2-oxo-acetyl]quinoxalin-2-yl] oxyethyl]carbamate (A2) (2 g, 4.58 mmol, 1.0 eq) and 2,2-dimethoxyacetaldehyde (954 mg, 9.16 mmol, 830 uL, 2.0 eq) in THF (20 mL) was added NH 4 OAc (1.77 g, 22.9 mmol, 5.0 eq), and then stirred at 35 °C for 12h.
- reaction mixture was poured into ice-water (20 mL) and stirred for 10 min.
- the aqueous phase was extracted with ethyl acetate (20 mL x 2).
- the combined organic phase was washed with brine (20 mL), dried with anhydrous Na 2 SO 4 , filtered and concentrated in vacuum.
- Step 3 Preparation of tert-butyl N-[2-[7-[2-formyl-4-(6-methyl-2-pyridyl)-1H-imidazol- 5-yl]quinoxalin-2-yl]oxyethyl]carbamate (Compound C4.1-c)
- To a solution of 5-[3-(2-aminoethoxy)quinoxalin-6-yl]-4-(6-methyl-2-pyridyl) -1H- imidazole-2-carbaldehyde (C4.1-b) (1.3 g, 3.47 mmol, 1 eq) in THF (20 mL) was added NaHCO 3 (584 mg, 6.94 mmol, 2 eq) in H 2 O (2 mL) and Boc 2 O (909 mg, 4.17 mmol, 957 uL, 1.2 eq) at 25°C, and then stirred at 25°C for 1 hour.
- Step 5 Preparation of N-[[5-[3-(2-aminoethoxy)quinoxalin-6-yl]-4-(6-methyl-2-pyridyl)- 1H-imidazol-2-yl]methyl]-5-fluoro-pyridin-2-amine (Compound C4.1)
- the resulting solution was stirred for 5 h at 50 ° C.
- the reaction mixture was cooled to 25 ° C with a water/ice bath, diluted with 100 mL of water and extracted with 3x100 mL of dichloromethane and the organic layers were combined, washed with 3x100 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
- the crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm.
- Step 3 Preparation of ethyl 3-(6-methylpyridin-2-yl)-3-oxopropanoate
- ethyl acetate 45.0 mL
- sodium hydride 1.6 g, 39.6 mmol, 2.0 equiv.
- Step 4 Preparation of ethyl (E)-3-(6-methylpyridin-2-yl)-3-((2-oxopyrrolidin-1- yl)imino)propanoate
- a solution of ethyl 3-(6-methylpyridin-2-yl)-3-oxopropanoate 1.5 g, 7.2 mmol, 1.0 equiv.
- 1- aminopyrrolidin-2-one hydrochloride 1.2 g, 8.6 mmol, 1.2 equiv.
- Step 5 Preparation of 2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole-3- carboxylic acid
- ethyl (E)-3-(6-methylpyridin-2-yl)-3-((2-oxopyrrolidin-1-yl)imino) propanoate 1.0 g, 3.5 mmol, 1.0 equiv.
- sodium ethoxide 476.0 mg, 7.0 mmol, 2.0 equiv.
- the resulting solution was stirred for 24 h at 80 ° C. LCMS showed the reaction was completed.
- Step 6 Preparation of 3-bromo-2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole
- 2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole-3- carboxylic acid (200.0 mg, 0.82 mmol, 1.0 equiv.) in N,N-dimethylformamide (5.0 mL) with an inert atmosphere of nitrogen, was added N-bromosuccinimide (445.0 mg, 2.5 mmol, 3.0 equiv.).
- N-bromosuccinimide 445.0 mg, 2.5 mmol, 3.0 equiv.
- the resulting solution was diluted with 20 mL of water and extracted with 3x20 mL of dichloromethane and the organic layers were combined, washed with 3x20 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
- the crude product was purified by Flash-Prep- HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm.
- Step 7 Preparation of benzyl (4-((6-(2-(6-methylpyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinolin-3-yl)oxy)butyl)carbamate
- 3-bromo-2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b] pyrazole (100.0 mg, 0.36 mmol, 1.0 equiv.) in 1,4-dioxane/water (5.0 mL/0.5 mL) with an inert atmosphere of nitrogen, was added (4-((6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinolin- 3-yl)oxy)butyl)carbamate (205.0 mg, 0.43 mmol, 1.2 equiv.), [1,1'-bis(diphenylphosphin
- the resulting solution was stirred for 4 h at 80 o C.
- the reaction mixture was cooled to 25 ° C with a water/ice bath and diluted with 10 mL of water and extracted with 3x10 mL of dichloromethane and the organic layers were combined, washed with 3x10 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
- the crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm.
- Step 8 Preparation of a TFA salt of 4-((6-(2-(6-methylpyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinolin-3-yl)oxy)butan-1-amine (Compound D-3.1 To a solution of benzyl (4-((6-(2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3- yl)quinolin-3-yl)oxy)butyl)carbamate (60.0 mg, 0.11 mmol, 1.0 equiv.) trifluoroacetic acid (2.0 mL) with inert atmosphere of nitrogen, the resulting solution was stirred for 4 h at 50 ° C.
- the reaction mixture was cooled to 25 ° C with a water/ice bath, diluted with 10 mL of dichloromethane and concentrated under vacuum.
- the crude product was purified by Flash-Prep- HPLC with the following conditions (IntelFlash-1): C18 Column; mobile phase, water (with 0.5% trifluoroacetic acid) and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm.
- Example 4-1B Synthesis of a TFA salt of N-(2-aminoethyl)-6-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro- 4H-pyrrolo[1,2-b]pyrazol-3-yl)quinoline-3-carboxamide (Compound D-3.15)
- Compound D-3.15 was prepared according to synthetic Method A for compounds of Formula (D-1) with the appropriate starting materials, as shown below.
- Step 1 Preparation of benzyl (2-(6-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinoline-3-carboxamido)ethyl)carbamate
- 3-bromo-2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazole (160.0 mg, 0.5 mmol, 1.0 equiv.) in 1,4-dioxane/water (5.0 mL/0.5 mL) with an inert atmosphere of nitrogen, was added benzyl (2-(6-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)quinoline-3-carboxamido)ethyl)carbamate (285.2 mg, 0.6 mmol, 1.2 equiv
- the resulting solution was stirred for 10 h at 80 o C.
- the reaction mixture was cooled to 25 o C with a water/ice bath and diluted with 50 mL of water and extracted with 3x50 mL of dichloromethane and the organic layers were combined, washed with 3x50 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
- the crude product was purified by Flash-Prep- HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm.
- Step 2 Preparation of N-(2-aminoethyl)-6-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinoline-3-carboxamide TFA salt (Compound D-3.15)
- benzyl (2-(6-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinoline-3-carboxamido)ethyl)carbamate 116.0 mg, 0.2 mmol, 1.0 equiv.
- trifluoroacetic acid 2.0 mL
- the reaction mixture was cooled to 25 o C with a water/ice bath, diluted with 10 mL of dichloromethane and concentrated under vacuum.
- the crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): C18 Column; mobile phase, water (with 0.5% trifluoroacetic acid) and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm.
- Example 4-1C Synthesis of a TFA salt of 2-(7-(2-(6-(trifluoromethyl)pyridin-2-yl)-5,6-dihydro-4H-pyrrolo [1,2-b]pyrazol-3-yl)quinoxalin-2-yl)ethan-1-amine (Compound D-3.22)
- Compound 3.22 was prepared according to synthetic Method A for compounds of Formula (D-1) with the appropriate starting materials, as shown below.
- Step 1 Preparation of benzyl (2-(7-(2-(6-(trifluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinoxalin-2-yl)ethyl)carbamate
- 3-bromo-2-(6-(trifluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazole 208 mg, 0.48 mmol, 1.2 equiv.
- 1,4-dioxane/water 5.0 mL/0.5 mL
- benzyl (2-(7-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)quinoxalin-2-yl)ethyl)carbamate (130.0 mg, 0.4 mmol, 1.0 equiv.),
- the resulting solution was stirred for 6 h at 80 o C.
- the reaction mixture was cooled to 25 o C with a water/ice bath and diluted with 50 mL of water and extracted with 3x50 mL of dichloromethane and the organic layers were combined, washed with 3x50 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
- the crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm.
- Step 2 Preparation of 2-(7-(2-(6-(trifluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinoxalin-2-yl)ethan-1-amine TFA salt (Compound D-3.22)
- benzyl (2-(7-(2-(6-(trifluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinoxalin-2-yl)ethyl)carbamate (102.0 mg, 0.18 mmol, 1.0 equiv.) in trifluoroacetic acid (3.0 mL) with inert atmosphere of nitrogen, the resulting solution was stirred for 4 h at 50 o C.
- the reaction mixture was cooled to 25 o C with a water/ice bath, diluted with 20 mL of dichloromethane and concentrated under vacuum.
- the crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): C18 Column; mobile phase, water (with 0.5% trifluoroacetic acid) and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm.
- Step 1 Preparation of (E)-3-(dimethylamino)-1-(6-methylpyridin-2-yl)prop-2-en-1-one
- 1-(6-methylpyridin-2-yl)ethanone 270.0 mg, 2.0 mmol, 1.0 equiv.
- (dimethoxy- methyl)dimethylamine 262.2 mg, 2.2 mmol, 1.1 equiv.
- the resulting solution was stirred for 7 h at 105 o C.
- the reaction mixture was cooled to 25 o C with a water/ice bath, diluted with 100 mL of water and extracted with 3x100 mL of dichloromethane and the organic layers were combined, washed with 3x100 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
- the crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm.
- Step 2 Preparation of 2-methyl-6-(1H-pyrazol-3-yl)pyridine To a solution of (E)-3-(dimethylamino)-1-(6-methylpyridin-2-yl)prop-2-en-1-one (190.0 mg, 1.0 mmol, 1.0 equiv.) in ethanol (5.0 mL) with an inert atmosphere of nitrogen, was added hydrazine hydrate (2.0 mL).
- Step 3 Preparation of 2-(4-bromo- pyrazol-3-yl)-6-methylpyridine
- 2-methyl-6-(1H-pyrazol-3-yl)pyridine 318 mg, 2.0 mmol, 1.0 equiv.
- dichloromethane 6.0 mL
- N-bromosuccinimide 391.6 mg, 2.2 mmol, 1.1 equiv.
- the resulting solution was diluted with 20 mL of water and extracted with 3x20 mL of dichloromethane and the organic layers were combined, washed with 3x20 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
- the crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm. 430 mg (90%) 2-(4-bromo-1H-pyrazol-3-yl)-6- methylpyridine was obtained as a yellow solid.
- Step 4 Preparation of tert-butyl 4-bromo-3-(6-methylpyridin-2-yl)-1H-pyrazole-1-carboxylate
- 2-(4-bromo-1H-pyrazol-3-yl)-6-methylpyridine 474.0 mg, 2.0 mmol, 1.0 equiv.
- tetrahydrofuran 8.0 mL
- di-tert- butyl pyrocarbonate 523.7 mg, 2.4 mmol, 1.2 equiv.
- triethylamine 303.0 mg, 3.0 mmol, 1.5 equiv.
- the resulting solution was stirred for 4 h at 25 o C.
- the resulting solution was diluted with 50 mL of water and extracted with 3x50 mL of dichloromethane and the organic layers were combined, washed with 3x50 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
- the crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm.
- Step 5 Preparation of tert-butyl 4-(3-(4-(((benzyloxy)carbonyl)amino)butoxy)quinolin-6-yl)-3- (6-methylpyridin-2-yl)-1H-pyrazole-1-carboxylate
- tert-butyl 4-bromo-3-(6-methylpyridin-2-yl)-1H-pyrazole-1-carboxylate (202.0 mg, 0.6 mmol, 1.0 equiv.) in 1,4-dioxane/water (5.0 mL/0.5 mL) with an inert atmosphere of nitrogen, was added (4-((6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinolin-3- yl)oxy)butyl)carbamate (343.0 mg, 0.72 mmol, 1.2 equiv.), [1,1'-Bis(diphenylphosphino)
- the resulting solution was stirred for 2 h at 80 o C.
- the reaction mixture was cooled to 25 o C with a water/ice bath and diluted with 20 mL of water and extracted with 3x20 mL of dichloromethane and the organic layers were combined, washed with 3x20 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
- the crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm.
- Step 6 Preparation of 4-((6-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)quinolin-3-yl)oxy)butan-
- 4-((6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinolin-3- yl)oxy)butyl)carbamate 200.0 mg, 0.33 mmol, 1.0 equiv.
- trifluoroacetic acid 5.0 mL
- the resulting solution was diluted with 10 mL of dichloromethane and concentrated under vacuum.
- Step 1 Preparation of (2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b] pyrazol-3-yl)boronic acid
- 3-bromo-2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazole 100.0 mg, 0.3 mmol, 1.0 equiv.
- 2-isopropoxy-4,4,5,5-tetramethyl-1,3,2-dioxaborolane 296.2 mg, 1.6 mmol, 5.0 equiv.
- the reaction mixture was cooled to -78 o C by the addition of n- butyllithium (51.0 mg, 0.8 mmol, 2.5 equiv.) to the reaction at -78 o C.
- the resulting solution was stirred for 3 h at -78 o C.
- the reaction was then quenched by the addition of water/ice.
- the resulting solution was diluted with 50 mL of water and extracted with 3x50 mL of dichloromethane and the organic layers were combined, washed with 3x50 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
- Step 2 Preparation of benzyl (2-(4-(5-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)-2-fluorophenyl)-1H-pyrazol-1-yl)ethyl)carbamate
- benzyl (2-(4-(5-bromo-2-fluorophenyl)-1H-pyrazol-1-yl)ethyl)carbamate 220.0 mg, 0.5 mmol, 1.0 equiv.
- 1,2-dimethoxyethane 5.0 mL
- the resulting solution was stirred for 2 h at 80°C.
- the reaction mixture was cooled to 25 o C with a water/ice bath, diluted with 50 mL of water and extracted with 3x50 mL of dichloromethane and the organic layers were combined, washed with 3x50 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
- the crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm.
- Step 3 Preparation of a TFA salt of 2-(4-(5-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)-2-fluorophenyl)-1H-pyrazol-1-yl)ethan-1-amine (Compound D-6.3)
- benzyl (2-(4-(5-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)-2-fluorophenyl)-1H-pyrazol-1-yl)ethyl)carbamate 17.
- the reaction mixture was cooled to 25 o C with a water/ice bath, diluted with 10 mL of dichloromethane and concentrated under vacuum.
- the crude product was purified by Flash-Prep- HPLC with the following conditions (IntelFlash-1): C18 Column; mobile phase, water (with 0.5% trifluoroacetic acid) and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm.
- Step 1 Preparation of 2-(2-(4-(2-fluoro-5-(3-(6-methylpyridin-2-yl pyrazol-4-yl)phenyl)- 1H-pyrazol-1-yl)ethyl)isoindoline-1,3-dione
- 2-(4-[2-fluoro-5-[3-(6-methylpyridin-2-yl)-1H-pyrazol-4- yl]phenyl]pyrazol-1-yl)ethanol 600.0 mg, 1.65 mmol, 1.0 equiv.
- phthalimide 485.1 mg, 3.3 mmol, 2.0 equiv.
- triphenyl phosphine 657.5 mg, 2.5 mmol, 1.5 equiv.
- diisopropyl azodiformate 505.0 mg, 2.5 mmol, 1.5 equiv.
- the resulting solution was stirred for 4 h at 25 o C.
- the resulting solution was diluted with 50 mL of water and extracted with 3x50 mL of dichloromethane and the organic layers were combined, washed with 3x50 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
- the crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 80.0% in 20 min); Detector, UV 254 nm.
- Step 2 Preparation of 2-(4-(2-fluoro-5-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)-1H- pyrazol-1-yl)ethan-1-amine
- the resulting solution was stirred for 1 h at 70 o C in an oil bath, cooled to 25 o C with a water/ice bath and concentrated under reduced pressure.
- the crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm.
- Step 1 Preparation of methyl (S)-4-((tert-butoxycarbonyl)amino)-5-((2-(4-(2-fluoro-5-(3-(6- methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)-1H-pyrazol-1-yl)ethyl)amino)-5-oxopentanoate
- 2-(4-(2-fluoro-5-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)-1H- pyrazol-1-yl)ethan-1-amine 150.0 mg, 0.41 mmol, 1.0 equiv.) in tetrahydrofuran (3.0 mL) with an inert atmosphere of nitrogen, was added N,N-carbonyldiimidazole (79.4 mg, 0.49 mmol, 1.2 equiv.), methyl (4S)-4-(benzy
- the resulting solution was stirred for 6 h at 25 o C.
- the resulting solution was diluted with 50 mL of water and extracted with 3x50 mL of dichloromethane and the organic layers were combined, washed with 3x50 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
- the crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 80.0% in 20 min); Detector, UV 254 nm.
- Step 2 Preparation of a TFA salt of methyl (S)-4-amino-5-((2-(4-(2-fluoro-5-(3-(6- methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)-1H-pyrazol-1-yl)ethyl)amino)-5-oxopentanoate T
- methyl (S)-4-((tert-butoxycarbonyl)amino)-5-((2-(4-(2-fluoro-5-(3-(6- methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)-1H-pyrazol-1-yl)ethyl)amino)-5-oxopentanoate 138.0 mg, 0.23 mmol, 1.0 equiv.
- the resulting solution was stirred for 2 h at 25 o C.
- the reaction mixture was diluted with 10 mL of dichloromethane and concentrated under vacuum.
- the crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): C18 Column; mobile phase, water (with 0.5% trifluoroacetic acid) and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm.
- Step 3 Preparation of (S)-4-amino-5-((2-(4-(2-fluoro-5-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4- yl)phenyl)-1H-pyrazol-1-yl)ethyl)amino)-5-oxopentanoic acid TFA salt (Compound D-7.14)
- Compound D-7.13 (190.0 mg, 0.38 mmol, 1.0 equiv.) in tetrahydrofuran/water (2.0 mL/2.0 mL) with an inert atmosphere of nitrogen, was added lithium hydroxide monohydrate (31.9 mg, 0.76 mmol, 2.0 equiv.).
- the resulting solution was stirred for 6 h at 25 o C.
- the pH of the reaction was adjusted to 7 with trifluoroacetic acid.
- the resulting mixture was concentrated under vacuum and purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): C18 Column; mobile phase, water (with 0.5% trifluoroacetic acid) and acetonitrile (10.0% acetonitrile up to 50.0% in 15 min); Detector, UV 254 nm.
- Step 1 Synthesis of 4-((S)-2-((S)-2-amino-3-methylbutanamido)-5- ureidopentanamido)benzyl ((5-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-4-(6-methylpyridin-2- yl)-1H-imidazol-2-yl)methyl)(2,6-difluorophenethyl)carbamate (Intermediate 3).
- Step 2 Preparation of tert-butyl 3-[2-amino-3-(3-tert-butoxy-3-oxo-propoxy) -2-[(3-tert- butoxy-3-oxo-propoxy)methyl]propoxy]propanoate.
- 2-amino-2-(hydroxymethyl)propane-1,3-diol (30 g, 247.66 mmol, 35.71 mL, 1 eq) in DMSO (83 mL) was added dropwise NaOH (5 M, 4.96 mL, 0.1 eq) at 0-15°C over 5 min.
- Step 3 Preparation of benzyl 5-[[2-(3-tert-butoxy-3-oxo-propoxy)-1,1-bis[(3-tert- butoxy- 3-oxo-propoxy)methyl]ethyl]amino]-5-oxo-pentanoate.
- 5-benzyloxy-5-oxo-pentanoic acid (4.40 g, 19.78 mmol, 1 eq) in DMF (20 mL) was added dropwise HATU (9.02 g, 23.73 mmol, 1.2 eq) at 15°C.
- reaction mixture was quenched by the addition or H 2 O (200 mL) at 0°C, and extracted with EtOAc (100 mL x 3). The combined organic layers were washed with brine (200 mL x 2), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue.
- Step 5 Preparation of benzyl 5-oxo-5-[[2-[3-oxo-3-(2,3,4,5,6-pentafluorophenoxy) propoxy]-1,1-bis[[3-oxo-3-(2,3,4,5,6-pentafluorophenoxy)propoxy]methyl]ethyl]amino] pentanoate.
- Step 6 Preparation of 5-[[2-[3-[4-[(2R,3R,4R,5R,6R)-3-acetamido-4,5-diacetoxy-6- (acetoxymethyl)tetrahydropyran-2-yl]oxybutylamino]-3-oxo-propoxy]-1,1-bis[[3-[4- [(2R,3R,4R,5R,6R)-3-acetamido-4,5-diacetoxy-6-(acetoxymethyl)tetrahydropyran-2- yl]oxybutylamino]-3-oxo-propoxy]methyl]ethyl]amino]-5-oxo-pentanoic acid.
- Step 7 Preparation of (2,3,4,5,6-pentafluorophenyl) 5-[[2-[3-[4-[(2R,3R,4R,5R,6R)-3- acetamido-4,5-diacetoxy-6-(acetoxymethyl)tetrahydropyran-2-yl]oxybutylamino]-3-oxo- propoxy]-1,1-bis[[3-[4-[(2R,3R,4R,5R,6R)-3-acetamido-4,5-diacetoxy-6- (acetoxymethyl)tetrahydropyran-2-yl]oxybutylamino]-3-oxo-propoxy]methyl]ethyl] amino]-5-oxo-pentanoate.
- Step 8 Preparation of Intermediate 4.
- Step 9 Preparation of Compound 3.1. To a solution of Intermediate 4 (16.2 mg, 6.5 umol, 1.0 equiv) in MeOH (5 mL) was added NaOMe (6.9 mg, 0.13 mmol, 20 equiv), the mixture was stirred at 20°C for 1 h. LCMS showed the reaction was complete. The reaction mixture was adjusted to pH ⁇ 5-6 using acetic acid, and then was concentrated under reduced pressure to give a residue.
- the residue was purified by prep-HPLC (column: Teledyne RediSep Prep C18, 100 ⁇ , 5 ⁇ M (length: 150 mm; ID: 20 mm); mobile phase: [water (0.1% TFA)-AcN (0.1% TFA)]; B%: 10%-60%, 30 min).
- HEK293 SBE-LUC reporter cells i.e., containing luciferase under the control of the SMAD-binding element (SBE), a TGF ⁇ responsive transcriptional element
- SBE SMAD-binding element
- TGF ⁇ responsive transcriptional element transfected to stably express full length human ASGR1 were seeded in 96 well plates at about 4 x 10 5 cells/well in an assay media of MEM containing 0.5% FBS, 1% NEAA, 1% NaPyr, and 1% Pen/Strep.
- ALK5 Inhibitor-GalNAc conjugates and vehicle controls were added to wells containing either HEK293 SBE-LUC or ASGR1-HEK293 SBE-LUC cells in a dose titration ranging from about 5 ⁇ M to about 0.06 nM.
- human TGF ⁇ 1 was added (PeproTech Inc.) to a final concentration of 1.6 ng/ml followed by an additional 18 hour incubation.
- Luciferase Steady Glo reagent Promega Corporation was added to each well, incubated for 10 minutes with shaking, and then luciferase activity was determined by measuring luminescence with an Envision Plate Reader (Perkin-Elmer Inc.). Using Prism Software v7.04 (GraphPad Inc.), data were fit with a four-parameter non-linear regression to calculate IC 50 values.
- ASGR mammalian asialoglycoprotein receptor
- GalNAc N-acetylgalactosamine
- GalNAc is a well-defined liver-targeted moiety having high affinity to ASGR1.
- HEK293 transfected with ASGR1 is known model for detecting uptake of GalNAc conjugates (see, e.g., Tanowitz et al., Nucleic Acid Res. 45:12388, 2017).
- the results for exposure of the ASGR1- positive cells (Conjugate X + ASGR1) and ASGR1-negative cells (Conjugate X) as compared to DMSO control are shown in Figures 1A (Conjugate 3.1), 1B (Conjugate 3.2), and 1C (Conjugate 3.3).
- DMEM-C with Geneticin DMEM supplemented with 10% fetal bovine serum, 1X NEAA, 1 mM Pyruvate, 2 mM glutamine, 50 ⁇ g/mL penicillin, 50 U/mL streptomycin and 400 ⁇ g/mL of Geneticin.
- the assay media is MEM supplemented with 0.5% fetal bovine serum, 1X NEAA, 1mM Pyruvate, 50 ⁇ g/mL penicillin and 50 U/mL streptomycin.
- Compounds are assayed to measure their activity as ALK5 inhibitors.
- Enzyme Inhibition Assays were performed by Reaction Biology Corp (Malvern, PA). 1 mg/mL of peptide subtrate (casein) and 10 uM ATP were prepared in a mixture of fresh reaction buffer. The kinase was delivered into the substrate solution which was gently mixed. Compounds in 100% DMSO were added to the kinase reaction mixture by Acoustic technology (Echo550; nanoliter range) and the mixture was incubated for 20 min at room temperature. 33 P- ATP (Specific activity 10 uCi/uL) was added into the reaction mixture to initiate the reaction and the reaction mixture was incubated for 2 hours at room temperature.
- Table 26 Key Compounds having an IC 50 value between 0.1 nM and 50 nM are denoted 50 nM and 100 nM as +++, 100 nM and 1000 nM as ++, and 1000 nM to 10,000 nM as +.
- the data in Table 26 shows that the ALK5 inhibitors of this disclosure are capable of diffusing into the cells and inhibiting ALK5 serine/threonine kinase activity at nanomolar and sub-nanomolar concentrations.
- TGF ⁇ Reporter Assay The HEK293 SBE-LUC reporter cells from Example 1 were grown in DMEM-C with Geneticin (DMEM supplemented with 10% fetal bovine serum, 1X NEAA, 1mM Pyruvate, 2mM glutamine, 50 ⁇ g/mL penicillin, 50 U/mL streptomycin and 400 ug/mL of Geneticin).
- the assay media was MEM supplemented with 0.5% fetal bovine serum, 1X NEAA, 1 mM Pyruvate, 50 ⁇ g/mL penicillin and 50 U/mL streptomycin.
- the HEK293 SBE-LUC reporter cells were harvested from the tissue culture flasks by incubation in small quantity of Versene at room temperature for three to five minutes after the media in the flask is removed and cells rinsed with PBS. Cells were counted and diluted in the assay media at ⁇ 0.8 x 10 6 cells/mL then 50 uL/well were added to 96-well assay plate. Test samples (at desired concentrations diluted in assay media) were added to assay plate containing the 50 uL/well of cells (or media only), 50 uL per well, and incubated for 5-6 hours at 37°C in a 5% CO 2 humidified incubator.
- TGF ⁇ diluted to 12.5ng/mL in the assay media was added to the plate.
- Controls included TGF ⁇ titration (from 25 to 0 ng/mL) without inhibitors, and media only (without cells, inhibitor or TGF ⁇ ). Plates were incubated at 37°C in a 5% CO 2 humidified incubator for 18h.
- Luciferase substrate solution was subsequently added at 75uL per well, incubated in dark with shaking at room temperature for 10 min, and luminescence Table 27 Key: Compounds having an IC 50 value between 0.1 nM and 10 nM are denoted as ++++, 10 nM and 100 nM as +++, 100 nM and 1000 nM as ++, and 1000 nM to 10,000 nM as +.
- ++++ Compounds having an IC 50 value between 0.1 nM and 10 nM are denoted as ++++, 10 nM and 100 nM as +++, 100 nM and 1000 nM as ++, and 1000 nM to 10,000 nM as +.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Public Health (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Veterinary Medicine (AREA)
- Epidemiology (AREA)
- Biochemistry (AREA)
- Molecular Biology (AREA)
- Gastroenterology & Hepatology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Conjugates comprising GalNAc moiety and a TGFβR1 inhibitor are provided. In some embodiments, the conjugates are useful for the treatment of viral infections, cancer, and fibrosis.
Description
GALNAC-TGFΒR1 INHIBITOR CONJUGATES FOR THE TREATMENT OF LIVER DISEASES BACKGROUND Fibrosis is the formation of excess fibrous connective tissue or scar tissue in an organ or tissue in a reparative or reactive process. Fibrosis can occur in many tissues within the body, typically as a result of inflammation or damage, which include the lungs, liver, heart, and brain. Scar tissue blocks arteries, immobilizes joints and damages internal organs, wreaking havoc on the body's ability to maintain vital functions. Every year, millions of people are hospitalized due to the damaging effects of fibrosis. However, current therapeutics for treating fibrotic diseases are lacking or have drawbacks. Thus, there remains a considerable need for alternative or improved treatments for fibrotic diseases. SUMMARY In one aspect, the present disclosure provides a conjugate of Formula (I): wherein
Region G comprises at least one GalNAc moiety; Region L3 is a connector that connects Region G to Region Inh; Region Inh comprises a TGFβR1 inhibitor; z is 1, 2, 3, 4, or 5; or a pharmaceutically acceptable salt thereof. In another aspect, the present disclosure relates to a pharmaceutical composition comprising a conjugate of Formula (I) as described herein and a pharmaceutically acceptable carrier. In another aspect, the present disclosure relates to a method for treating a disease mediated by TGFβR1 activity in subject in need thereof, such as liver cancer or liver fibrosis,
comprising administering to the subject an effective amount of a conjugate of Formula (I) or a pharmaceutical composition comprising a conjugate of Formula (I) as described herein. In another aspect, the present disclosure provides a method for enhancing an immune response (e.g., an anti-cancer immune response) in a subject comprising administering to the subject an effective amount of a conjugate of Formula (I) or a pharmaceutical composition comprising a conjugate of Formula (I) as described herein. Disclosed herein is a compound of Formula (XX): [Inh]−Z (XX) wherein Region Inh comprises a TGFβR1 inhibitor; and Z is the residual portion of a released, cleavable linker or comprises a non-cleavable linker and a residual portion of a degraded Region G; or a pharmaceutically acceptable salt thereof. BRIEF DESCRIPTION OF THE DRAWINGS The novel features of the disclosure are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present disclosure will be obtained by reference to the following detailed description that sets forth illustrative aspects, in which the principles of the disclosure are utilized. FIGS. 1A-1C illustrate the activity of GalNAc−TGFβR1 inhibitor conjugates in a HEK293 cell reporter assay (ASGR-positive and ASGR-negative cells: Fig. 1A, Conjugate 3.1; Fig. 1B, Conjugate 3.2; Fig. 1C, Conjugate 3.3). DETAILED DESCRIPTION Additional aspects and advantages of the present disclosure will become apparent to those skilled in this art from the following detailed description, wherein illustrative aspects of the present disclosure are shown and described. As will be appreciated, the present disclosure is capable of other and different aspects, and its several details are capable of modifications in
various respects, all without departing from the disclosure. Accordingly, the descriptions are to be regarded as illustrative in nature, and not as restrictive. Where values are described as ranges, it will be understood that such disclosure includes the disclosure of all possible sub-ranges within such ranges, as well as specific numerical values that fall within such ranges irrespective of whether a specific numerical value or specific sub- range is expressly stated. Activin receptor-like kinase 5 (ALK5), which is also commonly known as transforming growth factor beta receptor 1 (TGFβR1), is a serine/threonine kinase transmembrane receptor. It is a part of the TGFβ signaling pathway and is involved in signal transduction from the cell surface to the cytoplasm. The TGFβ signaling pathway regulates gene expression of genes involved in cellular processes such as differentiation, apoptosis, wound healing, and cell growth. In the absence of TGFβ ligands, ALK5 remains a homodimeric cell surface receptor. However, ligand binding to type II TGFβ receptor (TGFβR2) induces the formation of the TGFβR1/TGFβR2 complex, which leads to phosphorylation of Mothers Against Decapentaplegic homolog 2 (Smad2) and Mothers Against Decapentaplegic homolog 3 (Smad3) and subsequent modulation of a number of downstream signaling targets involved in the regulation of gene expression. ALK5 inhibitors and conjugates thereof may be useful for treatment and/or prevention, e.g., vaccination, of cancer, autoimmune diseases, inflammation, fibrosis, sepsis, allergy, asthma, graft rejection, graft-versus-host disease, immunodeficiencies, and infectious diseases. In some aspects, an ALK5 inhibitor has an IC50 value of between 0.1 nM and 1000 nM, between 0.1 nm and 100 nM, or between 0.1 nM and 80 nM in an ALK5 enzyme inhibition assay and/or in a TGFβR1 reporter assay. Exemplary ALK5 enzyme inhibition and TGFβR1 reporter assays are as set forth in the example section. Definitions Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this disclosure belongs. As used in the specification and claims, the singular form “a”, “an” and “the” includes plural references unless the context clearly dictates otherwise.
The term “about” as used herein in the context of a number refers to a range centered on that number and spanning 10% less than that number and 10% more than that number. The term “about” used in the context of a range refers to an extended range spanning 10% less than that the lowest number listed in the range and 10% more than the greatest number listed in the range. In the present description, any concentration range, percentage range, ratio range, or integer range is to be understood to include the value of any integer within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an integer), unless otherwise indicated. Also, any number range recited herein relating to any physical feature, such as polymer subunits, size or thickness, are to be understood to include any integer within the recited range, unless otherwise indicated. It should be understood that the terms "a" and "an" as used herein refer to "one or more" of the enumerated components. The use of the alternative (e.g., "or") should be understood to mean either one, both, or any combination thereof of the alternatives. As used herein, the terms "include" and "comprise" are used synonymously. The phrase “at least one of” when followed by a list of items or elements refers to an open ended set of one or more of the elements in the list, which may, but does not necessarily, include more than one of the elements. As used herein, a “TGFβR1 inhibitor” refers to a compound that reduces, minimizes, or inactivates activin receptor-linke kinase 5 (ALK5) activity (e.g., directly inhibiting serine/threonine kinase activity or indirectly inhibiting downstream TGFβ-dependent signaling activity, such as SBE-mediated responsiveness to TGFβ and SMAD proteins) by about 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% as compared to untreated ALK5. The terms ALK5 and TGFβR1 can be used interchangeably. As used herein, a “conjugate” refers to a GalNAc moiety covalently attached to at least one TGFβR1 inhibitor via a linker. As used herein, a “liver cell” refers to any cell type associated with normal liver tissue. For example, a liver cell can be a canalicular cell, a Kupffer cell, a hepatocyte, sinusoidal endothelial cell, or a stellate cell. As used herein, an “immune cell” refers to a T cell, B cell, NK cell, NKT cell, or an antigen presenting cell. In some embodiments, an immune cell is a T cell, B cell, NK cell, or
NKT cell. In some embodiments, an immune cell is an antigen presenting cell. In some embodiments, an immune cell is not an antigen presenting cell. The term “salt” or “pharmaceutically acceptable salt” refers to salts derived from a variety of organic and inorganic counter ions well known in the art. Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids. Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like. Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases. Inorganic bases from which salts can be derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, specifically such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine. In some embodiments, the pharmaceutically acceptable base addition salt is chosen from ammonium, potassium, sodium, calcium, and magnesium salts. The term “Cx-y” when used in conjunction with a chemical moiety, such as alkyl, alkenyl, or alkynyl is meant to include groups that contain from x to y carbons in the chain. For example, the term “C1-6alkyl” refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups that contain from 1 to 6 carbons. The term −Cx-yalkylene− refers to a substituted or unsubstituted alkylene chain with from x to y carbons in the alkylene chain. For example −C1-6alkylene− may be selected from methylene, ethylene, propylene, butylene, pentylene, and hexylene, any one of which is optionally substituted. The terms “Cx-yalkenyl” and “Cx-yalkynyl” refer to substituted or unsubstituted unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond, respectively. The term
−Cx-yalkenylene− refers to a substituted or unsubstituted alkenylene chain with from x to y carbons in the alkenylene chain. For example, −C2-6alkenylene− may be selected from ethenylene, propenylene, butenylene, pentenylene, and hexenylene, any one of which is optionally substituted. An alkenylene chain may have one double bond or more than one double bond in the alkenylene chain. The term −Cx-yalkynylene− refers to a substituted or unsubstituted alkynylene chain with from x to y carbons in the alkenylene chain. For example, −C2-6alkenylene− may be selected from ethynylene, propynylene, butynylene, pentynylene, and hexynylene, any one of which is optionally substituted. An alkynylene chain may have one triple bond or more than one triple bond in the alkynylene chain. “Alkyl” refers to a monovalent hydrocarbon consisting solely of carbon and hydrogen, containing no unsaturation, and preferably having from one to twelve carbon atoms, for example, methyl, ethyl, propyl, isopropyl, butyl, and the like. In other embodiments, an alkyl comprises one to five carbon atoms (i.e., C1-C5 alkyl). In other embodiments, an alkyl comprises one to four carbon atoms (i.e., C1-C4 alkyl). In other embodiments, an alkyl comprises one to three carbon atoms (i.e., C1-C3 alkyl). In other embodiments, an alkyl comprises one to two carbon atoms (i.e., C1-C2 alkyl). In other embodiments, an alkyl comprises one carbon atom (i.e., C1 alkyl or methyl). In other embodiments, an alkyl comprises five to eight carbon atoms (i.e., C5- C8 alkyl). In other embodiments, an alkyl comprises two to five carbon atoms (i.e., C2-C5 alkyl). In other embodiments, an alkyl comprises three to five carbon atoms (i.e., C3-C5 alkyl). Unless stated otherwise specifically in the specification, an alkyl chain is optionally substituted by one or more substituents such as those substituents described herein. “Alkylene” refers to a divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing no unsaturation, and preferably having from one to twelve carbon atoms, for example, methylene, ethylene, propylene, butylene, and the like. The alkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. The points of attachment of the alkylene chain to the rest of the molecule and to the radical group are through the terminal carbons respectively. In other embodiments, an alkylene comprises one to five carbon atoms (i.e., C1-C5 alkylene). In other embodiments, an alkylene comprises one to four carbon atoms (i.e., C1-C4 alkylene). In other embodiments, an alkylene comprises one to three carbon atoms
(i.e., C1-C3 alkylene). In other embodiments, an alkylene comprises one to two carbon atoms (i.e., C1-C2 alkylene). In other embodiments, an alkylene comprises one carbon atom (i.e., C1 alkylene). In other embodiments, an alkylene comprises five to eight carbon atoms (i.e., C5-C8 alkylene). In other embodiments, an alkylene comprises two to five carbon atoms (i.e., C2-C5 alkylene). In other embodiments, an alkylene comprises three to five carbon atoms (i.e., C3-C5 alkylene). Unless stated otherwise specifically in the specification, an alkylene chain is optionally substituted by one or more substituents such as those substituents described herein. “Alkenyl” refers to a monovalent hydrocarbon chain consisting solely of carbon and hydrogen, containing at least one carbon-carbon double bond, and preferably having from two to twelve carbon atoms. The alkenyl chain is attached to the rest of the molecule through a single bond. In other embodiments, an alkenyl comprises two to five carbon atoms (i.e., C2-C5 alkenyl). In other embodiments, an alkenyl comprises two to four carbon atoms (i.e., C2-C4 alkenyl). In other embodiments, an alkenyl comprises two to three carbon atoms (i.e., C2-C3 alkenyl). In other embodiments, an alkenyl comprises two carbon atom (i.e., C2 alkenyl). In other embodiments, an alkenyl comprises five to eight carbon atoms (i.e., C5-C8 alkenyl). In other embodiments, an alkenyl comprises three to five carbon atoms (i.e., C3-C5 alkenyl). Unless stated otherwise specifically in the specification, an alkenyl chain is optionally substituted by one or more substituents such as those substituents described herein. “Alkenylene” refers to a divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one carbon-carbon double bond, and preferably having from two to twelve carbon atoms. The alkenylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. The points of attachment of the alkenylene chain to the rest of the molecule and to the radical group are through the terminal carbons respectively. In other embodiments, an alkenylene comprises two to five carbon atoms (i.e., C2-C5 alkenylene). In other embodiments, an alkenylene comprises two to four carbon atoms (i.e., C2-C4 alkenylene). In other embodiments, an alkenylene comprises two to three carbon atoms (i.e., C2-C3 alkenylene). In other embodiments, an alkenylene comprises two carbon atom (i.e., C2 alkenylene). In other embodiments, an alkenylene comprises five to eight carbon atoms (i.e., C5-C8 alkenylene). In other embodiments, an alkenylene comprises three to five carbon atoms (i.e., C3-C5 alkenylene).
Unless stated otherwise specifically in the specification, an alkenylene chain is optionally substituted by one or more substituents such as those substituents described herein. “Alkynyl” refers to a monovalent hydrocarbon chain consisting solely of carbon and hydrogen, containing at least one carbon-carbon triple bond, and preferably having from two to twelve carbon atoms. The alkynyl chain is attached to the rest of the molecule through a single bond. In other embodiments, an alkynyl comprises two to five carbon atoms (i.e., C2-C5 alkynyl). In other embodiments, an alkynyl comprises two to four carbon atoms (i.e., C2-C4 alkynyl). In other embodiments, an alkynyl comprises two to three carbon atoms (i.e., C2-C3 alkynyl). In other embodiments, an alkynyl comprises two carbon atom (i.e., C2 alkynyl). In other embodiments, an alkynyl comprises five to eight carbon atoms (i.e., C5-C8 alkynyl). In other embodiments, an alkynyl comprises three to five carbon atoms (i.e., C3-C5 alkynyl). Unless stated otherwise specifically in the specification, an alkynyl chain is optionally substituted by one or more substituents such as those substituents described herein. “Alkynylene” refers to a divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one carbon-carbon triple bond, and preferably having from two to twelve carbon atoms. The alkynylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. The points of attachment of the alkynylene chain to the rest of the molecule and to the radical group are through the terminal carbons respectively. In other embodiments, an alkynylene comprises two to five carbon atoms (i.e., C2-C5 alkynylene). In other embodiments, an alkynylene comprises two to four carbon atoms (i.e., C2-C4 alkynylene). In other embodiments, an alkynylene comprises two to three carbon atoms (i.e., C2-C3 alkynylene). In other embodiments, an alkynylene comprises two carbon atom (i.e., C2 alkynylene). In other embodiments, an alkynylene comprises five to eight carbon atoms (i.e., C5-C8 alkynylene). In other embodiments, an alkynylene comprises three to five carbon atoms (i.e., C3-C5 alkynylene). Unless stated otherwise specifically in the specification, an alkynylene chain is optionally substituted by one or more substituents such as those substituents described herein. “Heteroalkyl” refers to a monovalent hydrocarbon chain including at least one heteroatom in the chain, containing no unsaturation, and preferably having from one to twelve carbon atoms and from one to 6 heteroatoms, e.g., −O−, −NH−, −S−. The heteroalkyl is attached
to the rest of the molecule through a single bond. In other embodiments, a heteroalkyl comprises one to five carbon atoms and from one to three heteroatoms. In other embodiments, a heteroalkyl comprises one to four carbon atoms and from one to three heteroatoms. In other embodiments, a heteroalkyl comprises one to three carbon atoms and from one to two heteroatoms. In other embodiments, a heteroalkyl comprises one to two carbon atoms and from one to two heteroatoms. In other embodiments, a heteroalkyl comprises one carbon atom and from one to two heteroatoms. In other embodiments, a heteroalkyl comprises five to eight carbon atoms and from one to four heteroatoms. In other embodiments, a heteroalkyl comprises two to five carbon atoms and from one to three heteroatoms. In other embodiments, a heteroalkyl comprises three to five carbon atoms and from one to three heteroatoms. Unless stated otherwise specifically in the specification, a heteroalkyl chain is optionally substituted by one or more substituents such as those substituents described herein. “Heteroalkylene” refers to a divalent hydrocarbon chain including at least one heteroatom in the chain, containing no unsaturation, and preferably having from one to twelve carbon atoms and from one to 6 heteroatoms, e.g., −O−, −NH−, −S−. The heteroalkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. The points of attachment of the heteroalkylene chain to the rest of the molecule and to the radical group are through the terminal atoms of the chain. In other embodiments, a heteroalkylene comprises one to five carbon atoms and from one to three heteroatoms. In other embodiments, a heteroalkylene comprises one to four carbon atoms and from one to three heteroatoms. In other embodiments, a heteroalkylene comprises one to three carbon atoms and from one to two heteroatoms. In other embodiments, a heteroalkylene comprises one to two carbon atoms and from one to two heteroatoms. In other embodiments, a heteroalkylene comprises one carbon atom and from one to two heteroatoms. In other embodiments, a heteroalkylene comprises five to eight carbon atoms and from one to four heteroatoms. In other embodiments, a heteroalkylene comprises two to five carbon atoms and from one to three heteroatoms. In other embodiments, a heteroalkylene comprises three to five carbon atoms and from one to three heteroatoms. Unless stated otherwise specifically in the specification, a heteroalkylene chain is optionally substituted by one or more substituents such as those substituents described herein.
The term “carbocycle” or “carbocyclyl” as used herein refers to a saturated, unsaturated or aromatic ring in which each atom of the ring is carbon. Carbocycle includes 3- to 10- membered monocyclic rings, 6- to 12-membered bicyclic rings, and 6- to 12-membered bridged rings. Each ring of a bicyclic carbocycle may be selected from saturated, unsaturated, and aromatic rings. In an exemplary embodiment, an aromatic ring, e.g., phenyl, may be fused to a saturated or unsaturated ring, e.g., cyclohexane, cyclopentane, or cyclohexene. A bicyclic carbocycle includes any combination of saturated, unsaturated and aromatic bicyclic rings, as valence permits. A bicyclic carbocycle includes any combination of ring sizes such as 4-5 fused ring systems, 5-5 fused ring systems, 5-6 fused ring systems, 6-6 fused ring systems, 5-7 fused ring systems, 6-7 fused ring systems, 5-8 fused ring systems, and 6-8 fused ring systems. Exemplary carbocycles include cyclopentyl, cyclohexyl, cyclohexenyl, adamantyl, phenyl, indanyl, and naphthyl. The term “unsaturated carbocycle” refers to carbocycles with at least one degree of unsaturation and excluding aromatic carbocycles. Examples of unsaturated carbocycles include cyclohexadiene, cyclohexene, and cyclopentene. The term “aryl” refers to an aromatic monocyclic or aromatic multicyclic hydrocarbon ring system. The aromatic monocyclic or aromatic multicyclic hydrocarbon ring system contains only hydrogen and carbon and from five to eighteen carbon atoms, where at least one of the rings in the ring system is aromatic, i.e., it contains a cyclic, delocalized (4n+2) π-electron system in accordance with the Hückel theory. The ring system from which aryl groups are derived include, but are not limited to, groups such as benzene, fluorene, indane, indene, tetralin and naphthalene. The term “cycloalkyl” refers to a saturated ring in which each atom of the ring is carbon. Cycloalkyl may include monocyclic and polycyclic rings such as 3- to 10-membered monocyclic rings, 6- to 12-membered bicyclic rings, and 6- to 12-membered bridged rings. In certain embodiments, a cycloalkyl comprises three to ten carbon atoms. In other embodiments, a cycloalkyl comprises five to seven carbon atoms. The cycloalkyl may be attached to the rest of the molecule by a single bond. Examples of monocyclic cycloalkyls include, e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Polycyclic cycloalkyl radicals include, for example, adamantyl, norbornyl (i.e., bicyclo[2.2.1]heptanyl), decalinyl, 7,7 dimethyl bicyclo[2.2.1]heptanyl, and the like.
The term “halo” or, alternatively, “halogen” or “halide,” means fluoro, chloro, bromo or iodo. In some embodiments, halo is fluoro, chloro, or bromo. The term “haloalkyl” refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, for example, trifluoromethyl, dichloromethyl, bromomethyl, 2,2,2-trifluoroethyl, 1-chloromethyl-2-fluoroethyl, and the like. The term “heterocycle” or “heterocyclyl” as used herein refers to a saturated, unsaturated or aromatic ring comprising one or more heteroatoms. Exemplary heteroatoms include N, O, Si, P, B, and S atoms. Heterocycles include 3- to 10-membered monocyclic rings, 6- to 12- membered bicyclic rings, and 6- to 12-membered bridged rings. A bicyclic heterocycle includes any combination of saturated, unsaturated and aromatic bicyclic rings, as valence permits. In an exemplary embodiment, an aromatic ring, e.g., pyridyl, may be fused to a saturated or unsaturated ring, e.g., cyclohexane, cyclopentane, morpholine, piperidine or cyclohexene. A bicyclic heterocycle includes any combination of ring sizes such as 4-5 fused ring systems, 5-5 fused ring systems, 5-6 fused ring systems, 6-6 fused ring systems, 5-7 fused ring systems, 6-7 fused ring systems, 5-8 fused ring systems, and 6-8 fused ring systems. The term “unsaturated heterocycle” refers to heterocycles with at least one degree of unsaturation and excluding aromatic heterocycles. Examples of unsaturated heterocycles include dihydropyrrole, dihydrofuran, oxazoline, pyrazoline, and dihydropyridine. The term “heteroaryl” includes aromatic single ring structures, preferably 5- to 7- membered rings, more preferably 5- to 6-membered rings, whose ring structures include at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms. The term “heteroaryl” also includes polycyclic ring systems having two or more rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heteroaromatic, e.g., the other rings can be aromatic or non-aromatic carbocyclic, or heterocyclic. Heteroaryl groups include, for example, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, pyrazole, pyridine, pyrazine, pyridazine, and pyrimidine, and the like. The term “heterocycloalkyl” refers to a saturated ring with carbon atoms and at least one heteroatom. Exemplary heteroatoms include N, O, Si, P, B, and S atoms. Heterocycloalkyl may include monocyclic and polycyclic rings such as 3- to 10-membered monocyclic rings, 6- to 12- membered bicyclic rings, and 6- to 12-membered bridged rings. The heteroatoms in the
heterocycloalkyl radical are optionally oxidized. One or more nitrogen atoms, if present, are optionally quaternized. The heterocycloalkyl is attached to the rest of the molecule through any atom of the heterocycloalkyl, valence permitting, such as any carbon or nitrogen atoms of the heterocycloalkyl. Examples of heterocycloalkyl radicals include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl. The term “substituted” refers to moieties having substituents replacing a hydrogen on one or more carbons or substitutable heteroatoms, e.g., −NH−, of the structure. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, i.e., a compound which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. In certain embodiments, substituted refers to moieties having substituents replacing two hydrogen atoms on the same carbon atom, such as substituting the two hydrogen atoms on a single carbon with an oxo, imino or thioxo group. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this disclosure, the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. In some embodiments, substituents may include any substituents described herein, for example: halogen, hydroxy, oxo (=O), thioxo (=S), cyano (−CN), nitro (−NO2), imino (=N−H), oximo (=N−OH), hydrazino (=N−NH2), −Rb−ORa, −Rb−OC(O)−Ra, −Rb−OC(O)−ORa, −Rb−OC(O)−N(Ra)2, −Rb−N(Ra)2, −Rb−C(O)Ra, −Rb−C(O)ORa, −Rb−C(O)N(Ra)2,
−Rb−O−Rc−C(O)N(Ra)2, −Rb−N(Ra)C(O)ORa, −Rb−N(Ra)C(O)Ra, −Rb−N(Ra)S(O)tRa (where t is 1 or 2), −Rb−S(O)tRa (where t is 1 or 2), −Rb−S(O)tORa (where t is 1 or 2), and −Rb−S(O)tN(Ra)2 (where t is 1 or 2); and alkyl, alkenyl, alkynyl, aryl, aralkyl, aralkenyl, aralkynyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl, and heteroarylalkyl any of which may be optionally substituted by alkyl, alkenyl, alkynyl, halogen, haloalkyl, haloalkenyl, haloalkynyl, oxo (=O), thioxo (=S), cyano (−CN), nitro (−NO2), imino (=N−H), oximo (=N−OH), hydrazine (=N−NH2), −Rb−ORa, −Rb−OC(O)−Ra, −Rb−OC(O)−ORa, −Rb−OC(O)−N(Ra)2, −Rb−N(Ra)2, −Rb−C(O)Ra, −Rb−C(O)ORa, −Rb−C(O)N(Ra)2, −Rb−O−Rc−C(O)N(Ra)2, −Rb−N(Ra)C(O)ORa, −Rb−N(Ra)C(O)Ra, −Rb−N(Ra)S(O)tRa (where t is 1 or 2), −Rb−S(O)tRa (where t is 1 or 2), −Rb−S(O)tORa (where t is 1 or 2) and −Rb−S(O)tN(Ra)2 (where t is 1 or 2); wherein each Ra is independently selected from hydrogen, alkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl, or heteroarylalkyl, wherein each Ra, valence permitting, may be optionally substituted with alkyl, alkenyl, alkynyl, halogen, haloalkyl, haloalkenyl, haloalkynyl, oxo (=O), thioxo (=S), cyano (−CN), nitro (−NO2), imino (=N−H), oximo (=N−OH), hydrazine (=N−NH2), −Rb−ORa, −Rb−OC(O)−Ra, −Rb−OC(O)−ORa, −Rb−OC(O)−N(Ra)2, −Rb−N(Ra)2, −Rb−C(O)Ra, −Rb−C(O)ORa, −Rb−C(O)N(Ra)2, −Rb−O−Rc−C(O)N(Ra)2, −Rb−N(Ra)C(O)ORa, −Rb−N(Ra)C(O)Ra, −Rb−N(Ra)S(O)tRa (where t is 1 or 2), −Rb−S(O)tRa (where t is 1 or 2), −Rb−S(O)tORa (where t is 1 or 2) and −Rb−S(O)tN(Ra)2 (where t is 1 or 2); and wherein each Rb is independently selected from a direct bond or a straight or branched alkylene, alkenylene, or alkynylene chain, and each Rc is a straight or branched alkylene, alkenylene or alkynylene chain. “Protecting group” refers to a moiety, except alkyl groups, that when attached to a reactive group in a molecule masks, reduces or prevents that reactivity. Examples of protecting groups can be found in T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3.sup.rd edition, John Wiley & Sons, New York, 1999, and Harrison and Harrison et al., Compendium of Synthetic Organic Methods, Vols. 1-8 (John Wiley and Sons, 1971-1996), which are incorporated herein by reference in their entirety. Representative amino or amine protecting groups include, formyl, acyl groups (such as acetyl, trifluoroacetyl, and benzoyl), benzyl, alkoxycarbonyl (such as benzyloxycarbonyl (CBZ), and tert-butoxycarbonyl (Boc)), trimethyl silyl (TMS), 2-trimethylsilyl-ethanesulfonyl (SES), trityl and substituted trityl groups,
allyloxycarbonyl, 9-fluorenylmethyloxycarbonyl (FMOC), nitro-veratryloxycarbonyl (NVOC), sulfonyl, and the like. Compounds described herein can include protecting groups (e.g., a hydrogen on a reactive nitrogen atom of a compound described herein can be replaced by an amino protecting group). It will be understood by those skilled in the art that substituents can themselves be substituted, if appropriate. Unless specifically stated as “unsubstituted,” references to chemical moieties herein are understood to include substituted variants. For example, reference to a “heteroaryl” group or moiety implicitly includes both substituted and unsubstituted variants. Chemical entities having carbon-carbon double bonds or carbon-nitrogen double bonds may exist in Z- or E- form (or cis- or trans- form). Furthermore, some chemical entities may exist in various tautomeric forms. Unless otherwise specified, chemical entities described herein are intended to include all Z-, E- and tautomeric forms as well. A “tautomer” refers to a molecule wherein a proton shift from one atom of a molecule to another atom of the same molecule is possible. T he compounds presented herein, in certain embodiments, exist as tautomers. In circumstances where tautomerization is possible, a chemical equilibrium of the tautomers will exist. The exact ratio of the tautomers depends on several factors, including physical state, temperature, solvent, and pH. Some examples of tautomeric equilibrium include:
.
The compounds described herein may in some cases exist as diastereomers, enantiomers, or other stereoisomeric forms. The compounds presented herein include all diastereomeric, enantiomeric, and epimeric forms as well as the appropriate mixtures thereof. Separation of stereoisomers may be performed by chromatography or by forming diastereomers and separating by recrystallization, or chromatography, or any combination thereof. (Jean Jacques, Andre Collet, Samuel H. Wilen, “Enantiomers, Racemates and Resolutions”, John Wiley And Sons, Inc., 1981, herein incorporated by reference for this disclosure). Stereoisomers may also be obtained by stereoselective synthesis. The compounds disclosed herein, in some embodiments, are used in different enriched isotopic forms, e.g., enriched in the content of 2H, 3H, 14
and/or C. In one particular embodiment, the compound is deuterated in at least one position. Such deuterated forms can be made by the procedure described in U.S. Patent Nos. 5,846,514 and 6,334,997. As described in U.S. Patent Nos. 5,846,514 and 6,334,997, deuteration can improve the metabolic stability and or efficacy, thus increasing the duration of action of drugs. Unless otherwise stated, structures depicted herein are intended to include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures except for the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by 13C- or 14C-enriched carbon are within the scope of the present disclosure. The compounds of the present disclosure optionally contain unnatural proportions of atomic isotopes at one or more atoms that constitute such compounds. For example, the compounds may be labeled with isotopes, such as for example, deuterium (2H), tritium (3H), iodine-125 (125I) or carbon-14 (14C). Isotopic substitution with 2H, 11C, 13C, 14C, 15C, 12N, 13N, 15N, 16N, 16O, 17O, 14F, 15F, 16F, 17F, 18F, 33S, 34S, 35S, 36S, 35Cl, 37Cl, 79Br, 81Br, 125I are all contemplated. All isotopic variations of the compounds of the present invention, whether radioactive or not, are encompassed within the scope of the present invention. In certain embodiments, the compounds disclosed herein have some or all of the 1H atoms replaced with 2H atoms. The methods of synthesis for deuterium-containing compounds are known in the art and include, by way of non-limiting example only, the following synthetic methods.
Deuterium substituted compounds are synthesized using various methods such as described in: Dean, Dennis C.; Editor. Recent Advances in the Synthesis and Applications of Radiolabeled Compounds for Drug Discovery and Development. [In: Curr., Pharm. Des., 2000; 6(10)] 2000, 110 pp; George W.; Varma, Rajender S. The Synthesis of Radiolabeled Compounds via Organometallic Intermediates, Tetrahedron, 1989, 45(21), 6601-21; and Evans, E. Anthony. Synthesis of radiolabeled compounds, J. Radioanal. Chem., 1981, 64(1-2), 9-32. Deuterated starting materials are readily available and are subjected to the synthetic methods described herein to provide for the synthesis of deuterium-containing compounds. Large numbers of deuterium-containing reagents and building blocks are available commercially from chemical vendors, such as Aldrich Chemical Co. The phrases “parenteral administration” and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion. The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. The phrase “pharmaceutically acceptable excipient” or “pharmaceutically acceptable carrier” as used herein means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as
peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen- free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations. Conjugates A conjugate as described herein comprises a GalNAc moiety linked to a TGFβR1 inhibitor via a linker. In one aspect, the present disclosure provides a conjugate of Formula (I):
(I) wherein Region G comprises at least one GalNAc moiety; Region L3 is a connector that connects Region G to Region Inh; Region Inh comprises a TGFβR1 inhibitor; z is 1, 2, 3, 4, or 5; or a pharmaceutically acceptable salt thereof. In some embodiments, Region G is as described herein. In some embodiments, Region L3 is as described herein. In some embodiments, the TGFβR1 inhibitor is as described herein. In some embodiments, z is 1. In some embodiments, z is 2. In some embodiments, z is 3. Synthetic chemistry transformations and methodologies useful in synthesizing the compounds and conjugates described herein are known in the art and include, for example, those described in R. Larock, Comprehensive Organic Transformations (1989); T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 2d. Ed. (1991); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis (1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis (1995).
Region G/GalNAc Moiety A conjugate as described herein comprises Region G, which comprises at least one GalNAc moiety. In some embodiments, Region G comprises a Display Element for display of the one or more GalNAc moieties. In some embodiments, Region G comprises 1, 2, or 3 GalNAc moieties. In some embodiments, Region G comprises two or three GalNAc moieties. In some embodiments, Region G comprises a structure of Formula (V):
wherein n is 1, 2, or 3; SP is a spacer, wherein each SP is independently a heteroalkylene, heteroalkenylene, or heteroalkynylene comprising 5 to 30 components in the longest linear chain, wherein the components are selected from −CH2−, −CH(C1-4alkyl), −C(C1-4alkyl)2, −CH=CH−, −C≡C−, −C(O)−, −O−, −NH−, −N(C1-4alkyl), −S−, −S(O)−, −S(O)2−, and −P(O)(O-)−; and DE is a branched Display Element, wherein the asterisk (*) is the position of connection to the rest of the conjugate. In some embodiments, n is 2 or 3. In some embodiments, n is 2. In some embodiments, n is 3. In some embodiments, when n is 1, the branched Display Element is absent. In some embodiments, one or more SP units comprise an ethylene glycol or poly(ethylene glycol) region. In some embodiments, one or more SP units comprise an amide bond. In some embodiments, one or more SP units comprise an unsubstituted ethylene or propylene unit. In some embodiments, each SP comprises 5 to 20 components in the longest linear chain. In some embodiments, each SP is −(CH2)2−C(O)−NH−(CH2)x−CH2− (where x is 1, 2, 3, 4, 5, or 6, or x is 1, 2, or 3, or x is 1, or x is 3, or x is 4, or x is 5); −CH2−(CH2)a−CH2−NH−C(O)−(CH2)b−, where a is 0, 1, 2, or 3 (or a is 1 or 2, or a is 1); and b is 1, 2, 3, 4, 5, or 6 (or b is 3, 4, or 5, or b is 5); −C(O)−NH−(CH2)(a+2)−NH−C(O)−(CH2)b−, where
a is 0, 1, 2, or 3 (or a is 1) and b is 1, 2, 3, 4, 5, or 6 (or b is 3, 4, or 5, or b is 5); or −C(O)−NH−(CH2)p− where p is 2, 3, 4, 5, or 6 (or p is 2, or p is 4, or p is 6). The Display Element (DE) comprises a central atom unit and two or three Arm Elements (AE) attached to the central atom unit that connect DE to each SP. In some embodiments, the central atom unit is methine (CH). In some embodiments, the central atom unit is carbon (C). In some embodiments, the central atom unit is nitrogen (N). In some embodiments, the central atom unit i
, where the two nitrogen substituents and one carbon substituent are each an SP, and the other carbon substituent connects the central atom unit to the linker. In some embodiments, each AE is independently −(CH2)1-3O−, −(CH2)1-3NR−, −NR−, −(CH2)1-3−, or a bond, wherein each CH2 is optionally substituted with one or more C1-4alkyl or fluoro substituents, and R is H or C1-4alkyl. In some embodiments, DE is one of the following structures:
. Representative structures of Formula (V) and representative Display Elements are described in U.S. Patent Nos. 8,450,467, 9,399,775, and 10,077,433, which are incorporated herein by reference. In some embodiments, the Region G has the structure:
, where x is 1, 2, 3, 4, 5, or 6. In some embodiments, x is 1, 2, or 3. In some embodiments, x is 1. In some embodiments, x is 3. In some embodiments, x is 4. In some embodiments, x is 5. In some embodiments, Region G has the structure:
where a is 0, 1, 2, or 3; and b is 1, 2, 3, 4, 5, or 6. In some embodiments, a is 1 or 2. In some embodiments, a is 1. In some embodiments, b is 3, 4, or 5. In some embodiments, b is 5. In some embodiments, Region G has the structure:
wherein each p is independently 2, 3, 4, 5, or 6. In some embodiments, each p is 2. In some embodiments, each p is 4. In some embodiments, each p is 6. TGFβR1 Inhibitors The conjugates described herein comprise a TGFβR1 inhibitor. The TGFβR1 inhibitor can provide a direct or indirect effect. In some preferred embodiments, the TGFβR1 inhibitor is not an siRNA, an antisense RNA, or other oligonucleotide. In some embodiments, the TGFβR1 inhibitor is a non-naturally occurring small molecule. A “small molecule” is an organic compound with a molecular weight of less than 1500, or 1000, or 900, or 750, or 600, or 500 Daltons. In some embodiments, a small molecule TGFβR1 inhibitor has an octanol-water partition coefficient (logP) in the range of 3 to 6, or from 4 to 5, or from 2 to 4. In some embodiments, a small molecule TGFβR1 inhibitor has a polar surface area of less than 200, or less than 150 Å2. In some embodiments, a small molecule TGFβR1 inhibitor has not more than five, or not more than three, hydrogen bond donors, and not more than 10, or not more than three hydrogen bond acceptors. A small molecule TGFβR1 inhibitor is not a protein, a polysaccharide, or a nucleic acid. TGFβR1 Inhibitors of Formula (A-I) In some aspects, the TGFβR1 inhibitor is a compound of Formula (A-I):
wherein
one of M1 and M2 is and the other of M1 and M2 is selected from:
R1 and R2 are, at each occurrence, independently selected from hydrogen, halogen, −OR11, −SR11, −N(R11)2, −NO2, −CN, phenyl, and −C1-C6 alkyl, wherein said −C1-C6 alkyl is optionally substituted with one or more substituents independently selected from halogen, −OR11, −SR11, −S(O)R10, −S(O)2R11, −S(O)2N(R11)2, −N(R11)2, −C(O)R10, −C(O)N(R11)2, −N(R11)C(O)R10, −C(O)OR11, −OC(O)R10, −NO2, and −CN; R3 is, at each occurrence, independently selected from halogen, −C1-C3 alkyl, −C1-C3 haloalkyl, −OH, -NO2, -CN, -OC1-C3 alkyl, and -OC1-C3 haloalkyl; each R4 is, at each occurrence, independently selected from hydrogen and C1-C3 alkyl or two R4 join together with atoms to which they are attached to form a 5- or 6-membered heterocycle optionally substituted with one or more substituents independently selected from halogen, C1-C3 alkyl, -OH, OC1-C3 alkyl, and -OC1-C3 haloalkyl; R5 is hydrogen, halogen, -OR61, -SR61, -N(R61)2, -NO2, -CN, and -C1-C6 alkyl, wherein said -C1-C6 alkyl is optionally substituted with one or more substituents independently selected from halogen, -OR61, -SR61, -N(R61)2, -NO2, and -CN; R6 is, at each occurrence, independently selected from: halogen, -OR21, -SR21, -N(R21)2, -C(O)R20, -C(O)N(R21)2, -N(R21)C(O)R20 , - C(O)OR21, -OC(O)R21, -S(O)R20, -S(O)2R21, -S(O)2N(R21)2, -OC(O)OR21, -OC(O)N(R21)2, −NR21C(=O)OR21, -N(R21)C(O)N(R21)2, -NO2, and -CN;
C1-C10 alkyl, C2-C10 alkenyl, and C2-C10 alkynyl, each of which is optionally substituted with one or more substituents independently selected from halogen, -OR21, -SR21, −N(R21)2, -C(O)R20, -C(O)N(R21)2, -N(R21)C(O)R20 , -C(O)OR21, -OC(O)R21, -S(O)R20, - S(O)2R21, -S(O)2N(R21)2, -OC(O)OR21, -OC(O)N(R21)2, -NR21C(=O)OR21, -N(R21)C(O)N(R21)2, -NO2, =O, =S, =N(R21), -CN, a C3-C10 carbocycle, and a 3- to 10-membered heterocycle wherein said C3-C10 carbocycle and said 3- to 10-membered heterocycle are optionally substituted with one or more substituents independently selected from RX; and a C3-C10 carbocycle and a 3- to 10-membered heterocycle, each of which is optionally substituted with one or more substituents independently selected from halogen, - OR20, -OH, -SR20, -SH, -N(R21)2, -C(O)R20, -C(O)N(R21)2, -N(R21)C(O)R20 , -C(O)OR21, −OC(O)R21, -S(O)R20, -S(O)2R21, -S(O)2N(R21)2, -OC(O)OR21, -OC(O)N(R21)2, −NR21C(=O)OR21, -N(R21)C(O)N(R21)2, -NO2, =O, =S, =N(R21), -CN, -C2-C6 alkenyl, −C2-C6 alkynyl and C1-C6 alkyl, wherein said C1-C6 alkyl is optionally substituted with one or more substituents independently selected from RY; R7 and R8 are independently selected from hydrogen, halogen, C1-C3 alkyl, -OH, OC1-C3 alkyl, and -OC1-C3 haloalkyl, or R7 and R8 join together with the atoms to which they are attached to form a C5-C6 carbocycle or 5- or 6- membered heterocycle each of which is optionally substituted with one or more substituents independently selected from halogen, -OR31, -SR31, -N(R31)2, -NO2, -CN and -C1-C6 alkyl wherein said C1-C6 alkyl is optionally substituted with one or more substituents independently selected from halogen, -OR31, -SR31, -N(R31)2, - NO2, and −CN; Y is selected from −O- and -N(R9)- and R9 is, at each occurrence, independently selected from: hydrogen; and -C1-C6 alkyl optionally substituted with one or more substituents independently selected from halogen, -OR41, -SR41, -S(O)R40, -S(O)2R41, -S(O)2N(R41)2, −N(R41)2, -C(O)R40, -C(O)N(R41)2, -N(R41)C(O)R40 , -C(O)OR41, -OC(O)R40, -NO2, and -CN; each R10, R20, and R40 is independently selected at each occurrence from: -C1-C10 alkyl, -C2-C10 alkenyl, and -C2-C10 alkynyl, each of which is optionally substituted with one or more substituents independently selected from RY; and a C3-C12 carbocycle and a 3- to 12-membered heterocycle, each of which is optionally substituted with one or more substituents independently selected from RX;
each R11, R21, R31, R41, and R61 is independently selected at each occurrence from: hydrogen; -C1-C10 alkyl, -C2-C10 alkenyl, and -C2-C10 alkynyl, each of which is optionally substituted with one or more substituents independently selected from RY; and a C3-C12 carbocycle and a 3- to 12-membered heterocycle, each of which is optionally substituted with one or more substituents independently selected from RX, or two R11, R21, R31, R41, or R61 on the same N atom are taken together with the N atom to which they are attached to form a N-containing heterocycle optionally substituted with RX; each RX is independently selected at each occurrence from: halogen, -OR51, -SR51, −N(R51)2, -C(O)R50, -C(O)N(R51)2, -N(R51)C(O)R50 , -C(O)OR51, -OC(O)R51, -S(O)R50, −S(O)2R51, -S(O)2N(R51)2, -OC(O)OR51, -OC(O)N(R51)2, -NR51C(=O)OR51, −N(R51)C(O)N(R51)2, -NO2, =O, =S, =N(R51), -CN, -C2-C6 alkenyl, -C2-C6 alkynyl, and C1-C6 alkyl, wherein said C1-C6 alkyl is optionally substituted with one or more substituents independently selected from -OR51, -SR51, -N(R51)2, -C(O)R50, -C(O)N(R51)2, -N(R51)C(O)R50 , −C(O)OR51, -OC(O)R51, -S(O)R50, -S(O)2R51, -S(O)2N(R51)2, -OC(O)OR51, -OC(O)N(R51)2, −NR51C(=O)OR51, -N(R51)C(O)N(R51)2, and =O; each RY is independently selected at each occurrence from: halogen, -OR51, -SR51, −N(R51)2, -C(O)R50, -C(O)N(R51)2, -N(R51)C(O)R50, -C(O)OR51, -OC(O)R51, -S(O)R50, −S(O)2R51, -S(O)2N(R51)2, -OC(O)OR51, -OC(O)N(R51)2, -NR51C(=O)OR51, −N(R51)C(O)N(R51)2, -NO2, =O, =S, =N(R51), and -CN; each R50 is independently selected at each occurrence from: -C1-C10 alkyl, -C2-C10 alkenyl, and -C2-C10 alkynyl, each of which is optionally substituted with one or more substituents independently selected from halogen, -OH, -CN, -NO2, −NH2, =O, =S, -O-C1-C10 alkyl, C3-C12 carbocycle, and a 3- to 12-membered heterocycle; and a C3-C12 carbocycle and a 3- to 12-membered heterocycle, each of which is optionally substituted with one or more substituents independently selected from halogen, -OH, - CN, -NO2, -NH2, =O, =S, -C1-C10 alkyl, -O-C1-C10 alkyl, and -C1-C10 haloalkyl; each R51 is independently selected at each occurrence from: hydrogen;
-C1-C10 alkyl, -C2-C10 alkenyl, and -C2-C10 alkynyl, each of which is optionally substituted with one or more substituents independently selected from halogen, -OH, -CN, -NO2, −NH2, =O, =S, -O-C1-C10 alkyl, C3-C12 carbocycle, and a 3- to 12-membered heterocycle; and a C3-C12 carbocycle and a 3- to 12-membered heterocycle, each of which is optionally substituted with one or more substituents independently selected from halogen, -OH, −CN, -NO2, -NH2, =O, =S, -C1-C10 alkyl, -O-C1-C10 alkyl, and -C1-C10 haloalkyl; Z1, Z2, Z3, and Z4 are each independently selected from N or C(H); n is selected from 1, 2, and 3; m is 0, 1, or 2; s is selected from 0 and 1; and w is selected from 0, 1, 2, 3, 4, and 5. or a pharmaceutically acceptable salt thereof. In a second aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein one of M1 and M2 is
and the other of M1 and M2 is
the remaining variables (e.g., R1-R8, R10, R20, R40, R11, R21, R31, R41, R50, R51, R61, Y, RX, RY, Z1, Z2, Z3, Z4, n, m, s, and w) are as set forth in the first aspect. In a third aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein one of M1
and the other of M1 and M2 is
the remaining variables are as set forth in the first aspect.
In a fourth aspect, disclosed herein is a compound represented by Formula (A-I) wherein one of one of M1 and M2 is and the other of M1 and M2 is ; and the remaining variables are as set forth in the first aspect. In a fifth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein one of one of M1 and M2 is and the other of M1 and M2 is ; and the remaining variables are as set forth in the first aspect. In a sixth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1 and M2 is as set forth in the fifth aspect and R7 and R8 are independently selected from hydrogen, halogen, C1-C3 alkyl, -OH, OC1-C3 alkyl, and -OC1-C3 haloalkyl, or R7 and R8 join together with the atoms to which they are attached to form a C5-C6 carbocycle or 5- or 6- membered heterocycle each of which is optionally substituted with one or more substituents independently selected from halogen, -OR31, -SR31, -N(R31)2, and -C1-C6 alkyl wherein said C1-C6 alkyl is optionally substituted with one or more substituents independently selected from halogen, -OR31, -SR31, and -N(R31)2; and the remaining variables are as set forth in the first aspect. In a seventh aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1 and M2 is as set forth in the fifth aspect and R7 and R8 are independently selected from hydrogen, halogen, C1-C3 alkyl, -OH, OC1-C3 alkyl, and -OC1-C3 haloalkyl, or R7 and R8 join together with the atoms to which they are attached to form an unsubstituted C5-C6 carbocycle or an unsubstituted 5- or 6- membered heterocycle; and the remaining variables are as set forth in the first aspect. In an eighth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1 and M2 is as set forth in any one of aspects 5-7 wherein the 5- or 6-
membered heterocycle of R7 and R8 is a 5- or 6- membered heterocycle contains one ring heteroatom selected from nitrogen contains one ring heteroatom selected from nitrogen; and the remaining variables are as set forth in the first aspect. In a ninth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1 and M2 is as set forth in any one of aspects 5-7 wherein R7 and R8 join together with the atoms to which they are attached to form a phenyl ring optionally substituted with one or more substituents independently selected from halogen, -OR31, -SR31, -N(R31)2, and - C1-C6 alkyl wherein said C1-C6 alkyl is optionally substituted with one or more substituents independently selected from halogen, -OR31, -SR31, and -N(R31)2; and the remaining variables are as set forth in the first aspect. In a tenth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1 and M2 is as set forth in any one of aspects 5-7 wherein R7 and R8 join together with the atoms to which they are attached to form an unsubstituted phenyl ring; and the remaining variables are as set forth in the first aspect. In an eleventh aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1 and M2 is as set forth in any one of aspects 5-7 wherein R7 and R8 are each hydrogen; and the remaining variables are as set forth in the first aspect. In a twelfth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7 and R8 are as set forth in any one of aspects 1-11 and wherein m is 1 or 2 and R3 is, at each occurrence, independently selected from halogen, -C1-C3 alkyl, -C1-C3 haloalkyl, -OH, OC1-C3 alkyl, and -OC1-C3 haloalkyl; and the remaining variables are as set forth in the first aspect. In a thirteenth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7 and R8 are as set forth in any one of aspects 1-11 and wherein m is 1 and R3 is, at each occurrence, independently selected from halogen, -C1-C3 alkyl, -C1-C3 haloalkyl, -OH, and -OC1-C3 alkyl; and the remaining variables are as set forth in the first aspect. In a fourteenth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7 and R8 are as set forth in any one of aspects 1-11 and wherein m is zero; and the remaining variables are as set forth in the first aspect.
In a fifteenth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein one of one of M1 and M2 is and the other of M1 and M2 is ; and the remaining variables are as set forth in the first aspect. In a sixteenth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1 and M2 is as set forth in aspect 15 wherein Z1, Z2, Z3, and Z4 are -C(H); and the remaining variables are as set forth in the first aspect. In a seventeenth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1 and M2 is as set forth in aspect 15 wherein Z2 is N and Z1, Z3, and Z4 are -C(H); and the remaining variables are as set forth in the first aspect. In a eighteenth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1 and M2 are as set forth in aspect 15 wherein Z1 is N and Z2, Z3, and Z4 are -C(H); and the remaining variables are as set forth in the first aspect. In a nineteenth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M2, R7, R8, m, R3, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1- 18 wherein M1 is ; and the remaining variables are as set forth in the first aspect. In a twentieth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, R7, R8, m, R3, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1- 18 wherein M2 is ; and the remaining variables are as set forth in the first aspect. In a twenty-first aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, R3, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-20 wherein R5 is hydrogen, halogen, or C1-C3 alkyl optionally substituted with halogen; and the remaining variables are as set forth in the first aspect.
In a twenty-second aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, R3, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-20 wherein R5 is hydrogen or C1-C3 alkyl; and the remaining variables are as set forth in the first aspect. In a twenty-third aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, R3, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-20 and wherein R5 is methyl; and the remaining variables are as set forth in the first aspect. In a twenty-fourth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein one of M1 and M
and the other of M1 and M2 is selected from:
and the remaining variables are as set forth in the first aspect. In a twenty-fifth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1 is as set forth in aspect 24 and wherein M2 is
; and the remaining variables are as set forth in the first aspect. In a twenty-sixth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, R3, R5, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-25 and wherein R1 and R2 are, at each occurrence, independently selected from hydrogen, halogen, -OR11, -SR11, -N(R11)2, phenyl, and -C1-C6 alkyl wherein said C1-C6 alkyl is optionally substituted with one or more substituents independently selected from halogen, −OR11, -SR11, -S(O)R10, -S(O)2R11, -S(O)2N(R11)2, -N(R11)2, -C(O)R10, -C(O)N(R11)2,
−N(R11)C(O)R10, -C(O)OR11, and -OC(O)R10; and the remaining variables are as set forth in the first aspect. In a twenty-seventh aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, R3, R5, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-25 and wherein R1 and R2 are independently selected at each occurrence from hydrogen, phenyl, and -C1-C3 alkyl wherein said -C1-C3 alkyl is optionally substituted with one or more substituents independently selected from halogen, -OR11, and -C(O)OR11; and the remaining variables are as set forth in the first aspect. In a twenty-eighth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, R3, R5, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-25 and wherein R1 and R2 are independently selected at each occurrence from hydrogen, -CH3, -CH2OH, CH2CO2CH3, and phenyl; and the remaining variables are as set forth in the first aspect. In a twenty-ninth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, R3, R5, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-25 and wherein R1 and R2 are each hydrogen; and the remaining variables are as set forth in the first aspect. In a thirtieth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, R1, R2, R3, R5, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-29 and wherein s is zero; and the remaining variables are as set forth in the first aspect. In a thirty-first aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, R1, R2, R3, R5, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-29 and wherein s is one; and the remaining variables are as set forth in the first aspect. In a thirty-second aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, R1, R2, R3, R5, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-29 and wherein s is one and n is two or three; and the remaining variables are as set forth in the first aspect.
In a thirty-third aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, R1, R2, R3, R5, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-29 and wherein s is zero or one and n is one; and the remaining variables are as set forth in the first aspect. In a thirty-fourth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, R1, R2, R3, R5, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-29 and wherein s is zero or one and n is two; and the remaining variables are as set forth in the first aspect. In a thirty-fifth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, R1, R2, R3, R5, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-29 and wherein s is zero or one and n is three; and the remaining variables are as set forth in the first aspect. In a thirty-sixth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, s, n R1, R2, R3, R5, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-35 and wherein Y is selected from −O- and -N(R9)- and R9 is, at each occurrence, independently selected from: hydrogen; and -C1-C6 alkyl optionally substituted with one or more substituents independently selected from halogen, -OR41, -SR41, -S(O)R40, −S(O)2R41, -S(O)2N(R41)2, -N(R41)2, -C(O)R40, -C(O)N(R41)2, -N(R41)C(O)R40, -C(O)OR41, and −OC(O)R40; and the remaining variables are as set forth in the first aspect. In a thirty-seventh aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, s, n R1, R2, R3, R5, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-35 and wherein Y is selected from −O- and -N(R9)- and R9 is, at each occurrence, independently selected from: hydrogen; and unsubstituted-C1-C6 alkyl; and the remaining variables are as set forth in the first aspect. In a thirty-eight aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, s, n R1, R2, R3, R5, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-35 and wherein Y is selected from O, N(H), and N(Me); and unsubstituted −C1-C6 alkyl; and the remaining variables are as set forth in the first aspect. In a thirty-ninth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, s, n R1, R2, R3, R5, R9, Y, Z1, Z2, Z3, and Z4 are as set
forth in any one of aspects 1-38 and wherein w is zero; and the remaining variables are as set forth in the first aspect. In a fortieth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, s, n R1, R2, R3, R5, R9, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-38 and wherein w is 1, 2, 3, 4, or 5; and the remaining variables are as set forth in the first aspect. In a forty-first aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, s, n R1, R2, R3, R5, R9, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-38 and wherein w is 1, 2, or 3; and the remaining variables are as set forth in the first aspect. In a forty-second aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, s, n R1, R2, R3, R5, R9, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-38 and wherein w is 1 or 2; and the remaining variables are as set forth in the first aspect. In a forty-third aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, s, n R1, R2, R3, R5, R9, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-38 having formula (A-IA), (A-IB), (A-IC), (A-ID) or (A-IE):
or a pharmaceutically acceptable salt of any one of formula (A-IA), (A-IB), (A-IC), (A-ID) or (A-IE); and the remaining variables are as set forth in the first aspect. In a forty-fourth aspect, disclosed herein is a compound represented by Formula (A-I) wherein M1, M2, R7, R8, m, s, n R1, R2, R3, R5, R9, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-38 having formula (A-IC) or (A-ID)
or a pharmaceutically acceptable salt of any one of formula (A-IC), or (A-ID); and the remaining variables are as set forth in the first aspect. In a forty-fifth aspect, disclosed herein is a compound represented by Formula (A-I) wherein M1, M2, R7, R8, m, s, n R1, R2, R3, R5, R9, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-38 having formula (A-IF):
wherein w is 1, 2, 3, or 4; or a pharmaceutically acceptable salt thereof); and the remaining variables are as set forth in the first aspect. In a forty-sixth aspect, disclosed herein is a compound represented by Formula (A-IF) or a salt thereof, wherein M1, M2, R7, R8, m, s, n, R1, R2, R3, R5, R9, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-38 and w is 2 or 3; and the remaining variables are as set forth in the first aspect. In a forty-seventh aspect, disclosed herein is a compound represented by Formula (A-IF) or a salt thereof, wherein M1, M2, R7, R8, m, s, n, R1, R2, R3, R5, R9, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-38 and w is 2; and the remaining variables are as set forth in the first aspect.
In a forty-eighth aspect, disclosed herein is a compound represented by Formula (A-I), (A-IA), (A-IB), (A-IC), (A-ID), (A-IE), or (A-IF) or a salt thereof, wherein M1, M2, R7, R8, m, s, n, w, R1, R2, R3, R5, R9, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-47 and R6 is independently selected at each occurrence from: halogen, -OR21, -N(R21)2 and -CN; and C1-C6 alkyl optionally substituted with one or more substituents independently selected from halogen, -OR21, -SR21, -N(R21)2, -C(O)R20, -C(O)N(R21)2, -N(R21)C(O)R20 , - C(O)OR21, −OC(O)R21, -S(O)R20, -S(O)2R21, -S(O)2N(R21)2, -OC(O)OR21, -OC(O)N(R21)2, −NR21C(=O)OR21, -N(R21)C(O)N(R21)2, -NO2, =O, =S, =N(R21), -CN, a C3-C10 carbocycle, and a 3- to 10-membered heterocycle wherein said C3-C10 carbocycle and said 3- to 10-membered heterocycle are optionally substituted with one or more substituents independently selected from RX; and a C3-C10 carbocycle and a 3- to 10-membered heterocycle, each of which is optionally substituted with one or more substituents independently selected from halogen, -OR20, −OH, - SR20, -SH, -N(R21)2, -C(O)R20, -C(O)N(R21)2, -N(R21)C(O)R20 , -C(O)OR21, −OC(O)R21, - S(O)R20, -S(O)2R21, -S(O)2N(R21)2, -OC(O)OR21, -OC(O)N(R21)2, −NR21C(=O)OR21, - N(R21)C(O)N(R21)2, -NO2, =O, =S, =N(R21), -CN, -C2-C6 alkenyl, −C2-C6 alkynyl and C1-C6 alkyl wherein said C1-C6 alkyl is optionally substituted with one or more substituents independently selected from RY; and the remaining variables are as set forth in the first aspect. In a forty-ninth aspect, disclosed herein is a compound represented by Formula (A-I), (A- IA), (A-IB), (A-IC), (A-ID), (A-IE) or (A-IF) or a salt thereof, wherein M1, M2, R7, R8, m, s, n, w, R1, R2, R3, R5, R9, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-47 and R6 is independently selected at each occurrence from: halogen, -OR21, and -N(R21)2; C1-C6 alkyl optionally substituted with one or more substituents independently selected from halogen, -OR21, -SR21, -N(R21)2, -C(O)R20, -C(O)N(R21)2, -N(R21)C(O)R20 , −C(O)OR21, -OC(O)R21, -S(O)R20, -S(O)2R21, -S(O)2N(R21)2, -OC(O)OR21, -OC(O)N(R21)2, −NR21C(=O)OR21, -N(R21)C(O)N(R21)2, -NO2, =O, =S, =N(R21), -CN, a C3-C10 carbocycle, and a 3- to 10-membered heterocycle, wherein said C3-C10 carbocycle and said 3- to 10-membered
heterocycle are optionally substituted with one or more substituents independently selected from RX; and phenyl and a 6-membered heterocycle comprising 1, 2, or 3 ring heteroatoms selected from nitrogen, each of which is optionally substituted with one or more substituents independently selected from halogen, -OR20, -OH, -SR20, -SH, -N(R21)2, -C(O)R20, −C(O)N(R21)2, -N(R21)C(O)R20 , -C(O)OR21, -OC(O)R21, -S(O)R20, -S(O)2R21, -S(O)2N(R21)2, −OC(O)OR21, -OC(O)N(R21)2, -NR21C(=O)OR21, -N(R21)C(O)N(R21)2, -NO2, =O, =S, =N(R21), - CN, -C2-C6 alkenyl, -C2-C6 alkynyl and C1-C6 alkyl wherein said C1-C6 alkyl is optionally substituted with one or more substituents independently selected from RY; and the remaining variables are as set forth in the first aspect. In some such aspects, at least one R6 is phenyl optionally substituted with one or more substituents independently selected from halogen, -OR20, -OH, -SR20, -SH, -N(R21)2, -C(O)R20, −C(O)N(R21)2, -N(R21)C(O)R20 , -C(O)OR21, -OC(O)R21, -S(O)R20, -S(O)2R21, -S(O)2N(R21)2, −OC(O)OR21, -OC(O)N(R21)2, -NR21C(=O)OR21, -N(R21)C(O)N(R21)2, -NO2, =O, =S, =N(R21), −CN, -C2-C6 alkenyl, -C2-C6 alkynyl and C1-C6 alkyl, wherein said C1-C6 alkyl is optionally substituted with one or more substituents independently selected from RY. In a fiftieth aspect, disclosed herein is a compound represented by Formula (A-I), (A-IA), (A-IB), (A-IC), (A-ID), (A-IE) or (A-IF) or a salt thereof, wherein M1, M2, R7, R8, m, s, n, w, R1, R2, R3, R5, R9, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-47 and R6 is independently selected at each occurrence from: halogen, -OR21, and -N(R21)2; C1-C6 alkyl optionally substituted with halogen; and a C3-C10 carbocycle and a 3- to 10-membered heterocycle, each of which is optionally substituted with C1-C6 alkyl, wherein said C1-C6 alkyl is optionally substituted with halogen; and the remaining variables are as set forth in the first aspect. In a fifty-first aspect, disclosed herein is a compound represented by Formula (A-I), (A- IA), (A-IB), (A-IC), (A-ID), (A-IE) or (A-IF) or a salt thereof, wherein M1, M2, R7, R8, m, s, n, w, R1, R2, R3, R5, R9, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-47 and R6 is independently selected at each occurrence from:
halogen, and -OR21; C1-C6 alkyl optionally substituted with halogen; and phenyl and a 6-membered heterocycle comprising 1, 2, or 3 ring heteroatoms selected from nitrogen, each of which is optionally substituted with C1-C6 alkyl wherein said C1- C6 alkyl is optionally substituted with one or more substituents independently selected from RY; and the remaining variables are as set forth in the first aspect. In a fifty-second aspect, disclosed herein is a compound represented by Formula (A-I), (A-IA), (A-IB), (A-IC), (A-ID), (A-IE) or (A-IF) or a salt thereof, wherein M1, M2, R6, R7, R8, m, s, n, w, R1, R2, R3, R5, R9, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-51 and R21 is C1-C3alkyl, phenyl, or a 6-membered heterocycle comprising 1, 2, or 3 ring heteroatoms selected from nitrogen; and the remaining variables are as set forth in the first aspect. In a fifty-third aspect, disclosed herein is a compound represented by Formula (A-I), (A- IA), (A-IB), (A-IC), (A-ID), (A-IE) or (A-IF) or a salt thereof, wherein M1, M2, R6, R7, R8, m, s, n, w, R1, R2, R3, R5, R9, R21, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-52 and RY on the phenyl or heterocycle of R6 is selected from halogen; and the remaining variables are as set forth in the first aspect. In a fifty-fourth aspect, disclosed herein is a compound represented by Formula (A-I), (A-IA), (A-IB), (A-IC), (A-ID), (A-IE) or (A-IF) or a salt thereof, wherein M1, M2, R6, R7, R8, m, s, n, w, R1, R2, R3, R5, R9, R21, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-52 and RY on the phenyl or heterocycle of R6 is selected from fluorine or chlorine; and the remaining variables are as set forth in the first aspect. In a fifty-fifth aspect, disclosed herein is a compound represented by Formula (A-I), (A- IA), (A-IB), (A-IC), (A-ID), (A-IE) or (A-IF) or a salt thereof, wherein M1, M2, R7, R8, m, s, n, w, R1, R2, R3, R5, R9, R21, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-54 and R6 is independently selected at each occurrence from: F, Cl, -OCH3, -CF3, -CN, -CH3, -CH2CH3, −CH(CH3)2, -OCF3, -CH2CF3, -CH(OH)(CF3), N(CH3)2, pyridyl, cyclohexyl, cyclopentyl, -O- phenyl, -O-pyridyl, and phenyl optionally substituted with one or more substituents independently selected from F, and -CH2NH2; and the remaining variables are as set forth in the first aspect. In some such aspects, R6 does not comprise cyano.
In a fifty-sixth aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, s, n, R1, R2, R3, R5, R9, R21, Y, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-55 and
is:
, wherein represents the point of attachment to
and the remaining variables are as set forth in the first aspect. In a fifty-seventh aspect, disclosed herein is a compound represented by Formula (A-I) or a salt thereof, wherein M1, M2, R7, R8, m, R3, R5, R9, R21, Z1, Z2, Z3, and Z4 are as set forth in any one of aspects 1-55 and wherein
is:
, wherein represents the point of attachment to and the remaining variables are as set forth in the first aspect.
In another aspect, the TGFβR1 inhibitor is a compound from Table 14. TGFβR1 Inhibitors of Formula (B-I) In some aspects, the TGFβR1 inhibitor is a compound of Formula (B-I): wherein:
M1 and M2 are independently selected from
R1 and R2 are independently selected at each occurrence from: a halogen, -OR10, -SR10, -N(R10)2, -C(O)R10, -C(O)N(R10)2, -N(R10)C(O)R10 , -C(O)OR10, −OC(O)R10, -S(O)R10, -S(O)2R10, -S(O)2N(R10)2, -P(O)(OR10)2, -OP(O)(OR10)2, -NO2, and -CN; -C1-C10 alkyl, -C2-C10 alkenyl, and -C2-C10 alkynyl, each of which is optionally substituted at each occurrence with one or more substituents independently selected from a halogen, −OR10, -SR10, -N(R10)2, -C(O)R10, -C(O)N(R10)2, -N(R10)C(O)R10 , -C(O)OR10, −OC(O)R10, -S(O)R10, -S(O)2R10, -S(O)2N(R10)2, -P(O)(OR10)2, -OP(O)(OR10)2, -NO2, =O, =S, =N(R10), -CN, a C3-C10 carbocycle, and a 3- to 10-membered heterocycle; and
a C3-C10 carbocycle and a 3- to 10-membered heterocycle, each of which is optionally substituted with one or more substituents independently selected from a halogen, - OR10, -SR10, -N(R10)2, -C(O)R10, -C(O)N(R10)2, -N(R10)C(O)R10 , -C(O)OR10, -OC(O)R10, −S(O)R10, -S(O)2R10, -S(O)2N(R10)2, -P(O)(OR10)2, -OP(O)(OR10)2, -NO2, =O, =S, =N(R10), -CN, -C1-C6 alkyl, -C2-C6 alkenyl, and -C2-C6 alkynyl; R3 is selected from hydrogen and -C1-C10 alkyl optionally substituted with one or more substituents independently selected from a halogen, -NO2, =O, =S, =N(R10), -CN, -OR10, −SR10, -N(R10)2, -C(O)R10, -C(O)N(R10)2, -N(R10)C(O)R10 , -C(O)OR10, and -OC(O)R10; n and m are independently selected from 0, 1, 2, 3, and 4; Q is selected from a bond, -(CR10 2)p-, -(CR10 2)qC(=O)(CR10 2)q-, -(CR10 2)qC(=S)(CR10 2)q-, −(CR10 2)qC(=NR10)(CR10 2)q-, -(CR10 2)qO(CR10 2)q-, -(CR10 2)qS(CR10 2)q-, −(CR10 2)qN(R10)(CR10 2)q-, -(CR10 2)qOC(=O)O(CR10 2)q-, -(CR10 2)qC(=O)N(R10)(CR10 2)q-, −(CR10 2)qN(R10)C(=O)(CR10 2)q-, and -(CR10 2)qN(R10)SO2(CR10 2)q-; p is selected from 1, 2, 3, 4, and 5; q is independently selected at each occurrence from 0, 1, 2, 3, 4, and 5; T is selected from an optionally substituted saturated C3-C7 carbocycle, an optionally substituted C5-12 bicyclic carbocycle, and an optionally substituted 4- to 12-membered heterocycle, wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13; R13 is independently selected at each occurrence from: a halogen, -OR10, -SR10, -N(R10)2, -C(O)R10, -C(O)N(R10)2, -N(R10)C(O)R10 , −C(O)OR10, -OC(O)R10, -S(O)R10, -S(O)2R10, -S(O)2N(R10)2, -P(O)(OR10)2, -OP(O)(OR10)2, −NO2, =O, =S, =N(R10), and -CN; -C1-C10 alkyl, -C2-C10 alkenyl, and -C2-C10 alkynyl, each of which is optionally substituted at each occurrence with one or more substituents independently selected from a halogen, -OR10, -SR10, -N(R10)2, -C(O)R10, -C(O)N(R10)2, -N(R10)C(O)R10 , -C(O)OR10, −OC(O)R10, -S(O)R10, -S(O)2R10, -S(O)2N(R10)2, -P(O)(OR10)2, -OP(O)(OR10)2, -NO2, =O, =S, =N(R10), -CN, a C3-C10 carbocycle, and a 3- to 10-membered heterocycle; and a C3-C10 carbocycle and a 3- to 10-membered heterocycle, each of which is optionally substituted with one or more substituents independently selected from a halogen,
−OR10, -SR10, -N(R10)2, -C(O)R10, -C(O)N(R10)2, -N(R10)C(O)R10 , -C(O)OR10, -OC(O)R10, −S(O)R10, -S(O)2R10, -S(O)2N(R10)2, -P(O)(OR10)2, -OP(O)(OR10)2, -NO2, =O, =S, =N(R10), -CN, -C1-C6 alkyl, -C2-C6 alkenyl, and -C2-C6 alkynyl; and R10 is independently selected at each occurrence from: hydrogen; -C1-C10 alkyl, -C2-C10 alkenyl, and -C2-C10 alkynyl, each of which is optionally substituted at each occurrence with one or more substituents independently selected from a halogen, -OH, -CN, -NO2, -NH2, =O, =S, -O-C1-C10 alkyl, C3-C12 carbocycle, and a 3- to 12- membered heterocycle; and a C3-C12 carbocycle and a 3- to 12-membered heterocycle, each of which is optionally substituted at each occurrence with one or more substituents independently selected from a halogen, -OH, -CN, -NO2, -NH2, =O, =S, -C1-C10 alkyl, -O-C1-C10 alkyl, and -C1-C10 haloalkyl; or a pharmaceutically acceptable salt thereof. In some embodiments, one of M1 and M2 is and the other of M1 and M2 is . For example, M1 may be . In some embodiments, the compound or salt is represented by Formula (B-Ia): (B-Ia); or a pharmaceutically acceptable salt thereof. In some embodiments, M2 is . In some embodiments, the compound or salt is represented by Formula (B-Ib):
(B-Ib); or a pharmaceutically acceptable salt thereof. In some embodiments, M1 is and M2 is . In other embodiments, M1 is and M2 is . In some embodiments, the compound or salt is represented by Formula (B-Ic) or Formula (B-Id): (B-Ic) (B-Id) or a salt thereof. In some embodiments, R3 for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), and (B-Id) is selected from hydrogen and -C1-C10 alkyl optionally substituted with one or more substituents independently selected from a halogen, -NO2, -CN, -OR10, -SR10, −N(R10)2, - C(O)R10, -C(O)N(R10)2, -N(R10)C(O)R10 , -C(O)OR10, and -OC(O)R10. For example, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), and (B-Id), R3 may be selected from hydrogen and -C1-C10 alkyl optionally substituted with one or more substituents independently selected from a halogen, -NO2, -CN, -OR10, -SR10, and -N(R10)2. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), and (B-Id), R3 is hydrogen. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), and (B-Id), n is 0. In other embodiments, for a compound or salt of any of Formulas (B-
I), (B-Ia), (B-Ib), (B-Ic), and (B-Id), n is selected from 1, 2, 3, and 4. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), and (B-Id), each R1 is independently selected from a halogen, -OR10, -SR10, -N(R10)2, -C(O)R10, -C(O)N(R10)2, −N(R10)C(O)R10 , -C(O)OR10, -OC(O)R10, -S(O)R10, -S(O)2R10, -S(O)2N(R10)2, -P(O)(OR10)2, −OP(O)(OR10)2, -NO2, and -CN; and -C1-C10 alkyl, -C2-C10 alkenyl, and -C2-C10 alkynyl, each of which is optionally substituted at each occurrence with one or more substituents independently selected from a halogen, -OR10, -SR10, -N(R10)2, -C(O)R10, -C(O)N(R10)2, -N(R10)C(O)R10 , −C(O)OR10, -OC(O)R10, -S(O)R10, -S(O)2R10, -S(O)2N(R10)2, -P(O)(OR10)2, -OP(O)(OR10)2, −NO2, =O, =S, =N(R10), -CN, a C3-C10 carbocycle, and a 3- to 10-membered heterocycle. In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), and (B-Id), each R1 is selected from a halogen, -OR10, -SR10, -N(R10)2, -NO2, and -CN; and -C1-C10 alkyl optionally substituted at each occurrence with one or more substituents independently selected from a halogen, -OR10, -SR10, -N(R10)2, -C(O)R10, -NO2, and -CN. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), and (B-Id), each R1 is independently selected from a -C1-C10 alkyl optionally substituted at each occurrence with one or more substituents independently selected from a halogen, -OR10, -SR10, - N(R10)2, -C(O)R10, -NO2, and −CN. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), and (B-Id), m is 0. In other embodiments, for a compound or salt of any of Formulas (B- I), (B-Ia), (B-Ib), (B-Ic), and (B-Id), m is selected from 1, 2, 3, and 4. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), and (B-Id), each R2 is independently selected from a halogen, -OR10, -SR10, -N(R10)2, -C(O)R10, -C(O)N(R10)2, −N(R10)C(O)R10 , −C(O)OR10, −OC(O)R10, −S(O)R10, −S(O)2R10, −S(O)2N(R10)2, −P(O)(OR10)2, −OP(O)(OR10)2, −NO2, and −CN; and −C1-C10 alkyl, −C2-C10 alkenyl, and −C2-C10 alkynyl, each of which is optionally substituted at each occurrence with one or more substituents independently selected from a halogen, −OR10, −SR10, −N(R10)2, −C(O)R10, −C(O)N(R10)2, −N(R10)C(O)R10 , −C(O)OR10, −OC(O)R10, −S(O)R10, −S(O)2R10, −S(O)2N(R10)2, −P(O)(OR10)2, −OP(O)(OR10)2, −NO2, =O, =S, =N(R10), −CN, a C3-C10 carbocycle, and a 3- to 10-membered heterocycle. In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), and (B-Id), each R2 is independently selected from a halogen, −OR10, −SR10,
−N(R10)2, −NO2, and −CN; and −C1-C10 alkyl optionally substituted at each occurrence with one or more substituents independently selected from a halogen, −OR10, −SR10, −N(R10)2, −C(O)R10, −NO2, and −CN. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), and (B-Id), each R2 is independently selected from −OR10 and −C1-C10 alkyl optionally substituted with one or more substituents independently selected from a halogen, −OR10, −SR10, −N(R10)2, −C(O)R10, −NO2, and −CN. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), and (B-Id), m is 1 and R2 is −CH3. For example, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), and (B-Id), M2 may be . In a particular example, for a compound or salt of any of Formulas (B-I), (B- Ia), (B-Ib), (B-Ic), and (B-Id), M2 is . In some embodiments, the compound of Formula (B-I) is represented by Formula (B-Ie): (B-Ie); or a salt thereof. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), Q is selected from −(CR10 2)p−, −(CR10 2)qO(CR10 2)q−, −(CR10 2)qS(CR10 2)q−, and −(CR10 2)qNR10(CR10 2)q−, where p is selected from 1, 2, 3, 4, and 5 and q is independently selected at each occurrence from 0, 1, 2, 3, 4, and 5. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), Q is selected from −(CR10 2)p−. For example, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), p may be 1 such that Q is −C(R10)2−. Alternatively, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), Q may be selected from −(CR10 2)qO(CR10 2)q−, −(CR10 2)qS(CR10 2)q−, and −(CR10 2)qNR10(CR10 2)q−. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B- Id), and (B-Ie), Q is −CH2NH−. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), Q is selected from
−(CR10 2)qNR10(CR10 2)q−. In some embodiments, for a compound or salt of any of Formulas (B- I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), Q is selected from −(CR10 2)qNR10−. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), Q is selected from −(CR10 2)qNR10−. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), Q is −CR10 2NR10(CR10 2)1-2−. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B- Id), and (B-Ie), Q is −CH2NHCH2− or −CH2NHCH2CH2−. In another example, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), Q is −CH2NHCH2−. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), each R10 is independently selected from hydrogen; and −C1-C10 alkyl, −C2-C10 alkenyl, and −C2-C10 alkynyl, each of which is optionally substituted at each occurrence with one or more substituents independently selected from a halogen, −OH, −CN, −NO2, and −NH2. In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), R10 is hydrogen at each occurrence. In some embodiments, for a compound or salt of any one of Formulas (B-I), (B-Ia), (B- Ib), (B-Ic), (B-Id), and (B-Ie), each R10 of Q is hydrogen at each occurence. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from a saturated C3-C7 carbocycle optionally substituted with one or more substituents independently selected at each occurrence from R13. For example, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T may be a saturated C3 carbocycle optionally substituted with one or more substituents independently selected at each occurrence from R13. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T may be selected from: , , , , and , each of which is optionally substituted with one or more substituents independently selected at each occurrence from R13. In some embodiments, for a compound or salt of any of Formulas (B- I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from , , ,
, , , and . For example, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T may be: . In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from a C5-12 bicyclic carbocycle optionally substituted with one or more substituents independently selected at each occurrence from R13. In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from a saturated C5-12 bridged carbocycle optionally substituted with one or more substituents independently selected at each occurrence from R13, such as an optionally substituted C5 bridged carbocycle. For example, for a compound or salt of any of Formulas (B- I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T may be selected from: , and , each of which is optionally substituted with one or more substituents independently selected at each occurrence from R13. In other embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from C8-11 bicyclic carbocycle optionally substituted with one or more substituents independently selected at each occurrence from R13. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is a fused bicyclic ring. In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from a C8-11 fused bicyclic carbocycle. In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B- Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from:
, , , , , , , , , , , , , , and , and wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. For example, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T may be selected from: , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B- Id), and (B-Ie), T may be selected from: and . In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T may be selected from: and .
In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from: , , , , , , , , , , and . In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from naphthalene, 1,2,3,4-tetrahydronaphthalene, decahydronaphthalene, octahydro-1H-indene, 2,3-dihydro-1H-indene, 1H-indene, octahydropentalene, decahydro-1H-benzo[7]annulene, 7H-benzo[7]annulene, 4aH- benzo[7]annulene, 6,7,8,9-tetrahydro-5H-benzo[7]annulene, 2,3,4,5-tetrahydro-1H- benzo[7]annulene, 2,3,4,7-tetrahydro-1H-benzo[7]annulene, azulene, and decahydroazulene, and wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from a 4- to 12-membered heterocycle optionally substituted with one or more substituents independently selected at each occurrence from R13. In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B- Id), and (B-Ie), T is selected from:
, , , , , , , , , , , , , , , , , , , and , any one of which is optionally substituted with one or more substituents independently selected at each occurrence from R13. In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from: , , , , , , , , , , , , , , , ,
In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from a 7- to 12-membered bicyclic heterocycle optionally substituted with one or more substituents independently selected at each occurrence from R13. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B- Id), and (B-Ie), T is selected from an 8- to 11-membered bicyclic heterocycle optionally substituted with one or more substituents independently selected at each occurrence from R13, such as an 8- to 11-membered bicyclic heteroaryl group. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from:
wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. In some embodiments, for a compound or salt of any one of Formulas (B-I), (B-Ia), (B- Ib), (B-Ic), (B-Id), and (B-Ie), −Q−T is selected from:
wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. In some embodiments, −Q−T is selected from
In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T has one to four ring heteroatoms independently selected from N, O, S, and B. In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B- Ib), (B-Ic), (B-Id), and (B-Ie), T has at least one ring heteroatom that is boron. For example, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T may be selected from:
wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T may be selected from , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is represented by: , wherein dashed lines represent single or double bonds, valence permitting; k is selected from 0, 1, 2, and 3; and W, X, Y, and Z are independently selected from N(R10)g and C(R10)h, wherein g is selected from 0 and 1 and h is selected from 1 and 2, and wherein a straight line linked to a wavy line indicates connectivity to Q from any position, valence permitting, of the bicyclic heterocycle. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is represented by: . In other embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is represented by:
. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from: , , , , , , , , , , , , , , , , , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. For example, for a compound or salt of any of Formulas (B-I), (B-Ia), (B- Ib), (B-Ic), (B-Id), and (B-Ie), T may be selected from: , , , , , , , , , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from: , , , , , , , , , , , , ,
, , , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T may be selected from: , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. For example, for a compound or salt of any of Formulas (B-I), (B-Ia), (B- Ib), (B-Ic), (B-Id), and (B-Ie), T may be selected from: , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T may be selected from: , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. For example, for a compound or salt of any of Formulas (B-I), (B-Ia), (B- Ib), (B-Ic), (B-Id), and (B-Ie), T may be selected from: , , , and ,
wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T may be selected from: and . In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T may be selected from: . For example, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T may be selected from: , , , , , , , and . In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from: , , , , , , , , , , , , , , , , , , , ,
, , , , , , , , , , , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from: , , , , , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from: , , , , , , , , , , , , , , , , , , , and ,wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13.
In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from: , , , , , , , , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), T is selected from 1,2,3,4-tetrahydroquinoline, 1,2,3,4- tetrahydroisoquinoline, 1,2-dihydroquinoline, 1,2-dihydroisoquinoline, 1,2,3,4- tetrahydroquinazoline, decahydroquinoline, decahydroisoquinoline, quinoline, isoquinoline, quinoxaline, phthalazine, quinazoline, and cinnoline, wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13. In some embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), each R13 may be independently selected from: a halogen, −OR10, −SR10, −N(R10)2, −C(O)R10, −C(O)N(R10)2, −N(R10)C(O)R10 , −C(O)OR10, −OC(O)R10, −S(O)R10, −S(O)2R10, −S(O)2N(R10)2, −P(O)(OR10)2, −OP(O)(OR10)2, −NO2, =O, =S, =N(R10), and −CN; and −C1-C10 alkyl, −C2-C10 alkenyl, and −C2-C10 alkynyl, each of which is optionally substituted at each occurrence with one or more substituents independently selected from a halogen, −OR10, −SR10, −N(R10)2, −C(O)R10, −C(O)N(R10)2, −N(R10)C(O)R10 , −C(O)OR10, −OC(O)R10, −S(O)R10, −S(O)2R10, −S(O)2N(R10)2, −P(O)(OR10)2, −OP(O)(OR10)2, −NO2, =O, =S, =N(R10), −CN, a C3-C10 carbocycle, and a 3- to 10-membered heterocycle. In certain embodiments, for a compound or salt of any of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie), each R13 may be independently selected from a halogen, −OR10, −SR10, −N(R10)2, −NO2, and −CN; and −C1-C10 alkyl optionally substituted at each occurrence
with one or more substituents independently selected from a halogen, −OR10, −SR10, −N(R10)2, −C(O)R10, −NO2, and −CN. In certain embodiments, for a compound or salt of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie): Q is selected from a bond, −(CR10 2)p−, and −(CR10 2)qNR10(CR10 2)q−; T is selected from an optionally substituted saturated C3-C7 carbocycle, an optionally substituted C5-12 bicyclic carbocycle, and an optionally substituted 4- to 12-membered heterocycle, wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13, wherein R13 is independently selected at each occurrence from halogen, −OR10, −N(R10)2, −C(O)R10, −C(O)OR10, −N(R10)C(O)R10, and −C1-C3 alkyl optionally substituted with one or more substituents independently selected from a halogen, −OR10 and −N(R10)2; and R10 is as set forth herein (and in some aspects, R10 is H or C1-C3 alkyl). In certain embodiments, for a compound or salt of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie): Q is selected from a bond, −(CR10 2)p−, and −(CR10 2)qNR10(CR10 2)q−; T is selected from an optionally substituted saturated C3-C7 carbocycle, an optionally substituted C5-12 bicyclic carbocycle, and an optionally substituted 4- to 12-membered heterocycle, wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13, wherein R13 is independently selected at each occurrence from halogen, −OR10, −N(R10)2, and −C1-C3 alkyl optionally substituted with one or more substituents independently selected from a halogen, −OR10 and −N(R10)2; and R10 is as set forth herein (and in some aspects, R10 is H or C1-C3 alkyl). In certain embodiments, for a compound or salt of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie): Q is selected from a bond, −CH2−, −CH2NH−, −CH2NHCH2−, and −CH2NHCH2CH2−; and T is selected from:
, , , , , , , , , , , , , , , , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13 wherein R13 is independently selected at each occurrence from halogen, −OR10, −N(R10)2, −C(O)R10, −C(O)OR10, −N(R10)C(O)R10, and −C1-C3 alkyl optionally substituted with one or more substituents independently selected from a halogen, −OR10 and −N(R10)2; and R10 is as set forth herein (and in some aspects, R10 is H or C1-C3 alkyl). In certain embodiments, for a compound or salt of Formulas (B-I), (B-Ia), (B-Ib), (B-Ic), (B-Id), and (B-Ie): Q is selected from −CH2−, −CH2NH−, −CH2NHCH2−, and −CH2NHCH2CH2−; and T is selected from: , , , , , , , , , and , wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13, wherein R13 is independently selected at each occurrence from halogen, −OH, −NH2, and −C1-C3 alkyl. In some embodiments, the compound of Formula (B-I) is selected from:
, and a salt of any one thereof. In some embodiments, the compound of Formula (B-I) is selected from the compounds in Table 15. TGFβR1 Inhibitors of Formula (C-I) In some aspects, the TGFβR1 inhibitor is a compound of Formula (C-I):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein: L is −[CR′2]p−L’−[CH2]q− L’ is absent, −S−, −O−, or −NH−; A is absent, carbocycle, or heterocycle; Q3 is N or CR3; Q4 is N or CR4; Q5 is N or CR5; Q6 is N or CR6; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R3 is H, halo, C1–3alkyl, C1–3haloalkyl, or C1–3 alkoxy; R4 is H, halo, C1–3 alkyl, C1–3haloalkyl, or C1–3 alkoxy; R5 is H, halo, C1–3 alkyl, C1–3haloalkyl, or C1–3 alkoxy; R6 is H, halo, C1–3 alkyl, C1–3haloalkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R3, R4, R5, R6, and R9 are each substituted with 0–3 R10;
R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; n is 0–5; p is 1-3; and q is 0-3. In certain embodiments, compounds are provided having the structure of Formula (C-I), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein L’ is absent. In other embodiments, L’ is −S−. In other embodiments, L’ is −O−. In still other embodiments, L’ is −NH−. In certain embodiments, compounds are provided having the structure of Formula (C-I), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein p is 1. In some embodiments, p is 2. In other embodiments, p is 3. In certain embodiments, compounds are provided having the structure of Formula (C-I), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein q is 0. In other embodiments, q is 1. In still other embodiments, q is 2. In further embodiments, q is 3. In certain embodiments, compounds are provided having the structure of Formula (C-II):
(C-II) or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; Q3 is N or CR3;
Q4 is N or CR4; Q5 is N or CR5; Q6 is N or CR6; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R3 is H, halo, C1–3alkyl, C1–3haloalkyl, or C1–3 alkoxy; R4 is H, halo, C1–3 alkyl, C1–3haloalkyl, or C1–3 alkoxy; R5 is H, halo, C1–3 alkyl, C1–3haloalkyl, or C1–3 alkoxy; R6 is H, halo, C1–3 alkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R3, R4, R5, R6, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of Formula (C-II-A):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R6 is H, halo, C1–3 alkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R3, R4, R5, R6, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of Formula (C-II-B):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of Formula (II-C):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R6 is H, halo, C1–3 alkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R6, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of Formula (C-III):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; Q3 is N or CR3; Q4 is N or CR4; Q5 is N or CR5; Q6 is N or CR6; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R3 is H, halo, C1–3alkyl, C1–3haloalkyl, or C1–3 alkoxy; R4 is H, halo, C1–3 alkyl, C1–3haloalkyl, or C1–3 alkoxy; R5 is H, halo, C1–3 alkyl, C1–3haloalkyl, or C1–3 alkoxy; R6 is H, halo, C1–3 alkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R3, R4, R5, R6, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy;
R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of Formula (C-III-A):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R6 is H, halo, C1–3 alkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R3, R4, R5, R6, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl;
m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of Formula (C-III-B):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; and n is 0–5.
In certain embodiments, compounds are provided having the structure of Formula (C-III-C):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R6 is H, halo, C1–3 alkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R6, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of Formula (C-IV):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; Q3 is N or CR3; Q4 is N or CR4; Q5 is N or CR5; Q6 is N or CR6; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R3 is H, halo, C1–3alkyl, C1–3haloalkyl, or C1–3 alkoxy; R4 is H, halo, C1–3 alkyl, C1–3haloalkyl, or C1–3 alkoxy; R5 is H, halo, C1–3 alkyl, C1–3haloalkyl, or C1–3 alkoxy; R6 is H, halo, C1–3 alkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R3, R4, R5, R6, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy;
R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of Formula (C-IV-A):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R6 is H, halo, C1–3 alkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R3, R4, R5, R6, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl;
m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of Formula (C-IV-B):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; and n is 0–5.
In certain embodiments, compounds are provided having the structure of Formula (C-IV-C):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R6 is H, halo, C1–3 alkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R6, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of Formula (C-V):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; Q3 is N or CR3; Q4 is N or CR4; Q5 is N or CR5; Q6 is N or CR6; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R3 is H, halo, C1–3alkyl, C1–3haloalkyl, or C1–3 alkoxy; R4 is H, halo, C1–3 alkyl, C1–3haloalkyl, or C1–3 alkoxy; R5 is H, halo, C1–3 alkyl, C1–3haloalkyl, or C1–3 alkoxy; R6 is H, halo, C1–3 alkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R3, R4, R5, R6, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy;
R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of Formula (C-V-A):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R6 is H, halo, C1–3 alkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R6, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl;
m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of Formula (C-V-B):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; and n is 0–5.
In certain embodiments, compounds are provided having the structure of Formula (C-V-C):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: A is carbocycle or heterocycle; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R6 is H, halo, C1–3 alkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R6, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of Formula (C-VI):
(C-VI) or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: Q3 is N or CR3; Q4 is N or CR4; Q5 is N or CR5; Q6 is N or CR6; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R3 is H, halo, C1–3alkyl, C1–3haloalkyl, or C1–3 alkoxy; R4 is H, halo, C1–3 alkyl, C1–3haloalkyl, or C1–3 alkoxy; R5 is H, halo, C1–3 alkyl, C1–3haloalkyl, or C1–3 alkoxy; R6 is H, halo, C1–3 alkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R3, R4, R5, R6, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl;
m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of Formula (C-VI-A):
(C-VI-A) or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R6 is H, halo, C1–3 alkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R6, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; and n is 0–5.
In certain embodiments, compounds are provided having the structure of Formula (C-VI-B):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of Formula (C-VI-C):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl, wherein R1 is substituted with 0–3 R10; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R6 is H, halo, C1–3 alkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R6, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein R1 is hydrogen. In other embodiments, R1 is unsubstituted C1–3 alkyl. In certain embodiments, R1 is methyl. In other embodiments, R1 is C1–3 alkyl substituted with 1–3 R10. In certain embodiments, R1 is
−CH2OCH3. In another embodiment, R1 is unsubstituted C1–3 haloalkyl. In certain embodiments, R1 is −CF3. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein m is 0. In other embodiments, m is 1–3. In certain embodiments, m is 1. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein at least one R2 is halo. In some embodiments, at least one R2 is F. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein A is heterocycle. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein A is carbocycle. In certain embodiments, A is phenyl. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein n is 0. In some embodiments, n is 1–5. In other embodiments, n is 1 or 2. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein at least one R9 is halo. In some embodiments, at least one R9 is F or Cl. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein at least one R9 is −OR′. In some embodiments, R′ of the at least one −OR′ is C1–6 alkyl.
In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein at least one R9 is C1–4 alkyl. In some embodiments, at least one R9 is methyl. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein at least one R9 is −CN. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein at least one R9 is −C(O)N(R′)2. In some embodiments, at least one R9 is −C(O)N(CH3)2. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein at least one R9 is −N(R′)2. In some embodiments, at least one R9 is −N(CH3)2. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein at least one R9 is −NO2. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein R6 is H. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein R6 is halo. In some embodiments, R6 is F or Cl. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein R6 is C1–3 alkyl. In some embodiments, R6 is methyl.
In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein R6 is C1–3 alkoxy. In some embodiments, R6 is −OCH3. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein the reactive moiety capable of attachment to a linker or the reactive moiety capable of attachment to an antibody is −Y−(CR11R12)p−NHR13, −Y−(CR11R12)p−C(O)OH, or −Y−(CR11R12)p−C(O)NHR13, wherein: Y is −O−, −CH2−, −OCH2CH2−, or −OCH2CH2NR14−; R11 is at each occurrence, independently, H, halogen, C1–3 alkyl, or C1–3 haloalkyl; R12 is at each occurrence, independently, H, halogen, C1–3 alkyl, or C1–3 haloalkyl; R13 is H; or R11 at one occurrence and R12 at one occurrence, together with the atom to which they are attached, form a ring; or R11 at one occurrence and R13 form a ring; R14 is H, halogen, C1–3 alkyl, or C1–3 haloalkyl; and p is 0–5. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein R7 is −Y−(CR11R12)p−NHR13. In some embodiments, Y is
. In other embodiments, Y is −CH2−. In still other embodiments, Y is −OCH2CH2−. In further embodiments, Y is−OCH2CH2NR14−. In certain embodiments, compounds are provided having the structure of any one of Formula (C-I), (C-II), (C-III), (C-IV), (C-V), or (C-VI), or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein R7 is:
Representative compounds of Formula (C-I), and Formulas (C-II) through (C-VI) as applicable, include the compounds listed in Tables 16–20 below, as well as pharmaceutically acceptable salts thereof. To this end, representative compounds are identified herein by their respective “Compound Number”, which is sometimes abbreviated as “Compound No.”, “Cmpd No.” or “No.” TGFβR1 Inhibitors of Formula (D-I) In some aspects, the TGFβR1 inhibitor is a compound of Formula (D-I):
(D-I) or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: QA is CRA or N; QB is CRB; Q3 is N or CR3; Q4 is N or CR4; Q5 is N or CR5; RA is H, halo, C1-3 alkyl, or C1-3 haloalkyl and RB is B
R , together with the atoms to which they are attached, form a heterocyclic ring; Ra and Rb are each H, or Ra and Rb, together with the atoms to which they are attached, form a heterocyclic ring; ring B is carbocycle or heterocycle; R1 is H, C1-3 alkyl, or C1-3 haloalkyl;
R2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; R3 is H, halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; R4 is H, halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; R5 is H, halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; wherein R1, R2, R3, R4, and R5 are, at each occurrence, independently substituted with 0-3 R10; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R9 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, or C1-3 haloalkoxy; R10 is, at each occurrence, independently C1-3 alkoxy or C1-3 haloalkoxy; m is 0–3; and n is 0–5. In certain embodiments, compounds are provided having the structure of Formula (D-II):
(D-II) or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: Q3 is N or CR3; Q4 is N or CR4; Q5 is N or CR5; Q6 is N or CR6;
Ra and Rb are each H, or Ra and Rb, together with the atoms to which they are attached, form a heterocyclic ring; R1 is H, C1-3 alkyl, or C1-3 haloalkyl; R2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; R3 is H, halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; R4 is H, halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; R5 is H, halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; R6 is H, halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; wherein R1, R2, R3, R4, R5, and R6 are, at each occurrence, independently substituted with 0-3 R10; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R10 is, at each occurrence, independently C1-3 alkoxy or C1-3 haloalkoxy; and m is 0–3. In certain embodiment of Formula (II), Q6 is CR6 when R1 is methyl and Ra and Rb, together with the atoms to which they are attached, form a heterocyclic ring. In certain embodiments, compounds are provided having the structure of Formula (D-III):
(D-III) or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: Q6 is N or CR6;
R1 is H, C1-3 alkyl, or C1-3 haloalkyl; R2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; R6 is H, halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; wherein R1, R2, and R6 are, at each occurrence, independently substituted with 0-3 R10; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody; R10 is, at each occurrence, independently C1-3 alkoxy or C1-3 haloalkoxy; and m is 0–3. wherein Q6 is CR6 when R1 is methyl. In certain embodiments, compounds are provided having the structure of Formula (D-IV):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: Q6 is N or CR6; R1 is H, C1-3 alkyl, or C1-3 haloalkyl; R2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; R6 is H, halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; wherein R1, R2, and R6 are each substituted with 0–3 R10;
R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R10 is, at each occurrence, independently C1-3 alkoxy or C1-3 haloalkoxy; and m is 0–3. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is H. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is C1-3 alkyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is methyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, R1 is ethyl or propyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, R1 is C1-3 haloalkyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is −CFH2, −CF2H, or −CF3. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein m is 0. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein m is 1–3. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein Q6 is N. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein Q6 is CR6. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R6 is H. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R6 is halo, C1-3 alkyl, or C1-3 haloalkyl.
In certain embodiments, compounds are provided wherein the TGFβR1 inhibitor is a compound having the structure of Formula (D-V):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: Ra and Rb are each H, or Ra and Rb, together with the atoms to which they are attached, form a heterocyclic ring; QA is CRA or N; Q3 is N or CR3; Q4 is N or CR4; Q5 is N or CR5; RA is H, halo, C1-3 alkyl, or C1-3 haloalkyl; Ra and Rb are each H, or Ra and Rb, together with the atoms to which they are attached, form a heterocyclic ring; ring B is carbocycle or heterocycle; R1 is H, C1-3 alkyl, or C1-3 haloalkyl; R2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; R3 is H, halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; R4 is H, halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; R5 is H, halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; wherein R1, R2, R3, R4, and R5 are, at each occurrence, independently substituted with 0– 3 R10;
R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R9 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, or C1-3 haloalkoxy; R10 is, at each occurrence, independently C1-3 alkoxy or C1-3 haloalkoxy; m is 0–3; and n is 0–5. In certain embodiments, compounds are provided wherein the TGFβR1 inhibitor is a compound having the structure of Formula (D-VI):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: QA is CRA or N; RA is H, halo, C1-3 alkyl, or C1-3 haloalkyl; ring B is carbocycle or heterocycle; R1 is H, C1-3 alkyl, or C1-3 haloalkyl; R2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; wherein R1 and R2 are, at each occurrence, independently substituted with 0-3 R10; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety;
R9 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, or C1-3 haloalkoxy; R10 is, at each occurrence, independently C1-3 alkoxy or C1-3 haloalkoxy; m is 0–3; and n is 0–5. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is heterocycle. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is heteroaryl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, isoxazolyl, oxazolyl, isothiazolyl, thiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, or pyrazinyl. In certain embodiments, compounds are provided having the structure of Formula (D-VI-A):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: QA is CRA or N; RA is H, halo, C1-3 alkyl, or C1-3 haloalkyl; R1 is H, C1-3 alkyl, or C1-3 haloalkyl; R2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy;
R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; wherein R1 and R2 are, at each occurrence, independently substituted with 0-3 R10; R9 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, or C1-3 haloalkoxy; R10 is, at each occurrence, independently C1-3 alkoxy or C1-3 haloalkoxy; m is 0–3; and n is 0–2. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is carbocycle. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is aryl. In certain embodiments, compounds are provided having the structure of Formula (D-VI-B):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: QA is CRA or N; RA is H, halo, C1-3 alkyl, or C1-3 haloalkyl; R1 is H, C1-3 alkyl, or C1-3 haloalkyl; R2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy;
wherein R1 and R2 are, at each occurrence, independently substituted with 0-3 R10; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R9 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, or C1-3 haloalkoxy; R10 is, at each occurrence, independently C1-3 alkoxy or C1-3 haloalkoxy; m is 0–3; and n is 0–4. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein QA is N. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein QA is CRA. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein RA is H. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein RA is halo. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein RA is F. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein RA is C1-3 alkyl, or C1-3 haloalkyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is H. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is C1-3 alkyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is methyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is ethyl or propyl.
In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is C1-3 haloalkyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is −CFH2, −CF2H, or −CF3. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein m is 0. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein m is 1–3. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein n is 0. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein n is 1–2. In certain embodiments, compounds are provided, wherin the TGFβR1 inhibitor is a compound having the structure of Formula (D-VII):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: QA is CRA or N; RA is H, halo, C1-3 alkyl, or C1-3 haloalkyl; ring B is carbocycle or heterocycle; R1 is H, C1-3 alkyl, or C1-3 haloalkyl;
R2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; wherein R1 and R2 are, at each occurrence, independently substituted with 0-3 R10; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R9 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, or C1-3 haloalkoxy; R10 is, at each occurrence, independently C1-3 alkoxy; m is 0–3; and n is 0–5. In certain embodiments of Formula (D-VII), compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: R1 is not H or methyl when ring B is carbocycle; ring B is a five membered heteroaryl when R1 is H and QA is N; and R7 is substituted with an acidic amino acid sidechain when R1 is methyl and QA is CRA. In certain embodiments, compounds are provided having the structure of Formula (D-VIII):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: RA is H, halo, C1-3 alkyl, or C1-3 haloalkyl; ring B is carbocycle or heterocycle;
R1 is H, ethyl, propyl, or C1-3 haloalkyl; R2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; wherein R1 and R2 are, at each occurrence, independently substituted with 0-3 R10; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R9 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, or C1-3 haloalkoxy; R10 is, at each occurrence, independently C1-3 alkoxy; m is 0–3; and n is 0–5; wherein R1 is ethyl, propyl, or C1-3 haloalkyl when ring B is carbocycle. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is heterocycle. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is heteroaryl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, isoxazolyl, oxazolyl, isothiazolyl, thiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, or pyrazinyl. In certain embodiments, compounds are provided having the structure of Formula (D-VIII-A):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: RA is H, halo, C1-3 alkyl, or C1-3 haloalkyl; R1 is H, ethyl, propyl, or C1-3 haloalkyl; R2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; wherein R1 and R2 are, at each occurrence, independently substituted with 0-3 R10; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R9 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, or C1-3 haloalkoxy; R10 is, at each occurrence, independently C1-3 alkoxy; m is 0–3; and n is 0–2. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is carbocycle. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is aryl. In certain embodiments, compounds are provided having the structure of Formula (D-VIII-B):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: RA is H, halo, C1-3 alkyl, or C1-3 haloalkyl; R1 is ethyl, propyl, or C1-3 haloalkyl; R2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; wherein R1 and R2 are, at each occurrence, independently substituted with 0-3 R10; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R9 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, or C1-3 haloalkoxy; R10 is, at each occurrence, independently C1-3 alkoxy or C1-3 haloalkoxy; m is 0–3; and n is 0–4. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein RA is H. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein RA is halo. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein RA is F.
In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein RA is C1-3 alkyl, or C1-3 haloalkyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is H. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is ethyl or propyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is C1-3 haloalkyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is −CFH2, −CF2H, or −CF3. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein m is 0. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein m is 1–3. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein n is 0. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein n is 1–2. In certain embodiments, compounds are provided having the structure of Formula (D-IX):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: ring B is carbocycle or heterocycle; R1 is H, C1-3 alkyl, or C1-3 haloalkyl; R2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; wherein R1 and R2 are, at each occurrence, independently substituted with 0-3 R10; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R9 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, or C1-3 haloalkoxy; R10 is, at each occurrence, independently C1-3 alkoxy; m is 0–3; and n is 0–5; wherein: R1 is ethyl, propyl, or C1-3 haloalkyl when ring B is carbocycle; and ring B is a 5-membered heteroaryl when R1 is H In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is heterocycle. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is heteroaryl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is a 5-membered heteroaryl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, isoxazolyl, oxazolyl, isothiazolyl, thiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, or pyrazinyl. In certain embodiments, compounds are provided having the structure of Formula (D-IX-A):
or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: R1 is H, C1-3 alkyl, or C1-3 haloalkyl; R2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; wherein R1 and R2 are, at each occurrence, independently substituted with 0-3 R10; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R9 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, or C1-3 haloalkoxy; R10 is, at each occurrence, independently C1-3 alkoxy or C1-3 haloalkoxy; m is 0–3; and n is 0–2. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is carbocycle. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein ring B is aryl. In certain embodiments, compounds are provided having the structure of Formula (D-IX-B):
R1 is ethyl, propyl, or C1-3 haloalkyl; R2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; wherein R1 and R2 are, at each occurrence, independently substituted with 0-3 R10; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R9 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, or C1-3 haloalkoxy; R10 is, at each occurrence, independently C1-3 alkoxy or C1-3 haloalkoxy; m is 0–3; and n is 0–4. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is H. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is C1-3 alkyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is methyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is ethyl or propyl. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is C1-3 haloalkyl.
In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R1 is −CFH2, −CF2H, or −CF3. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein m is 0. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein m is 1–3. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein n is 0. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein n is 1–2. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R7, the reactive moiety capable of attachment to a linker or the reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety, is −Y−(CR11R12)p−NHR13, −Y−(CR11R12)p−C(O)OR13, or −Y−(CR11R12)p−C(O)NHR13, wherein: Y is −O−, −CH2−, −OCH2CH2−, −OCH2CH2NR14−, −C(O)NR14−; −CH2C(O)NR14−, −CH2CH2C(O)NR14−, −C(O)O−; −CH2C(O)O−, −CH2CH2C(O)O−,, or −CH2CH2NR14C(O)C(O)−, R11 is at each occurrence, independently, H, halo, C1–3 alkyl, C1–3 haloalkyl, or R15; R12 is at each occurrence, independently, H, halo, C1–3 alkyl, C1–3 haloalkyl, or R15; R13 is H, C1-3 alkyl, or C1-3 haloalkyl; or R11 at one occurrence and R12 at one occurrence, together with the atom to which they are attached, form a ring; or R11 at one occurrence and R13 form a ring; R14 is H, halo, C1-3 alkyl, or C1-3 haloalkyl; and R15 is an amino acid side chain;−C1-3 alkyl−C(O)OR13; p is 0–5. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R15 is −C1-3 alkyl−C(O)OR13;
In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R7 is −Y−(CR11R12)p−NHR13. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein Y is −O−. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein Y is −CH2−. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein Y is −OCH2CH2−. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein Y is−OCH2CH2NR14−. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R7 is:
. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R7, the reactive moiety capable of attachment to a linker or the reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety, comprises a substitutable nitrogen atom. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein R7, the reactive moiety capable of attachment to a linker or the reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety, comprises a moiety that enhances cell permeability. In certain embodiments, compounds are provided, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein the compound has a pKa of about 3.5 to about 5.5. Representative compounds of Formula (D-I), and Formulas (D-II) through (D-IX) as applicable, include the compounds listed in Tables 21–24 below, as well as pharmaceutically acceptable salts thereof. To this end, representative compounds are identified herein by their respective “Compound Number”, which is sometimes abbreviated as “Compound No.”, “Cmpd No.” or “No.”
Linker Region L3 The conjugates of the present disclosure include a linker region, L3. As shown in Formula (I), Region L3 covalently links Region G, which comprises the GalNAc moiety, to Region Inh, which comprises the TGFβR1 inhibitor. The covalent linkages can be formed by reaction between functional groups in synthetic precursors for each region. Linkers of the disclosure (L3) may have from about 10 to about 500 atoms in a linker, such as from about 10 to about 400 atoms, such as about 10 to about 300 atoms in a linker. In certain embodiments, linkers of the disclosure have from about 30 to about 400 atoms, such as from about 30 to about 300 atoms in the linker. In the conjugates, an inhibitor as described herein is covalently bound to a GalNAc moiety by way of a linker, also referred to herein as L3. L3, as used herein, may be selected from any of the linker moieties discussed herein. As will be appreciated by skilled artisans, a conjugate may be prepared by contacting an GalNAc moiety with a linker-compound described herein under conditions in which the linker- compound covalently links to the GalNAc moiety. One embodiment pertains to a method of making a conjugate by contacting a linker-compound with a unit comprising Region G under conditions in which the linker-compound covalently links to the GalNAc moiety. In certain embodiments, any one of the inhibitors described herein is covalently bound to a linker (L3). The linker may be covalently bound to any position on the inhibitor, valence permitting. The linker may comprise a reactive moiety, e.g., a nucleophile or an an electrophile that can react to form a covalent bond. Linkers can be short, long, flexible, rigid, cleavable, non-cleavable, hydrophilic, hydrophobic, unbranched (e.g., where z is 1), or branched (e.g., where z is greater than 1). A linker can contain connector segments that have different characteristics, such as segments of flexibility and segments of rigidity, or segments that are cleavable and segments that are cleavable. A linker can contain multiple segments, such as one or more non-cleavable segments and one or more cleavable segments. The linkers may be polyvalent such that they covalently link more than one inhibitor to a single site on the GalNAc moiety, or monovalent such that covalently they link a single inhibitor to a single site on the GalNAc moiety. A linker can comprise alkylene, alkenylene, alkynylene, polyether, polyester, polyamide, polyamino acid,
polypeptide, cleavable peptide, and/or para-aminobenzylcarbamate groups. In some embodiments, the linker comprises a “non-cleavable” segment, such as a “non-cleavable linker,” that is chemically stable to extracellular environments, for example, chemically stable in the blood stream and in intracellular environments. In some embodiments, the linker comprises a “cleavable” segment, such as a “cleavable linker,” that includes one or more linkages that are not stable, such as linkages that are designed to cleave and/or immolate or otherwise breakdown specifically or non-specifically in the blood stream and/or inside cells (i.e., in an intracellular environment). Linkers comprise one or more cleavable segments, one or more non-cleavable segments, or a combination thereof. In some embodiments, a moiety, construct, or conjugate described herein includes the symbol , which indicates the point of attachment, e.g., the point of attachment of a chemical or functional moiety to the compound, the point of attachment of a linker to a compound of the disclosure, or the point of attachment of a linker to a GalNAc moiety. Sulfamide linkers may be used to link many compounds of the present invention to a GalNAc moiety. Sulfamide linkers are as described herein and e.g., U.S. Patent Publication Number 2019/0038765, the linkers of which are incorporated by reference herein. Cleavable linkers can be cleavable in vitro and in vivo. Cleavable linkers can include chemically or enzymatically unstable or degradable linkages. Cleavable linkers can rely on processes inside the cell to liberate a TGFβR1 inhibitor, such as reduction in the cytoplasm, exposure to acidic conditions in the lysosome, or cleavage by specific proteases or other enzymes within the cell. A cleavable linker can be sensitive to (i.e., cleavable by) enzymes at a specific site. A cleavable linker can be cleaved by enzymes such as proteases. Cleavable linkers can incorporate one or more chemical bonds that are either chemically or enzymatically cleavable while the remainder of the linker can be non-cleavable. In some embodiments, L is a linker comprising a reactive moiety. In some embodiments, for a linker-payload comprising a compound of the present disclosure, or pharmaceutically acceptable isomer, tautomer, racemate, hydrate, solvate, isotope, or salt thereof, −L is represented by the formula:
. In some embodiments, −L is represented by the formula:
, wherein each R30 is independently selected from optionally substituted C1-C6 alkyl and optionally substituted phenyl, and RX is the reactive moiety. RX may comprise a leaving group. RX may be a maleimide. L may be further covalently bound to a GalNAc moiety. In some embodiments, −L− is represented by the formula:
, wherein RX* is a bond, a succinimide moiety, or a hydrolyzed succinimide moiety bound to a GalNAc moiety, wherein
on RX* represents the point of attachment; and each R30 is independently selected from optionally substituted C1-C6 alkyl and optionally substituted phenyl. In some embodiments, for a linker-payload comprising a compound of the present disclosure, or pharmaceutically acceptable isomer, tautomer, racemate, hydrate, solvate, isotope, or salt thereof, and linker L; L comprises a methylene carbamate unit. A linker can contain a chemically labile group such as hydrazone and/or disulfide groups. Linkers comprising chemically labile groups can exploit differential properties between the plasma and some cytoplasmic compartments. The intracellular conditions that can facilitate TGFβR1 inhibitor release for hydrazone containing linkers can be the acidic environment of endosomes and lysosomes, while the disulfide containing linkers can be reduced in the cytosol, which can contain high thiol concentrations, e.g., glutathione. The plasma stability of a linker containing a chemically labile group can be increased by introducing steric hindrance using substituents near the chemically labile group. Acid-labile groups, such as hydrazone, can remain intact during systemic circulation in the blood’s neutral pH environment (pH 7.3-7.5) and can undergo hydrolysis once the conjugate is internalized into mildly acidic endosomal (pH 5.0-6.5) and lysosomal (pH 4.5-5.0)
compartments of the cell. This pH dependent release mechanism can be associated with nonspecific release of the drug. To increase the stability of the hydrazone group of the linker, the linker can be varied by chemical modification, e.g., substitution, allowing tuning to achieve more efficient release in the lysosome with a minimized loss in circulation. In some embodiments, for a linker-payload comprising a compound of the present disclosure, or pharmaceutically acceptable isomer, tautomer, racemate, hydrate, solvate, isotope, or salt thereof; and a linker L, −L comprises a hydrazone moiety. For example, L may be selected from:
wherein M is selected from C1-C6 alkyl, aryl, and −O−C1-C6 alkyl. Hydrazone-containing linkers can contain additional cleavage sites, such as additional acid-labile cleavage sites and/or enzymatically labile cleavage sites. Conjugates including exemplary hydrazone-containing linkers can include, for example, the following structures:
In certain linkers, such as linker (Ia), the linker can comprise two cleavable groups– a disulfide and a hydrazone moiety. For such linkers, effective cleavage can require acidic pH or disulfide reduction and acidic pH. Linkers such as (Ib) and (Ic) can be effective with a single hydrazone cleavage site.
Other acid-labile groups that can be included in linkers include cis-aconityl-containing linkers. cis-Aconityl chemistry can use a carboxylic acid juxtaposed to an amide bond to accelerate amide hydrolysis under acidic conditions. Cleavable linkers can also include a disulfide group. Disulfides can be thermodynamically stable at physiological pH and can be designed to release upon internalization inside cells, wherein the cytosol can provide a significantly more reducing environment compared to the extracellular environment. Scission of disulfide bonds can require the presence of a cytoplasmic thiol cofactor, such as (reduced) glutathione (GSH), such that disulfide-containing linkers can be reasonably stable in circulation, selectively releasing the TGFβR1 inhibitor in the cytosol. The intracellular enzyme protein disulfide isomerase, or similar enzymes capable of cleaving disulfide bonds, can also contribute to the preferential cleavage of disulfide bonds inside cells. GSH can be present in cells in the concentration range of 0.5-10 mM compared with a significantly lower concentration of GSH or cysteine, the most abundant low-molecular weight thiol, in circulation at approximately 5 μM. Tumor cells, where irregular blood flow can lead to a hypoxic state, can result in enhanced activity of reductive enzymes and therefore even higher glutathione concentrations. The in vivo stability of a disulfide-containing linker can be enhanced by chemical modification of the linker, e.g., use of steric hindrance adjacent to the disulfide bond. For example, L may be selected from:
, wherein M is selected from C1-C6 alkyl, aryl, and −O-C1-C6 alkyl. Exemplary cleavable linkers including disulfide moieties can include the following
wherein R is independently selected at each occurrence from hydrogen or alkyl, for example. Increasing steric hindrance adjacent to the disulfide bond can increase the stability of the linker. Structures such as (Id) and (If) can show increased in vivo stability when one or more R groups is selected from a lower alkyl such as methyl. Another type of cleavable linker is specifically cleaved by an enzyme. For example, the linker can be cleaved by a lysosomal enzyme. Such linkers can be peptide-based or can include peptidic regions that can act as substrates for enzymes. Peptide-based linkers can be more stable in plasma and extracellular milieu than chemically labile linkers. Peptide bonds can have good serum stability, as lysosomal proteolytic enzymes can have very low activity in blood due to endogenous inhibitors and the unfavorably high pH value of blood compared to lysosomes. Release of a TGFβR1 inhibitor from a conjugate can occur due to the action of lysosomal proteases, e.g., cathepsin and/or plasmin. These proteases can be present at elevated levels in certain tumor tissues. The linker can be cleavable by a lysosomal enzyme. The lysosomal enzyme can be, for example, cathepsin B, β-glucuronidase, or β-galactosidase. The cleavable peptide can be selected from tetrapeptides such as Gly-Phe-Leu-Gly, Ala- Leu-Ala-Leu or dipeptides such as Val-Cit, Val-Ala, and Phe-Lys. Dipeptides can have lower hydrophobicity compared to longer peptides. In some embodiments, the cleavable linker comprises a cleavable peptide. In some embodiments, the cleavable peptide is a dipeptide, tripeptide, or tetrapeptide. In some embodiments, the cleavable peptide is Val−Cit; Cit−Val; Ala−Ala; Ala−Cit; Cit−Ala; Asn−Cit; Cit−Asn; Cit−Cit; Val−Glu; Glu−Val; Ser−Cit; Cit−Ser; Lys−Cit; Cit−Lys; Asp−Cit; Cit−Asp; Ala−Val; Val−Ala; Phe−Lys; Lys−Phe; Val−Lys; Lys−Val; Ala−Lys; Lys−Ala; Phe−Cit; Cit−Phe; Leu− Cit; Cit−Leu; Ile−Cit; Cit−Ile; Phe−Arg; Arg−Phe; Cit−Trp; Trp−Cit; Ala−Ala−Asn; Gly−Phe−Leu−Gly; Gly−Gly−Phe−Gly; or Ala−Leu−Ala−Leu. In some embodiments, the cleavable linker comprises a structure of formula:
wherein −AA1−AA2− is the cleavable dipeptide and AA1 and AA2 are each an amino acid. In some embodiments, the cleavable dipeptide is Val−Cit.
Enzymatically cleavable linkers can include a self-immolative spacer to spatially separate the TGFβR1 inhibitor from the site of enzymatic cleavage. The direct attachment of a TGFβR1 inhibitor to a peptide linker can result in proteolytic release of an amino acid adduct of the TGFβR1 inhibitor, thereby impairing its activity. The use of a self-immolative spacer can allow for the elimination of the fully active, chemically unmodified TGFβR1 inhibitor upon amide bond hydrolysis. One self-immolative spacer can be a bifunctional para-aminobenzyl alcohol (PABA) group, which can link to the peptide through the amino group, forming an amide bond, while amine containing TGFβR1 inhibitors can be attached through carbamate functionalities to the benzylic hydroxyl group of the linker to form a para-aminobenzyl carbamate (PMBC) group. The resulting pro-TGFβR1 inhibitor can be activated upon protease-mediated cleavage, leading to a 1,6-elimination reaction releasing the unmodified TGFβR1 inhibitor, carbon dioxide, and remnants of the linker group. The following scheme depicts the fragmentation of a substituted p- aminobenzyl carbamate and release of the compound:
wherein HX−D represents the unmodified inhibitor. In some embodiments, a cleavable linker comprises a valine-citrulline peptide or a valine-alanine peptide. A valine-citrulline or valine-alanine-containing linker can contain a succimide group. A valine-citrulline- or valine-alanine-containing linker can contain a para- aminobenzyl (PAB) group. In some embodiments, the PAB group is a para-aminobenzyl alcohol (PABA) group or a para-aminobenzyl carbamate (PABC) group. In some embodiments, a valine-citrulline- or valine-alanine-containing linker also includes a PAB group, such as a PABC group. In some embodiments, a cleavable linker comprises a −(valine−citrulline)−(para−aminobenzyloxycarbonyl) group. In some embodiments, a cleavable linker comprises a lysine with an N-terminal amine acetylated, and a valine-citrulline cleavage site. In some embodiments, the cleavable linker comprises a succinimide group.
The enzymatically cleavable linker can be a ß-glucuronic acid-based linker. Facile release of the TGFβR1 inhibitor can be realized through cleavage of the ß-glucuronide glycosidic bond by the lysosomal enzyme ß-glucuronidase. This enzyme can be abundantly present within lysosomes and can be overexpressed in some tumor types, while the enzyme activity outside cells can be low. ß-Glucuronic acid-based linkers can be used to circumvent the tendency of a conjugate to undergo aggregation due to the hydrophilic nature of ß-glucuronides. In certain embodiments, ß-glucuronic acid-based linkers can link the GalNAc moiety to a hydrophobic TGFβR1 inhibitor. A variety of cleavable β-glucuronic acid-based linkers useful for linking drugs such as auristatins, camptothecin and doxorubicin analogues, CBI minor-groove binders, and psymberin to antibodies have been described, and such linkers are useful for the present conjugates as well. All of these β-glucuronic acid-based linkers may be used in the conjugates comprising a TGFβR1 inhibitor described herein. In certain embodiments, the enzymatically cleavable linker is a β-galactoside-based linker. β-Galactoside is present abundantly within lysosomes, while the enzyme activity outside cells is low.Additionally, cleavable linkers may comprise a phenol and connection through the phenolic oxygen. One such linker employs diamino-ethane unit (e.g., “Space Link”) in conjunction with traditional “PABO”-based self-immolative groups to deliver a phenol. Inhibitors containing an aromatic or aliphatic hydroxyl group can be covalently bonded to a linker through the hydroxyl group using a methodology that relies on a methylene carbamate linkage, as described in WO 2015/095755. Cleavable linkers can include non-cleavable portions or segments, and/or cleavable segments or portions can be included in an otherwise non-cleavable linker to render it cleavable. By way of example only, polyethylene glycol (PEG) and related polymers can include cleavable groups in the polymer backbone. For example, a polyethylene glycol or polymer linker can include one or more cleavable groups such as a disulfide, a hydrazone or a dipeptide. Other degradable linkages that can be included in cleavable linkers include ester linkages formed by the reaction of PEG carboxylic acids or activated PEG carboxylic acids with alcohol groups on a TGFβR1 inhibitor, wherein such ester groups can hydrolyze under physiological conditions to release the TGFβR1 inhibitor. Hydrolytically degradable linkages can include, but are not limited to, carbonate linkages; imine linkages resulting from reaction of an amine and an
aldehyde; phosphate ester linkages formed by reacting an alcohol with a phosphate group; acetal linkages that are the reaction product of an aldehyde and an alcohol; orthoester linkages that are the reaction product of a formate and an alcohol; and oligonucleotide linkages formed by a phosphoramidite group, including but not limited to, at the end of a polymer, and a 5’ hydroxyl group of an oligonucleotide. A linker can be a link created by a microbial transglutaminase, wherein the link can be created between an amine-containing moiety and a moiety engineered to contain glutamine as a result of the enzyme catalyzing a bond formation between the acyl group of a glutamine side chain and the primary amine of a lysine chain. A linker can contain a reactive primary amine. A linker can be a Sortase A linker. A Sortase A linker can be created by a Sortase A enzyme fusing an LPXTG (SEQ ID NO:1) recognition motif to an N-terminal GGG motif to regenerate a native amide bond. The linker created can therefore link a moiety attached to the LPXTG (SEQ ID NO:1) recognition motif with a moiety attached to the N-terminal GGG motif. A linker can be a link created between an unnatural amino acid on one moiety reacting with oxime bond that was formed by modifying a ketone group with an alkoxyamine on another moiety. A moiety can be a TGFβR1 inhibitor. Although cleavable linkers can provide certain advantages, the linkers in the conjugates described herein need not include cleavable linkers. For non-cleavable linkers, the TGFβR1 inhibitor release may not depend on the differential properties between the plasma and some cytoplasmic compartments. A non-cleavable linker can be protease insensitive. A non-cleavable linker can contain a succinimide group. A non-cleavable linker can contain a caproyl group or a succinimidocaproyl group. A succinimidocaproyl linker can comprise N-succinimidomethylcyclohexane-1- carboxylate. A non-cleavable linker can be a combination of a succinimidocaproyl group and one or more ethylene glycol units. A non-cleavable linker can be a PEG4 linker or a succinimido-PEG4 linker. A non-cleavable linker can be a combination of a succinimidocaproyl linker containing a succinimide group and one or more ethylene glycol units. A non-cleavable linker can contain one or more succinimido groups linked to polyethylene glycol units in which the polyethylene glycol can allow for more linker flexibility or can be used lengthen the linker.
In some embodiments, a non-cleavable linker independently comprises a heteroalkylene, heteroalkenylene, or heteroalkynylene comprising 3 to 30 components in the longest linear chain, wherein the components are selected from −CH2−, −CH(C1-4alkyl), −C(C1-4alkyl)2, −CH=CH−, −C≡C−, −C(O)−, −O−, −NH−, −N(C1-4alkyl), −S−, −S(O)−, −S(O)2−, and −P(O)(O-)−. In some embodiments, a non-cleavable linker independently comprises a heteroalkylene comprising 3 to 15 components in the longest linear chain, wherein the components are selected from −CH2−, −CH(C1-4alkyl), −C(O)−, −O−, −NH−, and −N(C1-4alkyl). In some embodiments, the non-cleavable linker comprises a polyethylene glycol region comprising two to six ethylene glycol units. In some embodiments, a non-cleavable linker is −NH−C(O)−(CH2)2-5−C(O)−, −NH−C(O)−(CH2)2-5−C(O)NH−(CH2)2-7−C(O)−, or −NH−C(O)−(CH2)2-5−C(O)NH−(CH2)2-7−NH−C(O)−(CH2)2-4−C(O)−. In some embodiments, a non-cleavable linker is −NH−C(O)−(CH2)3−C(O)−. Attachment groups that are used to attach the linkers in a conjugate can be electrophilic in nature and include, for example, active esters such as NHS esters and HOBt esters, haloformates, acid halides, carboxylic acids, amines, alkyl halides, benzyl halides such as haloacetamides, maleimide groups, or activated disulfides. In some embodiments, Region L3 comprises or is the following structure:
, wherein: L4 represents the C-terminus of the peptide; L5 is selected from a bond, alkylene, and heteroalkylene, wherein L5 is optionally substituted with one or more groups independently selected from R32; R32 is independently selected at each occurrence from halogen, −OH, −CN, −O−alkyl, −SH, =O, =S, −NH2, −NO2; and C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, each of which is optionally substituted with one or more substituents independently selected from halogen, −OH, −CN, −O−alkyl, −SH, =O, =S, −NH2, −NO2; and RX* is a bond. In some embodiments, the peptide comprises Val—Cit or Val—Ala.
In any of the aforementioned embodiments, −L3 is:
A linker can comprise a cleavable peptide, for example, a linker comprising structural formula (IVa), (IVb), (IVc), or (IVd):
or a salt thereof, wherein: peptide represents a cleavable peptide (illustrated N→C, wherein peptide includes the amino and carboxy “termini”) as described herein; T represents a polymer comprising one or more ethylene glycol units or an alkylene chain, or combinations thereof; Ra is selected from hydrogen, alkyl, sulfonate and methyl sulfonate; Ry is hydrogen or C1-4 alkyl−(O)r−(C1-4 alkylene)s−G1 or C1-4 alkyl−(N)−[(C1-4 alkylene)−G1]2; Rz is C1-4 alkyl−(O)r−(C1-4 alkylene)s−G2; G1 is SO3H, CO2H, PEG 4-32, or sugar moiety; G2 is SO3H, CO2H, or PEG 4-32 moiety; r is 0 or 1; s is 0 or 1; p is an integer ranging from 0 to 5; q is 0 or 1; x is 0 or 1; y is 0 or 1; represents one point of attachment of the linker to the inhibitor; and * represents the point of attachment to the GalNAc moiety. Exemplary polyvalent linkers that may be used to link inhibitors include Fleximer® linker technology that has the potential to enable high loading of conjugates with good physicochemical properties. The Fleximer® linker technology is based on incorporating drug molecules into a solubilizing poly-acetal backbone via a sequence of ester bonds.
The methodology renders highly-loaded conjugates (drug loading up to 20) while maintaining good physicochemical properties. In certain embodiments, to utilize the Fleximer® linker technology depicted above, an aliphatic alcohol can be present or introduced into an inhibitor or salt thereof (e.g., an ALK5 inhibitor, as described herein). The alcohol moiety is then conjugated to an alanine moiety, which is then synthetically incorporated into the Fleximer® linker. Liposomal processing of the conjugate in vitro releases the parent alcohol-containing drug. Sulfamide linkers may also be used in the disclosed conjugates. Sulfamide linkers are as described herein and e.g., U.S. Patent Publication No. 2019/0038765, the linkers of which are incorporated by reference herein. Exemplary embodiments of linkers according to structural formula (IVa) that can be included in the conjugates described herein can include the structures illustrated below:
. wherein one of represents the point of attachment of the linker (L3) to the inhibitor. Exemplary embodiments of linkers according to structural formula (IVb), (IVc), and (IVd) that can be included in the conjugates described herein can include the structures illustrated below:
.
wherein one of represents the point of attachment of the linker (L3) to the inhibitor. Exemplary embodiments of linkers according to structural formula (IVa) that can be included in the conjugates described herein can include the structures illustrated below:
wherein one of represents the point of attachment of the linker (L3) to the inhibitor.
The cleavable linker can contain an enzymatically cleavable sugar moiety, for example, a linker comprising structural formula (Va), (Vb), (Vc), (Vd), or (Ve):
or a salt thereof, wherein: q is 0 or 1; r is 0 or 1; X1 is CH2, O or NH;
represents the point of attachment of the linker to the TGFβR1 inhibitor; and * represents the point of attachment to the remainder of the conjugate. Exemplary embodiments of linkers according to structural formula (IVa) that may be included in the conjugates described herein can include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the TGFβR1 inhibitor:
, Exemplary embodiments of linkers according to structural formula (IVb) that may be included in the conjugates described herein can include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the TGFβR1 inhibitor:
. Exemplary embodiments of linkers according to structural formula (IVc) that may be included in the conjugates described herein can include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the TGFβR1 inhibitor:
. Exemplary embodiments of linkers according to structural formula (IVd) that may be included in the conjugates described herein can include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the TGFβR1 inhibitor:
. Exemplary embodiments of linkers according to structural formula (IVe) that may be included in the conjugates described herein can include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the TGFβR1 inhibitor:
. The linker can be non-cleavable in vivo, for example, a linker according to the formulations below:
or salts thereof, wherein: Ra is selected from hydrogen, alkyl, sulfonate and methyl sulfonate; Rx is a moiety that covalently links the linker to the rest of the conjugate, such as a bond; and represents the point of attachment of the linker to the rest of the conjugate. Exemplary embodiments of linkers according to structural formula (Va)-(Vf) that may be included in the conjugates described herein include the linkers illustrated:
. Attachment groups that are used to attach the linkers to a TGFβR1 inhibitor can be electrophilic in nature and include, for example, maleimide groups, alkynes, alkynoates, allenes and allenoates, activated disulfides, active esters such as NHS esters and HOBt esters, haloformates, acid halides, alkyl, and benzyl halides such as haloacetamides. There are also emerging technologies related to "self-stabilizing" maleimides and "bridging disulfides" that can be used in accordance with the disclosure. A linker described herein with a maleimide group may include an electron withdrawing group such as, but not limited to, -C(O)R, =O, -CN, -NO2, -CX3, -X, -COOR, -CONR2, -COR, - COX, -SO2R, -SO2OR, -SO2NHR, -SO2NR2, PO3R2, -P(O)(CH3)NHR, -NO, -NR3 +, -CR=CR2, and -C≡CR, where each R is independently selected from H and C1-6 alkyl and each X is independently selected from F, Br, Cl, and I. Self-stabilizing linkers may also include aryl, e.g., phenyl, or heteroaryl, e.g., pyridine, groups optionally substituted with electron withdrawing groups such as those described herein. Examples of self-stabilizing linkers are provided in, e.g., U.S. Patent Publication No. US 2013/0309256, the linkers of which are incorporated by reference herein. It will be understood that a self-stabilizing linker useful in conjunction with immune-stimulatory compounds may be equivalently described as unsubstituted maleimide-including linkers, thio- substituted succinimide-including linkers, or hydrolyzed, ring-opened thio-substituted succinimide-including linkers. In certain embodiments, a linker comprises a stabilizing linker moiety selected from:
. In the scheme provided above, the bottom structure may be referred to as (maleimido)- DPR-Val-Cit-PAB, where DPR refers to diaminopropinoic acid, Val refers to valine, Cit refers to citrulline, and PAB refers to para-aminobenzylcarbonyl, wherein represents the point of attachment of the linker to a nitrogen of the inhibitor. A method for bridging a pair of sulfhydryl groups derived from reduction of a native hinge disulfide bond has been disclosed and is depicted in the schematic below. An advantage of this methodology is the ability to synthesize homogenous DAR4 conjugates by full reduction of IgGs (to give 4 pairs of sulfhydryls from interchain disulfides) followed by reaction with 4 equivalents of the alkylating agent. Conjugates containing "bridged disulfides" are also claimed to have increased stability.
Similarly, as depicted below, a maleimide derivative that is capable of bridging a pair of sulfhydryl groups has been developed.
A linker can contain the following structural formulas (VIa), (VIb), or (VIc):
or salts thereof, wherein: Rq is H or–O-(CH2CH2O)11-CH3; x is 0 or 1; y is 0 or 1; G2 is -CH2CH2CH2SO3H or–CH2CH2O-(CH2CH2O)11-CH3; Rw is–O-CH2CH2SO3H or -NH(CO)-CH2CH2O-(CH2CH2O)12-CH3; and * represents the point of attachment to the remainder of the linker.
Exemplary embodiments of linkers according to structural formula (VIa) and (VIb) that can be included in the conjugates described herein can include the linkers illustrated:
, wherein represents the point of attachment of the linker to a nitrogen of the inhibitor. Exemplary embodiments of linkers according to structural formula (Vic) that can be included in the immune-stimulatory conjugates described herein can include the linkers illustrated below:
wherein represents the point of attachment of the linker to a nitrogen of the inhibitor. Some exemplary linkers (L3) are described in the following paragraphs. In some embodiments for an inhibitor wherein attachment of the linker is to a nitrogen of the inhibitor, −L3 is represented by the formulas set forth in Table 1 below: TABLE 1
wherein
represents attachment to a nitrogen of a TGFβR1 inhibitor; L4 represents the C-terminus of the peptide and L5 is selected from a bond, alkylene and heteroalkylene, wherein L5 is optionally substituted with one or more groups independently selected from R30, and R30 is independently selected at each occurrence from halogen, −OH, −CN, −O−alkyl, −SH, =O, =S, −S(O)2OH, −NH2, −NO2; and C1−C10alkyl, C2-C10alkenyl, and C2-C10alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, −OH, −CN, −O−alkyl, −SH, =O, =S, −S(O)2OH, −NH2, and −NO2; and RX represents a reactive moiety. The reactive moiety may be selected, for example, from an electrophile, e.g., an αǡ ^-unsaturated carbonyl, such as a maleimide, and a leaving group. For example, –L can be represented by the formulas set forth in Table 2 below: TABLE 2
wherein
represents attachment to a nitrogen of a TGFβR1 inhibitor and L4 represents the C-terminus of the peptide and L5 is selected from a bond, alkylene and heteroalkylene, wherein L5 is optionally substituted with one or more groups independently selected from R30, and R30 is independently selected at each occurrence from halogen, −OH, −CN, −O−alkyl, −SH, =O, =S, −NH2, −NO2; and C1-C10alkyl, C2-C10alkenyl, and C2-C10alkynyl,
each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, −OH, −CN, −O−alkyl, −SH, =O, =S, −NH2, and −NO2. When conjugated to the GalNAc moiety, such linkers can be, for example, represented by the Formulas set forth in Table 3 below: TABLE 3
where represents attachment to a nitrogen of a TGFβR1 inhibitor and −RX* represents the point of attachment to the GalNAc moiety; L4 represents the C-terminus of the peptide and L5 is selected from a bond, alkylene and heteroalkylene, wherein L5 is optionally substituted with one or more groups independently selected from R30, and R30 when present is independently selected at each occurrence from halogen, −OH, −CN, −O−alkyl, −SH, =O, =S, −S(O)2OH, −NH2, −NO2; and C1-C10alkyl, C2-C10alkenyl, and C2-C10alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, −OH, −CN, −O−alkyl, −SH, =O, =S, −S(O)2OH, −NH2, and −NO2; and RX represents the point of attachment to the GalNAc moiety.
In some embodiments, –L3 is represented by the formulas set forth in Table 4 below: TABLE 4
wherein
represents attachment to a nitrogen of a TGFβR1 inhibitor and RX represents a reactive moiety. The reactive moiety may be selected from, for example, a leaving group. For example, –L can be represented by the formulas set forth in Table 5 below:
TABLE 5
The linkers, can, for example, be represented by the Formulas set forth in Table 6 below wherein RX* is a bond to the GalNAc moiety, wherein
on RX* represents the point of attachment to such residue: TABLE 6
represents the point of attachment to the GalNAc moiety. As is known by skilled artisans, the linker selected for a particular conjugate may be influenced by a variety of factors, including the site of attachment to GalNAc moiety structural constraints of the drug pharmacophore, and the lipophilicity of the drug. The specific linker selected for a conjugate should seek to balance these different factors. For example, cytotoxic conjugates have been observed to effect killing of bystander antigen-negative cells present in the vicinity of the antigen-positive tumor cells. The mechanism of the bystander effect by cytotoxic conjugates has indicated that metabolic products formed during intracellular processing of the conjugates may play a role. Neutral cytotoxic metabolites generated by metabolism of the conjugates in antigen-positive cells appear to play a role in bystander cell killing while charged metabolites may be prevented from diffusing across the membrane into the medium, or from the medium across the membrane and, therefore, cannot effect cell killing via the bystander effect. In some embodiments, a linker is selected to attenuate
the bystander effect caused by cellular metabolites of the conjugate. In further embodiments, a linker is selected to increase the bystander effect. The properties of the linker, or linker-payload, may also impact aggregation of a conjugate under conditions of use and/or storage. Conjugates reported in the literature contain about 3-4 drug molecules per antibody molecule. Attempts to obtain higher drug-to-antibody ratios (“DAR”) often failed, particularly if both the drug and the linker were hydrophobic, due to aggregation of the conjugate. In many instances, DARs higher than 3-4 could be beneficial as a means of increasing potency. In instances where the payload compound is more hydrophobic in nature, it may be desirable to select linkers that are relatively hydrophilic as a means of reducing conjugate aggregation, especially in instances where DARs greater than 3-4 are desired. Thus, in some embodiments, a linker incorporates chemical moieties that reduce aggregation of the conjugates during storage and/or use. A linker may incorporate polar or hydrophilic groups such as charged groups or groups that become charged under physiological pH to reduce the aggregation of the conjugates. For example, a linker may incorporate charged groups such as salts or groups that deprotonate, e.g., carboxylates, or protonate, e.g., amines, at physiological pH. In preferred embodiments, aggregation of conjugates during storage or use is less than about 40% as determined by size-exclusion chromatography (SEC). In particular embodiments, the aggregation of the conjugates during storage or use is less than about 35%, such as less than about 30%, about 25%, about 20%, about 15%, about 10%, about 5%, about 4%, or even less, as determined by size-exclusion chromatography (SEC). Some exemplary linkers (L) are described in the following paragraphs. In some embodiments for a compound wherein attachment of the linker is to a nitrogen of the of a TGFβR1 inhibitor and −RX represents a reactive group for attachment to the GalNAc moiety. –L is represented by the formulas set forth in Table 7 below:
wherein
represents attachment to a nitrogen of a TGFβR1 inhibitor. L4 represents the C-terminus of the peptide and L5 is selected from a bond, alkylene and heteroalkylene, wherein L5 is optionally substituted with one or more groups independently selected from R30, and R30 is independently selected at each occurrence from halogen, −OH, −CN, −O−alkyl, −SH, =O, =S, −S(O)2OH, −NH2, −NO2; and C1-C10alkyl, C2-C10alkenyl, and C2-C10alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, −OH, −CN, −O−alkyl, −SH, =O, =S, −S(O)2OH, −NH2, and −NO2; and RX represents a reactive moiety. The reactive moiety may be selected, for example, from an electrophile, e.g., an α,β-unsaturated carbonyl, such as a maleimide, and a leaving group. For example, –L3 can be represented by the formulas set forth in Table 8 below: TABLE 8
wherein
represents attachment to a nitrogen of a TGFβR1 inhibitor and L4 represents the C-terminus of the peptide and L5 is selected from a bond, alkylene and
heteroalkylene, wherein L5 is optionally substituted with one or more groups independently selected from R30, and R30 is independently selected at each occurrence from halogen, −OH, −CN, −O−alkyl, −SH, =O, =S, −S(O)2OH, −NH2, −NO2; and C1-C10alkyl, C2-C10alkenyl, and C2-C10alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, −OH, −CN, −O−alkyl, −SH, =O, =S, −S(O)2OH, −NH2, and −NO2. Additional linkers can be, for example, represented by the Formulas set forth in Table 9 below: TABLE 9
wherein RX* is a bond, a succinimide moiety, or a hydrolyzed succinimide moiety, wherein on RX* represents the point of attachment to such residue; L4 when present represents the C-terminus of the peptide and L5 is selected from a bond, alkylene and heteroalkylene, wherein L5 is optionally substituted with one or more groups independently selected from R30; and R30 when present is independently selected at each occurrence from halogen, −OH, −CN, −O−alkyl, −SH, =O, =S, −S(O)2OH, −NH2, −NO2; and C1-C10alkyl, C2-C10alkenyl, and C2-C10alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, −OH, −CN, −O−alkyl, −SH, =O, =S, −S(O)2OH, −NH2, and −NO2. A particularly preferred pepide is val-ala or val-cit. In some embodiments wherein attachment of the linker is to a nitrogen of a TGFβR1 inhibitor and −RX represents the reactive moiety for the point of attachment to the GalNAc moiety, –L3 is represented by the formulas set forth in Table 10 below: TABLE 10
The reactive moiety may be selected from, for example, a leaving group. For example, – L3 can be represented by the formulas set forth in Table 11 below: TABLE 11
L3 can be represented by the Formulas set forth in Table 12 below wherein RX* is a bond to the GalNAc moiety, wherein on RX* represents the point of attachment to such residue:
TABLE 12
As noted,
represents attachment to a nitrogen of a TGFβR1 inhibitor and −RX* represents the point of attachment to the GalNAc moiety. In exemplary embodiments, the linkers described herein, including those in the preceding paragraphs, are attached to a compound of the present invention through the nitrogen of a secondary acyclic amine depicted in the structure of any one the compounds of the present invention. In exemplary embodiments, the linkers described herein, including those in the preceding paragraphs, are attached to a compound of the present invention at a nitrogen atom. As is known by skilled artisans, the linker selected for a particular conjugate may be influenced by a variety of factors, including but not limited to, the site of attachment to the
antibody construct (e.g., lys, cys or other amino acid residues), structural constraints of the drug pharmacophore and the lipophilicity of the drug. The specific linker selected for a conjugate should seek to balance these different factors for the specific GalNAc construct/drug combination. In some aspects, the conjugate comprises one of the following structures: ,
or a salt thereof. In some aspects, the conjugate comprises a structure as shown in Table 13:
TABLE 13
In some aspects, the conjugate comprises one of the following structures, where the wavy bond is the point of attachment to the rest of the conjugate:
. In some embodiments, Region L3 comprises a dipeptide. In some embodiments, the dipeptide is Val−Cit. In some embodiments, Region L3 comprises a para-aminobenzyl moiety. In some embodiments, Region L3 comprises a PABC moiety. In some embodiments, Region L3 comprises −NHC(O)−(CH2)p−C(O)− where p is 2, 3, 4, 5, or 6. In some embodiments, Region L3 is −NHC(O)−(CH2)p−C(O)−Val−Cit−PAB−O−C(O)−. A TGFβR1 inhibitor-linker compound can be synthesized by various methods known in the art. See, for example, WO2018170179. As is known by skilled artisans, the linker selected for a particular conjugate may be influenced by a variety of factors, including but not limited to, the site of attachment to the GalNAc moiety, structural constraints of the drug pharmacophore, and the lipophilicity of the drug. The specific linker selected for a conjugate should seek to balance these different factors for the specific conjugate.
Additional Inhibitors In some aspects, the disclosure includes a TGFβR1 inhibitor of Formula (XX): [Inh]−Z (XX) wherein Region Inh comprises a TGFβR1 inhibitor (e.g., a compound of Formula (A-I), (B-I), or (C-1); and Z is the residual portion of a released, cleavable linker or comprises a non-cleavable linker and a residual portion of a degraded Region G; or a salt thereof. In such aspects, Z is a moiety that remains covalently attached to the rest of the inhibitor structure following cleavage (e.g., in vivo) of a cleavable linker of a conjugate as described herein. In some embodiments, Z comprises an amino acid. In some embodiments, Z comprises valine or citrulline. In some embodiments, Z is −NH2. In some embodiments, Z is a moiety that remains covalently attached to the inhibitor following degradation (e.g., in vivo) of Region G of a conjugate as described herein. In some embodiments, Z comprises a degraded Region G and a non-cleavable linker as described herein. In some embodiments, the degradation is by enzyme- or acid-mediated degradation, such as hydrolysis. In some embodiments, a degraded Region G comprises one or more degraded GalNAc groups. Pharmaceutical Formulations The conjugates described herein are useful as pharmaceutical compositions for administration to a subject in need thereof. Pharmaceutical compositions can comprise the conjugates described herein and one or more pharmaceutically acceptable carriers, diluents, excipients, stabilizers, dispersing agents, suspending agents, and/or thickening agents. A pharmaceutical composition can comprise any conjugate described herein. A pharmaceutical composition can further comprise buffers, antibiotics, steroids, carbohydrates, drugs (e.g., chemotherapy drugs), radiation, polypeptides, chelators, adjuvants and/or preservatives. In a pharmaceutical composition, conjugates for which a is 1 comprise a Region Fc. These conjugates can have an average drug loading. The average drug loading is the average
number of TGFβR1 inhibitor molecules per Region Fc. The average number of TGFβR1 inhibitors per Region Fc in a preparation may be characterized by conventional means such as mass spectroscopy, HIC, ELISA assay, and HPLC. In some embodiments, the average drug loading is 1 to 8, or 1 to 6, or 1 to 4, or 1 to 3, or 1 to 2, or 2 to 4, or 2 to 3, TGFβR1 inhibitor molecules per Region Fc. Pharmaceutical compositions can be formulated using one or more physiologically- acceptable carriers comprising excipients and auxiliaries. Formulation can be modified depending upon the route of administration chosen. Pharmaceutical compositions comprising a conjugate as described herein can be manufactured, for example, by lyophilizing the conjugate, mixing, dissolving, emulsifying, encapsulating or entrapping the conjugate. The pharmaceutical compositions can also include the conjugates described herein in a free-base form or pharmaceutically-acceptable salt form. Methods for formulation of the pharmaceutical compositions can include formulating any of the conjugates described herein with one or more inert, pharmaceutically-acceptable excipients or carriers to form a solid, semi-solid, or liquid composition. Solid compositions can include, for example, powders, tablets, dispersible granules and capsules, and in some aspects, the solid compositions further contain nontoxic, auxiliary substances, for example wetting or emulsifying agents, pH buffering agents, and other pharmaceutically-acceptable additives. Alternatively, the compositions described herein can be lyophilized or in powder form for re- constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use Pharmaceutical compositions of the conjugates described herein can comprise at least a conjugate as an active ingredient, respectively. The active ingredients can be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization (e.g., hydroxymethylcellulose or gelatin microcapsules and poly- (methylmethacylate) microcapsules, respectively), in colloidal drug-delivery systems (e.g., liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Pharmaceutical compositions as described herein often further can comprise more than one active compound as necessary for the particular indication being treated. The active compounds can have complementary activities that do not adversely affect each other. For
example, the pharmaceutical composition can also comprise a cytotoxic agent, cytokine, growth- inhibitory agent, anti-hormonal agent, anti-angiogenic agent, and/or cardioprotectant. Such molecules can be present in combination in amounts that are effective for the purpose intended. The pharmaceutical compositions and formulations can be sterilized. Sterilization can be accomplished by filtration through sterile filtration. The pharmaceutical compositions described herein can be formulated for administration as an injection. Non-limiting examples of formulations for injection can include a sterile suspension, solution or emulsion in oily or aqueous vehicles. Suitable oily vehicles can include, but are not limited to, lipophilic solvents or vehicles such as fatty oils or synthetic fatty acid esters, or liposomes. Aqueous injection suspensions can contain substances which increase the viscosity of the suspension. The suspension can also contain suitable stabilizers. Injections can be formulated for bolus injection or continuous infusion. Alternatively, the pharmaceutical compositions described herein can be lyophilized or in powder form for reconstitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. For parenteral administration, the conjugates can be formulated in a unit dosage injectable form (e.g., use letter solution, suspension, emulsion) in association with a pharmaceutically acceptable parenteral vehicle. Such vehicles can be inherently nontoxic, and non-therapeutic. A vehicle can be water, saline, Ringer’s solution, dextrose solution, and 5% human serum albumin. Nonaqueous vehicles such as fixed oils and ethyl oleate can also be used. Liposomes can be used as carriers. The vehicle can contain minor amounts of additives such as substances that enhance isotonicity and chemical stability (e.g., buffers and preservatives). Sustained-release preparations can also be prepared. Examples of sustained-release matrices can include polyesters, hydrogels (e.g., poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides, copolymers of L-glutamic acid and γ ethyl-L-glutamate, non- degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTM (i.e., injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-( – )-3-hydroxybutyric acid. Pharmaceutical formulations of the compositions described herein can be prepared for storage by mixing a conjugate with a pharmaceutically acceptable carrier, excipient, and/or a stabilizer. This formulation can be a lyophilized formulation or an aqueous solution. Acceptable
carriers, excipients, and/or stabilizers can be nontoxic to recipients at the dosages and concentrations used. Acceptable carriers, excipients, and/or stabilizers can include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives, polypeptides; proteins, such as serum albumin or gelatin; hydrophilic polymers; amino acids; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes; and/or non- ionic surfactants or polyethylene glycol.
The conjugates, pharmaceutical compositions, and methods of the present disclosure can be useful for treating a subject such as, but not limited to, a mammal, a human, a non-human mammal, a domesticated animal (e.g., laboratory animals, household pets, or livestock), a non- domesticated animal (e.g., wildlife), a dog, a cat, a rodent, a mouse, a hamster, a cow, a bird, a chicken, a fish, a pig, a horse, a goat, a sheep, or a rabbit. In preferred embodiments, conjugates, pharmaceutical compositions, and methods of the present disclosure are used for treating a human. The conjugates, pharmaceutical compositions, and methods described herein can be useful as a therapeutic, for example a treatment that can be administered to a subject in need thereof. A therapeutic effect can be obtained in a subject by reduction, suppression, remission, or eradication of a disease state, including, but not limited to, a symptom thereof. A therapeutic effect in a subject having a disease or condition, or pre-disposed to have or is beginning to have the disease or condition, can be obtained by a reduction, a suppression, a prevention, a remission, or an eradication of the condition or disease, or pre-condition or pre-disease state. In practicing the methods described herein, therapeutically-effective amounts of the conjugates, or pharmaceutical compositions described herein can be administered to a subject in need thereof, often for treating and/or preventing a condition or progression thereof. A pharmaceutical composition can affect the physiology of the subject, such as the immune system, inflammatory response, or other physiologic affect. A therapeutically-effective amount can vary
widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compounds used, and other factors. Treat and/or treating can refer to any indicia of success in the treatment or amelioration of the disease or condition. Treating can include, for example, reducing, delaying or alleviating the severity of one or more symptoms of the disease or condition, or it can include reducing the frequency with which symptoms of a disease, defect, disorder, or adverse condition, and the like, are experienced by a patient. Treat can be used herein to refer to a method that results in some level of treatment or amelioration of the disease or condition, and can contemplate a range of results directed to that end, including but not restricted to prevention of the condition entirely. Prevent, preventing and the like can refer to the prevention of the disease or condition, e.g., viral infection, in the patient. For example, if an individual at risk of contracting a viral infection is treated with the methods of the present disclosure and does not later become infected with the virus, then the disease has been prevented, at least over a period of time, in that individual. A therapeutically effective amount can be the amount of conjugates or pharmaceutical compositions or an active component thereof sufficient to provide a beneficial effect or to otherwise reduce a detrimental non-beneficial event to the individual to whom the composition is administered. A therapeutically effective dose can be a dose that produces one or more desired or desirable (e.g., beneficial) effects for which it is administered, such administration occurring one or more times over a given period of time. An exact dose can depend on the purpose of the treatment, and can be ascertainable by one skilled in the art using known techniques. The conjugates or pharmaceutical compositions described herein that can be used in therapy can be formulated and dosages established in a fashion consistent with good medical practice taking into account the disorder to be treated, the condition of the individual patient, the site of delivery of the conjugate, or pharmaceutical composition, the method of administration and other factors known to practitioners. The conjugates or pharmaceutical compositions can be prepared according to the description of preparation described herein. One of ordinary skill in the art would understand that the amount, duration and frequency of administration of a pharmaceutical composition or conjugate described herein to a subject in need thereof depends on several factors including, for example but not limited to, the health of
the subject, the specific disease or condition of the patient, the grade or level of a specific disease or condition of the patient, the additional therapeutics the subject is being or has been administered, and the like. The methods, conjugates and pharmaceutical compositions described herein can be for administration to a subject in need thereof. Often, administration of the conjugates, or pharmaceutical compositions can include routes of administration, non-limiting examples of administration routes include intravenous, intraarterial, subcutaneous, subdural, intramuscular, intracranial, intrasternal, intratumoral, or intraperitoneally. Additionally, a pharmaceutical composition, or conjugate can be administered to a subject by additional routes of administration, for example, by inhalation, oral, dermal, intranasal, or intrathecal administration. Pharmaceutical compositions or conjugates of the present disclosure can be administered to a subject in need thereof in a first administration, and in one or more additional administrations. The one or more additional administrations can be administered to the subject in need thereof minutes, hours, days, weeks or months following the first administration. Any one of the additional administrations can be administered to the subject in need thereof less than 21 days, or less than 14 days, less than 10 days, less than 7 days, less than 4 days or less than 1 day after the first administration. The one or more administrations can occur more than once per day, more than once per week or more than once per month. The conjugates or pharmaceutical compositions can be administered to the subject in need thereof in cycles of 21 days, 14 days, 10 days, 7 days, 4 days or daily over a period of one to seven days. In one aspect, the disclosure relates to a method for treating a disease mediated by TGFβR1 activity, comprising administering an effective amount of a conjugate or a pharmaceutical composition as described herein to a subject in need thereof. In some aspects, the disease is cancer. In certain embodiments, the cancer is hepatocellular carcinoma (HCC). In further embodiments, the cancer is metastatic liver cancer from colon, lung, breast, neuroendocrine, stomach or pancreatic cancer. In some aspects the disease is liver fibrosis. In some aspects, the fibrosis is liver fibrosis. In some aspects, the fibrosis is cancer-associated. In some aspects, the fibrosis is not cancer- associated. In some aspects, the fibrosis is associated with scleroderma, systemic fibrosis,
steatohepatitis, or non-alcoholic steatohepatitis (NASH) , chronic liver viral disease (such as HBV and HCV infection), autoimmune hepatitis, or primary biliary cholangitis. In certain embodiments, the disease is a liver metabolic disease, such as steatosis. In further embodiments, treatment with conjugates or pharmaceutical compositions of the present disclosure reduces hepatocyte apoptosis or reduces altered lipid metabolism in subject consuming a fatty-diet. In one aspect is a method of treating a subject having liver cancer, such as hepatocellular carcinoma (HCC), comprising administering to the subject an effective amount of a conjugate as described herein or a pharmaceutical composition comprising the conjugate. In certain embodiments, the presently described conjugates or pharmaceutical compositions can be used to decrease HCC tumor cell growth and invasiveness. In further embodiments, the presently described conjugates or pharmaceutical compositions can be utilized to enhance liver-localized immune responses against liver cancer, such as HCC. In some embodiments, the administering is in a regimen that comprises administering the conjugates or pharmaceutical compositions intravenously or subcutaneouly. In certain embodiments, the dose level and schedule are chosen to maintain a high ratio of liver exposure to systemic exposure. In further embodiments, the dose and schedule are chosen to attain about an IC90, IC80, IC50 or IC30 of phospho-SMAD2/3 levels in hepatocytes as compared to non- conjugated treatment for at least 24 hours. In still furthre embodiments, the dose and schedule are chosen to attain about an IC90, IC80, IC50 or IC30 of non-hepatocyte liver cells compared to non-conjugate treatment for 24 hours. Increased dosages and reduced side-effects In certain embodiments, using a conjugate of this disclosure can allow administration of the conjugate at greater levels of the inhibitor in the form of the conjugate than the level of inhibitor alone. For example, the conjugate can be administered at a level higher than the maximum tolerated dose for that inhibitor administered in the absence of the being conjugated to the antibody construct in the conjugate. In certain embodiments, administration of the conjugate can be associated with fewer side effects than when administered as the inhibitor alone.
Diseases, Conditions and the Like The conjugates, pharmaceutical compositions, and methods provided herein can be useful for the treatment of a plurality of diseases, conditions, preventing a disease or a condition in a subject or other therapeutic applications for subjects in need thereof. The conjugates, pharmaceutical compositions, and methods provided herein can be useful for treatment of liver diseases, such as liver fibrosis or liver cancer. In some aspects, the fibrosis is cancer-associated. In some aspects, the fibrosis is not cancer-associated. In some aspects, the fibrosis is associated with scleroderma, systemic fibrosis, steatohepatitis, or non-alcoholic steatohepatitis (NASH), chronic viral HBV and HCV infection, autoimmune hepatitis, or primary biliary cholangitis. The disclosure further provides any conjugates disclosed herein for use in a method of treatment of the human or animal body by therapy. Therapy may be by any mechanism disclosed herein, such as inhbiting, reducing, or reducing progression of fibrosis or other liver damage. The disclosure further provides any conjugate disclosed herein for prevention or treatment of any condition disclosed herein, for example, fibrosis or cancer. The disclosure also provides any conjugate or pharmaceutical composition thereof disclosed herein for obtaining any clinical outcome disclosed herein for any condition disclosed herein, such as treating fibrosis or cancer. The disclosure also provides use of any conjugate disclosed herein in the manufacture of a medicament for preventing or treating any condition disclosed herein. EXAMPLES The following examples illustrate the various methods of making compounds described herein. The examples are included to further describe some embodiments of the present disclosure, and should not be used to limit the scope of the disclosure. The following synthetic schemes are provided for purposes of illustration, not limitation. It is understood that one skilled in the art may be able to make these compounds by similar methods or by combining other methods known to one skilled in the art. It is also understood that one skilled in the art would be able to make, in a similar manner as described below by using the appropriate starting materials and modifying the synthetic route as needed. In general, starting materials and reagents can be obtained from commercial vendors or synthesized according to sources known to those skilled in the art or prepared as described herein.
General Scheme 1 for the Preparation of Exemplary Inhibitors of
Formula (A-I) or (B-I)
In one method, compounds of Formula (A-I), or compounds of Formula (B-I) wherein Q is CH2N, are prepared according to Scheme 1. Specifically, R2-substituted pyridine-2- carbaldehydes are reacted with aniline and diphenyl phosphite to give N,P-acetal (iv), which can be further coupled with R1 substituted [1,2,4]triazolo[1,5-a]pyridine-6-carbaldehydes (iii) followed by hydrolysis in acidic condition to produce a monoketone (v). The monoketone (v) may be oxidized to a diketone (vi) with HBr in DMSO. Certain diketone analogs (vi) were commercially available (e.g. R1 = H and R2 = CH3: CAS No. 356560-84-4) and were used as a starting point when appropriate. Diketones (vi) can be condensed with 2,2- dimethoxyacetaldehyde in the presence of ammonium acetate to yield an acetal-protected imidazole (vii), which can be hydrolyzed in acidic condition to produce an imidazole-2- carbaldehyde (viii). The imidazole-2-carbaldehyde (viii) can be reductively aminated in the presence of an amine and a reducing agent such as sodium borohydride or sodium
cyanoborohydride to yield a compound of Formula (A-I) or Formula (B-I), where substituents are selected as appropriate. General Scheme 2 for the Preparation of Exemplary ALK5 Inhibitors of Formula (C-I)
In one method, compounds of Formula (C-I) wherein Q3, Q4, Q5 and Q6 are CH are prepared according to Scheme 1. Specifically, R1-substituted pyridine-2-carbaldehydes are reacted with aniline and diphenyl phosphite to give N,P-acetal (ii), which can be further coupled with R7, R8 substituted quinoline-6-carbaldehydes (i) followed by hydrolysis in acidic condition to produce a monoketone (iii). The monoketone (iii) may be oxidized to a diketone (iv) with HBr in DMSO. Diketones (iv) can be condensed with aldehydes (v) in the presence of ammonium acetate to yield an imidazole (vi). The protecting group (PG) substituted nitrogen can then be depretoected under appropriate conditions to yield a compound of Formula (C-I).
General Scheme 3 for the Preparation of Exemplary ALK5 Inhibitors of Formula (D-I)
In one method, compounds of Formula (I) are prepared according to Scheme 1. Specifically, bromo-substituted pyrazoles (i) are reacted with an aryl- or heteroaryl-boronic acid (or ester) (iii) in the presence of an appropriate transition metal catalyst (e.g. palladium-based) to generate compounds of Formula (I). Alternatively, bromo-substituted pyrazoles can be converted to a boronic ester, or ester, (ii) via a borylation reaction. The resultant compound (ii) can then be coupled with an appropriate aryl- or heteroaryl-halide (or pseudohalide) (iv) to generate compounds of Formula (I).
EXAMPLE 1: Synthesis of 1-(5-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-4-(6-methylpyridin-2-yl)-1H-imidazol-2- yl)-N-benzylmethanamine (Compound 1)
Step A. Preparation of Int 1a
(Int 1a) To a stirred solution of 1-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-2-(6-methylpyridin-2- yl)ethane-1,2-dione (CAS 356560-84-4; 2.7 g, 10.1 mmol) in a 2:1 mixture of tert-butyl methyl ether and methanol (25 mL) were added 60% 2,2-dimethoxyacetaldehyde in H2O (3.5 mL, 20 mmol) and NH4OAc (1.95 g, 25.2 mmol). The mixture was stirred at room temperature for 5 h before the solvent was removed. The pH of the reaction mixture was adjusted to 8 with saturated aqueous NaHCO3 solution and extracted with CH2Cl2 (2 × 10 mL). The combined organic extracts were washed with brine (10 mL) and dried over anhydrous Na2SO4, filtered, and evaporated. The residue was purified on silica gel (ISCO gold, 40 g; 0% to 20% CH2Cl2/MeOH over 15 min) to give the desired imidazole product, which was dissolved in 1 N HCl (20 mL) and heated at 70°C for 4 h. The reaction mixture was allowed to cool to 0°C and then it was neutralized with saturated aqueous NaHCO3 solution. The precipitate was collected and washed with water (20 mL) and ether (40 mL) to give Int 1a as a yellow-brown solid. 1H NMR (DMSO- d6) δ 10.0 (dd, J=1.6, 0.8 Hz, 1H), 9.56 (s, 1H), 8.42 (s, 1H), 8.27 (dd, J=9.2, 1.6Hz, 1H), 7.82 (br d, J = 0.8 Hz, 1H), 7.72 (dd, J = 9.2, 0.8Hz, 1H), 7.65 (t, J=7.8Hz, 1H), 7.05 (d, J=7.6Hz, 1H), 2.46 (s, 3H). LCMS (M+H) = 305.1.
Step B. Preparation of Compound 1
To a solution of 4-(6-methyl-2-pyridyl)-5-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-1H- imidazole-2-carbaldehyde (200 mg, 0.66 mmol) in dichloroethane (30 mL) was added acetic acid (79 mg, 1.3 mmol, 75 μL) and phenylmethanamine (106 mg, 0.99 mmol). The mixture was stirred at 60°C for 2 h then cooled to 0°C. Methanol (20 mL) and THF (10 mL) were added followed by NaBH3CN (165 mg, 2.63 mmol, 4.0 eq) and then the reaction mixture was allowed to warm to 15°C and stirred for an additional 3 h, at which time LCMS showed the reaction to be complete. The reaction mixture was quenched by addition of water (0.10 mL) at 0°C and was then concentrated under reduced pressure to give a residue that was purified by silica gel chromatography (0 Æ 10% MeOH in DCM) to afford 240 mg of 1-(5-([1,2,4]triazolo[1,5- a]pyridin-6-yl)-4-(6-methylpyridin-2-yl)-1H-imidazol-2-yl)-N-benzylmethanamine as an off- white solid. 1H NMR (DMSO-d6, 400MHz) δ 9.54 (s, 1H), 8.55 (s, 1H), 7.99-7.96 (m, 1H), 7.92-7.88 (m, 1H), 7.79 (t, J = 7.8 Hz, 1H), 7.58-7.53 (m, 3H), 7.49-7.41 (m, 3H), 7.26 (d, J = 7.7 Hz, 1H), 4.37 (d, J = 12.8 Hz, 4H), 2.52 (s, 3H). LCMS M/z 396.1 [M+H]+. EXAMPLE 2: Synthesis of 6-(4-(6-methylpyridin-2-yl)-2-(piperidin-4-yl)-1H-imidazol-5-yl)- [ ound 1.47)
Step A. Preparation of Int. 1.47a
To a solution of 1-(6-methyl-2-pyridyl)-2-([1,2,4]triazolo[1,5-a]pyridin-6-yl) ethane-1,2- dione (0.2 g, 751.16 umol, 1 eq) and tert-butyl 4-formylpiperidine-1-carboxylate (160.20 mg, 751.16 umol, 1 eq) in MTBE (5 mL) and MeOH (10 mL) was added NH4OAc (289.51 mg, 3.76 mmol, 5 eq). The mixture was stirred at 25°C for 4 h. The reaction mixture was quenched by addition of H2O (20 mL), and was extracted with EtOAc (20 mL x 3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give tert-butyl 4-(5-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-4-(6-methylpyridin- 2-yl)-1H-imidazol-2-yl)piperidine-1-carboxylate (0.3 g, crude) was obtained as a yellow solid. Step B. Preparation of Compound 1.47
To a solution of tert-butyl 4-[4-(6-methyl-2-pyridyl)-5-([1,2,4]triazolo[1,5-a]pyridin-6- yl) -1H-imidazol-2-yl]piperidine-1-carboxylate (Int.1.47a) (0.3 g, 652.82 umol, 1 eq) in DCM (10 mL) was added HCl/EtOAc (4 M, 326.41 uL, 2 eq). The mixture was stirred at 25°C for 1 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Nano-micro Kromasil C18100*30mm 5um; mobile phase: [water (0.1%TFA) -ACN]; B%: 1%-20%, 10 min) and freeze-drying. Compound 6-[4-(6- methyl-2-pyridyl)-2-(4-piperidyl)-1H-imidazol-5-yl]-[1,2,4]triazolo[1,5-a]pyridine (Compound 1.47) (0.17 g, 359.07 umol, 55.00% yield, TFA) was obtained as a yellow solid. 1H NMR (CDCl3, 400 MHz) δ 9.44 (s, 1H), 8.94 (s, 1H), 8.65 (d, J = 6.0 Hz, 1H), 8.60 (s, 1H), 7.94 (d, J = 9.2 Hz, 1H), 7.87-7.80 (m, 3H), 7.52-7.47 (m, 1H), 7.36 (d, J = 7.6 Hz, 1H), 3.86 (s, 1H), 3.46
(d, J = 12.4 Hz, 2H), 3.41-3.33 (m, 1H), 3.17-3.04 (m, 2H), 2.56-2.53 (m, 3H), 2.27 ( d, J = 12.4 Hz, 2H), 2.11-2.00 (m, 2H); HPLC: 96.356% (220 nm), 98.486% (254 nm); MS (ESI): mass calcd. for C20H21N7359.19, m/z found 360.0[M+H]+. The compounds in Tables 14 and 15 were prepared in a manner similar to that described for Compounds 1 or 1.47 using the appropriately substituted amine. TABLE 14
TABLE 15
EXAMPLE 3 Preparation of ALK5 Inhibitors of Formula (C-I) Synthesis of tert-butyl (2-((6-(2-(6-methylpyridin-2-yl)-2-oxoacetyl)quinolin-3-yl)oxy)ethyl) carbamate (Compound A1)
Step 1: Preparation of 6-bromo-3-hydroxy-quinoline-4-carboxylic acid (Compound A1-b)
To a solution of KOH (40.3 g, 718 mmol, 8.0 equiv) in H2O (400 mL) was added 5- bromoindoline-2,3-dione (A1-a) (20.3 g, 89.8 mmol, 1.0 equiv) at 20°C under N2. The mixture was stirred at 50°C for 1.5 hours, then 3-bromo-2-oxo-propanoic acid (15.0 g, 89.8 mmol, 1.0 equiv) was added and it was stirred at 25°C for 90 hours. The reaction mixture was quenched with aq HCl (4 M) at 0°C until pH = 4, desired solid was precipitated and filtered to afford 6- bromo-3-hydroxy-quinoline-4-carboxylic acid (A1-b) (21.0 g, 78.3 mmol, 87.2% yield) as a brown solid. 1H NMR (DMSO-d6, 400 MHz) δ8.69 (s, 1H), 8.54 (s, 1H), 7.82 (d, J = 8.4 Hz, 1H), 7.60 (d, J = 8.4 Hz, 1H) Step 2: Preparation of 6-bromoquinolin-3-ol (Compound A1-c)
A solution of 6-bromo-3-hydroxy-quinoline-4-carboxylic acid (A1-b) (15.0 g, 56.0 mmol, 1.0 equiv) in nitrobenzene (150 mL) was heated to 180°C and then stirred for 6 hours at this temperature. The reaction mixture was cooled to 0 °C and methyl tert-butyl ether (300 mL) was added, desired solid was precipitated and then filtered to afford 6-bromoquinolin-3-ol (A1-c) (7.0 g, 31.2 mmol, 55.8% yield) as brown solid. 1H NMR (DMSO-d6, 400 MHz) δ8.57 (d, J = 2.4 Hz, 1H), 8.01 (d, J = 1.2 Hz, 1H), 7.77 (d, J = 9.2 Hz, 1H), 7.53 (dd, J = 2.0, 9.2 Hz, 1H), 7.45 (d, J = 2.0 Hz, 1H)
Step 3: Preparation 2-[(6-bromo-3-quinolyl)oxy]ethyl]carbamate
(Compound A1-d)
To a mixture of 6-bromoquinolin-3-ol (A1-c) (1.00 g, 4.46 mmol, 1.0 equiv) and tert- butyl N-(2-bromoethyl)carbamate (5.00 g, 22.3 mmol, 5.0 equiv) in DMF (30 mL) was added KI (741 mg, 4.46 mmol, 1.0 equiv) and K2CO3 (1.85 g, 13.4 mmol, 3.0 equiv) at 25°C under N2. The mixture was stirred at 45 °C for 10 hours. Water (70 mL) was added to the reaction mixture and the aqueous phase was extracted with ethyl acetate (50 mL x 3). The combined organic phase was washed with brine (40 mL x 2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (column height: 250 mm, diameter: 100 mm, 100-200 mesh silica gel, Petroleum ether/Ethyl acetate=10/1, 1/1) to afford tert-butyl N-[2-[(6-bromo-3-quinolyl)oxy]ethyl]carbamate (A1-d) (700 mg, 1.91 mmol, 42.7% yield) as brown solid. 1H NMR (400 MHz, DMSO-d6) δ8.60 (d, J = 2.4 Hz, 1H), 8.08 (d, J = 2.4 Hz, 1H), 7.83 (d, J = 8.8 Hz, 1H), 7.72 (d, J = 2.4 Hz, 1H), 7.64 (dd, J = 2.4, 8.8 Hz, 1H), 7.05 (br t, J = 5.6 Hz, 1H), 4.08 (t, J = 5.6 Hz, 2H), 3.34 (q, J = 5.6 Hz, 2H), 1.33 (s, 9H) Step 4: Preparation o butyl N-[2-[(6-formyl-3-quinolyl)oxy]ethyl]carbamate
(Compound A1-e)
To a solution of tert-butyl N-[2-[(6-bromo-3-quinolyl)oxy]ethyl]carbamate (A1-d) (1.80 g, 4.90 mmol, 1.0 equiv) in DMF (30 mL) was added Pd(dppf)Cl2 (357 mg, 490 umol, 0.1 equiv), Et3N (1.49 g, 14.7 mmol, 2.05 mL, 3.0 equiv) and Et3SiH (11.4 g, 98.0 mmol, 15.7 mL,
20.0 equiv) under N2. The suspension was degassed under vacuum and purged with CO several times. The mixture was stirred under CO (50 psi) at 80°C for 15 hours. Water (70 mL) was added to the reaction mixture and the aqueous phase was extracted with ethyl acetate (20 mL x 3). The combined organic phase was washed with brine (50 mL x 2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (column height: 250 mm, diameter: 100 mm, 100-200 mesh silica gel, Petroleum ether/Ethyl acetate = 10/1, 1/1) to afford tert-butyl N-[2-[(6-formyl-3-quinolyl) oxy]ethyl]carbamate (A1-e) (800 mg, 2.53 mmol, 51.6% yield) as yellow solid. 1H NMR (CDCl3, 400 MHz) δ10.13 (s, 1H), 8.75 (d, J = 2.4 Hz, 1H), 8.21 (d, J = 1.2 Hz, 1H), 8.10 (d, J = 8.4 Hz, 1H), 8.00 (d, J = 1.6 Hz, 1H), 7.49 (d, J = 2.4 Hz, 1H), 4.18-4.13 (m, 2H), 3.62-3.58 (m, 2H), 1.41 (s, 9H) Step 5: Preparation of tert-butyl N-[2-[[6-[2-(6-methyl-2-pyridyl)-2-oxo-ethyl]-3- quinolyl]oxy]ethyl]carbamate (Compound A1-f)
To a mixture of tert-butyl N-[2-[(6-formyl-3-quinolyl)oxy]ethyl]carbamate (A1-e) (760 mg, 2.40 mmol, 1.0 equiv) and N-[diphenoxyphosphoryl-(6-methyl-2-pyridyl)methyl]aniline (1.03 g, 2.40 mmol, 1.0 equiv) in THF (15 mL) and i-PrOH (5 mL) was added Cs2CO3 (783 mg, 2.40 mmol, 1.0 equiv) at 25°C under N2 and then stirred at 25°C for 2 hours. The reaction mixture was quenched with aq HCl (4 M) at 0°C until pH = 4 and then stirred for another 2 hours. Water (20 mL) was added and the aqueous phase was extracted with ethyl acetate (10 mL x 3). The combined organic phase was washed with brine (20 mL x 2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel
chromatography (column height: 250 mm, diameter: 100 mm, 100-200 mesh silica gel, Petroleum ether/Ethyl acetate = 10/1, 1/1) to afford tert-butyl N-[2-[[6-[2-(6-methyl-2-pyridyl)- 2-oxo-ethyl]-3-quinolyl]oxy]ethyl] carbamate (A1-f) (0.45 g, 1.07 mmol, 44.4% yield) as yellow oil. Step 6: Preparation of tert-butyl N-[2-[[6-[2-(6-methyl-2-pyridyl)-2-oxo-acetyl]-3- quinolyl]oxy]ethyl]carbamate (Compound A1)
To a solution of tert-butyl N-[2-[[6-[2-(6-methyl-2-pyridyl)-2-oxo-ethyl]-3-quinolyl]oxy] ethyl]carbamate (A1-f) (450 mg, 1.07 mmol, 1.0 equiv) in MeCN (15 mL) was added Pd/C (518 mg, 5% purity, 0.2 equiv) under N2. The suspension was degassed under vacuum and purged with O2 several times. The mixture was stirred under O2 (15 psi) at 80°C for 5 hours. The mixture was filtered and the filtrate was concentrated in vacuum to afford tert-butyl N-[2-[[6-[2- (6-methyl-2-pyridyl)-2-oxo-acetyl]-3-quinolyl]oxy]ethyl]carbamate (Compound A1) (390 mg, 896 umol, 83.9% yield) as yellow oil. 1H NMR (CDCl3, 400 MHz) δ8.75 (d, J = 2.4 Hz, 1H), 8.22 (s, 1H), 8.15-8.07 (m, 2H), 8.02 (d, J = 7.6 Hz, 1H), 7.80 (t, J = 7.6 Hz, 1H), 7.40 (d, J = 2.8 Hz, 1H), 7.35 (d, J = 8.0 Hz, 1H), 5.01 (s, 1H), 4.12 (t, J = 5.2 Hz, 2H), 3.59 (q, J = 5.2 Hz, 2H), 2.44 (s, 3H), 1.42 (s, 9H)
Synthesis of tert-butyl (2-((7-(2-(6-methylpyridin-2-yl)-2-oxoacetyl)quinoxalin-2-yl) ethyl)
carbamate (Compound A2)
Step 1: Preparation 2-(7-bromoquinoxalin-2-yl)oxyethyl]carbamate
(Compound A2-b)
To a mixture of 7-bromo-2-chloro-quinoxaline (A2-a) (2 g, 8.21 mmol, 1.0 equiv) and tert-butyl N-(2-hydroxyethyl)carbamate (1.99 g, 12 mmol, 1.9 mL, 1.5 equiv) in DMF (20 mL) was added K2CO3 (1.70 g, 12 mmol, 1.5 equiv), and then stirred at 80°C for 12 h. The reaction mixture was quenched by addition H2O (20 mL) at 20°C, and then extracted with EtOAc (20 mL x 2). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered
and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=1/0 to 4/1) to give compound tert-butyl N- [2-(7-bromoquinoxalin-2-yl)oxyethyl]carbamate (A2-b) (2.5 g, 7 mmol, 82.66% yield) as a yellow solid. 1H NMR (CDCl3, 400MHz) δ8.45 (s, 1H), 7.99 (d, J = 2.4 Hz, 1H), 7.85 (d, J = 8.8 Hz, 1H), 7.64 (dd, J = 2.4, 8.8 Hz, 1H), 4.53 (t, J = 5.2 Hz, 2H), 3.65-3.60 (m, 2H), 1.44 (s, 9H) Step 2: Preparation of tert-butyl N-[2-(7-formylquinoxalin-2-yl)oxyethyl]carbamate (Compound A2-c)
To a solution of tert-butyl N-[2-(7-bromoquinoxalin-2-yl)oxyethyl]carbamate (A2-b) (2.5 g, 6.79 mmol, 1.0 equiv) in DMF (30 mL) was added Et3N (2.06 g, 20.4 mmol, 2.83 mL, 3.0 equiv), Et3SiH (15.8 g, 136 mmol, 21.7 mL, 20.0 equiv), and Pd(dppf)Cl2 (496 mg, 678 umol, 0.1 equiv), the suspension was degassed under vacuum and purged with CO several times. The mixture was stirred under CO (50 psi) at 80°C for 20 hours. The reaction mixture was quenched by addition H2O (50 mL) at 20°C, and then extracted with EtOAc (30 mL x 3). The combined organic layers were washed with brine (30 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 3/1) to give compound tert-butyl N-[2-(7- formylquinoxalin-2-yl)oxyethyl]carbamate (A2-c) (1 g, 3 mmol, 46.41% yield) as a yellow solid. 1H NMR (CDCl3, 400MHz) δ10.22 (s, 1H), 8.59 (s, 1H), 8.32 (d, J = 1.6 Hz, 1H), 8.15 (d, J = 8.4 Hz, 1H), 8.07 (d, J = 8.4 Hz, 1H), 5.00 (s, 1H), 4.60 (t, J = 5.2 Hz, 2H), 3.68-3.62 (m, 2H), 1.46 (s, 9H)
Step 3: Preparation of tert-butyl N-[2-[7-[2-(6-methyl-2-pyridyl)-2-oxo-ethyl]quinoxalin- 2-yl]oxyethyl]carbamate (Compound A2-d) To a mixture of tert-butyl N-[2-(7-formylquinoxalin-2-yl)oxyethyl]carbamate (A2-c) (1 g, 3.15 mmol, 1.0 equiv) and N-[diphenoxyphosphoryl-(6-methyl-2-pyridyl)methyl]aniline (2.03 g, 4.73 mmol, 1.5 equiv) in THF (10 mL) and i-PrOH (4 mL) was added Cs2CO3 (1.54 g, 4.73 mmol, 1.5 equiv) at 20°C, and then stirred at 35°C for 6 h, the pH of the mixture was adjusted to pH~3 with 2N HCl at 0°C, and it was stirred at 20°C for another 2 h. The reaction mixture was poured into H2O (20 mL), and then the pH of the mixture was adjusted to ~8 by adding aq. NaHCO3, extracted with EtOAc (20 mL) for twice. The organic lawyers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 3/1) to give compound tert-butyl N-[2-[7-[2-(6-methyl-2-pyridyl)-2-oxo-ethyl]quinoxalin-2- yl]oxyethyl]carbamate (A2-d) (1.5 g, crude) as a yellow solid. 1H NMR (MeOD, 400MHz) δ8.39 (s, 1H), 7.88 (d, J = 8.4 Hz, 1H), 7.85-7.82 (m, 1H), 7.80 (d, J = 7.6 Hz, 1H), 7.74 (d, J = 1.2 Hz, 1H), 7.56 (dd, J = 1.6, 8.4 Hz, 1H), 7.45 (dd, J = 1.2, 7.2 Hz, 1H), 4.73 (s, 2H), 4.52 (t, J = 5.2 Hz, 2H), 3.53 (t, J = 5.2 Hz, 2H), 2.66 (s, 3H), 1.42 (s, 9H)
Step 3: Preparation of tert-butyl N-[2-[7-[2-(6-methyl-2-pyridyl)-2-oxo-acetyl] quinoxalin-2-yl]oxyethyl]carbamate (Compound A2)
To a solution of tert-butyl N-[2-[7-[2-(6-methyl-2-pyridyl)-2-oxo-ethyl]quinoxalin-2-yl] oxyethyl]carbamate (A2-d) (1.5 g, 3.55 mmol, 1.0 equiv) in MeCN (20 mL) was added Pd/C (0.5 g, 10% purity) at 20°C, the suspension was degassed under vacuum and purged with O2 several times. The mixture was stirred under O2 (15psi) at 80°C for 2 hours. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 2/1) to give compound tert-butyl N-[2-[7-[2-(6-methyl-2-pyridyl)-2-oxo-acetyl]quinoxalin-2- yl]oxyethyl]carbamate (Compound A2) (0.94 g, 2.15 mmol, 60.66% yield) as a yellow solid. 1H NMR (CDCl3, 400MHz) δ8.57 (s, 1H), 8.27 (d, J = 1.2 Hz, 1H), 8.21-8.18 (m, 1H), 8.17-8.13 (m, 1H), 8.06 (d, J = 7.6 Hz, 1H), 7.84 (t, J = 7.6 Hz, 1H), 7.39 (d, J = 7.6 Hz, 1H), 4.54 (t, J = 5.2 Hz, 2H), 3.61 (d, J = 5.2 Hz, 2H), 2.48 (s, 3H), 1.44 (s, 9H) ALK5 INHIBITORS OF FORMULA (C-I)—SYNTHETIC METHOD A Synthesis of N-[[5-[3-(2-aminoethoxy)-6-quinolyl]-4-(6-methyl-2-pyridyl)-1H-imidazol-2- yl] methyl]-2-fluoro-aniline (Compound C1.1)
Synthetic Scheme
Step 1: Preparation of tert-butyl N-(2-fluorophenyl)carbamate (Compound C1.1-b)
To a solution of 2-fluoroaniline (C1.1-a) (25 g, 225 mmol, 1.0 equiv) in THF (300 mL) was added Boc2O (73.6 g, 337 mmol, 77.5 mL, 1.5 equiv) at 25°C, and then stirred at 80°C for 12h. The reaction mixture was concentrated under reduced pressure to give tert-butyl N-(2- fluorophenyl) carbamate (C1.1-b) (47 g, 223 mmol, 98.90% yield) as colorless oil. 1H NMR (DMSO-d6, 400MHz) δ8.98 (s, 1H), 7.68-7.60 (m, 1H), 7.27-7.10 (m, 3H), 1.50 (s, 9H)
Step 2: Preparation of tert-butyl N-allyl-N-(2-fluorophenyl)carbamate (Compound C1.1-c)
To a mixture of tert-butyl N-(2-fluorophenyl)carbamate (C1.1-b) (10 g, 47 mmol, 1.0 equiv) in DMF (100 mL) was added NaH (2.84 g, 71 mmol, 60% purity, 1.5 equiv) slowly at 0°C, the mixture was stirred 10 min at 0°C, then 3-bromoprop-1-ene (8.59 g, 71 mmol, 1.5 equiv) was added, and it was stirred at 20°C for 2 h. The reaction mixture was poured into H2O (100 mL), extracted with EtOAc (100 mL) for twice. The organic lawyers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=1/0 to 1/0) to give compound tert-butyl N-allyl-N-(2-fluorophenyl)carbamate (C1.1-c) (10.3 g, 41 mmol, 86.58% yield) as a colorless liquid. 1H NMR (DMSO-d6,400 MHz) δ7.35-7.10 (m, 4H), 5.86-5.72 (m, 1H), 5.14-5.02 (m, 2H), 4.14 (d, J = 5.6 Hz, 2H), 1.38-1.31 (m, 9H) Step 3: Preparation of tert-butyl N-(2-fluorophenyl)-N-(2-oxoethyl)carbamate (Compound C1.1-d)
To a mixture of tert-butyl N-allyl-N-(2-fluorophenyl)carbamate (1.1-c) (10.3 g, 41.0 mmol, 1.0 equiv) in THF (100 mL) and H2O (100 mL) was added K2OsO4 .2H2O (453 mg, 1 mmol, 0.03 equiv) and NaIO4 (43.8 g, 205 mmol, 5.0 equiv) at 20°C, and then stirred at 35°C
for 5h. The reaction mixture was filtered and the filtrate was poured into H2O (100 mL), extracted with EtOAc(100 mL) for twice. The organic lawyers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=1/0 to 9/1) to give compound tert-butyl N-(2-fluorophenyl)-N -(2-oxoethyl)carbamate (1.1-d) (4.26 g, 17 mmol, 41.04% yield) as a brown oil. 1H NMR (CDCl3, 400 MHz) δ9.71 (s, 1H), 7.34-7.17 (m, 2H), 7.12 (d, J = 8.8 Hz, 2H), 4.28-4.16 (m, 2H), 1.40 (d, J = 6.4 Hz, 9H) Step 4: Preparation of tert-butyl N-[[5-[3-[2-(tert-butoxycarbonylamino)ethoxy]-6- quinolyl]-4-(6-methyl-2-pyridyl)-1H-imidazol-2-yl] methyl]-N-(2-fluorophenyl) carbamate (Compound C1.1-e)
To a mixture of tert-butyl N-[2-[[6-[2-(6-methyl-2-pyridyl)-2-oxo-acetyl]-3- quinolyl]oxy] ethyl]carbamate (A1) (100 mg, 223 umol, 1.0 equiv) and tert-butyl N-(2- fluorophenyl)-N-(2-oxoethyl)carbamate (1.1-d) (87.0 mg, 344 umol, 1.5 equiv) in THF (5 mL) was added NH4OAc (89 mg, 1.15 mmol, 5 equiv), and then stirred at 35°C for 12 hr. The reaction mixture was quenched by addition of H2O ^10 mL ^at 20°C, and then extracted with EtOAc (10 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 1/1) to give compound tert-butyl N-[[5-[3-[2-(tert-butoxycarbonylamino)ethoxy]-6-quinolyl]-4-(6-methyl-2- pyridyl)-1H-imidazol-2-yl]methyl]-N-(2-fluorophenyl)carbamate (1.1-e) (127 mg, 190 umol, 82.7% yield) as a white solid. 1H NMR (MeOD, 400MHz) δ8.59 (d, J = 2.8 Hz, 1H), 7.97 (s, 1H), 7.91 (d, J = 8.8 Hz, 1H), 7.68 (d, J = 2.8 Hz, 1H), 7.63 (d, J = 8.0 Hz, 1H), 7.57 (s, 1H), 7.31 (d, J = 6.4 Hz, 1H),
7.20-7.11 (m, 4H), 6.95 (s, 1H), 4.94 (s, 2H), 4.19 (t, J = 5.2 Hz, 2H), 3.52 (q, J = 5.2 Hz, 2H), 2.50 (s, 3H), 1.44 (s, 18H) Step 5: Preparation of N-[[5-[3-(2-aminoethoxy)-6-quinolyl]-4-(6-methyl-2-pyridyl)-1H- imidazol-2-yl] methyl]-2-fluoro-aniline (Compound C1.1)
To a mixture of tert-butyl N-[[5-[3-[2-(tert-butoxycarbonylamino)ethoxy]-6-quinolyl]-4- (6- methyl-2-pyridyl)-1H-imidazol-2-yl]methyl]-N-(2-fluorophenyl)carbamate (0.127 g, 189.91 umol, 1 equiv) in EtOAc (2 mL) was added HCl/EtOAc (4M, 5 mL) and then stirred at 20°C for 30 min. The reaction mixture was concentrated under reduced pressure to give N-[[5-[3-(2- aminoethoxy)-6-quinolyl]-4-(6-methyl-2-pyridyl)-1H-imidazol-2-yl]methyl]-2-fluoro-aniline (Compound C1.1) (80.0 mg, 146 umol, 77.0% yield, 92.3% purity, HCl) as a yellow solid. 1H NMR (MeOD, 400 MHz) δ9.22 (d, J = 2.4 Hz, 1H), 8.85 (d, J = 2.4 Hz, 1H), 8.53 (d, J = 1.6 Hz, 1H), 8.31 (d, J = 9.2 Hz, 1H), 8.16-8.02 (m, 2H), 7.69 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 8.0 Hz, 1H), 7.10-7.00 (m, 2H), 6.91-6.83 (m, 1H), 6.80-6.71 (m, 1H), 4.89 (s, 2H), 4.65 (t, J = 4.8 Hz, 2H), 3.54 (t, J = 4.4 Hz, 2H), 2.76 (s, 3H) HPLC: 92.30% (220 nm), 95.72% (254 nm) MS (ESI): mass calcd. For C27H25FN6O 468.21, m/z found 469.2[M+H]+
Synthesis of N-[[5-[3-(2-aminoethoxy)-6-quinolyl]-4-(6-methyl-2-pyridyl)-1H-imidazol-2- yl] methyl]-2-fluoro-aniline (Compound C1.2)
Step 1: Preparation of tert-butyl N-[[5-[3-[2-(tert-butoxycarbonylamino)ethoxy] quinoxalin-6-yl]-4-(6-methyl-2-pyridyl)-1H-imidazol-2-yl]methyl]-N-(2-fluorophenyl) carbamate (Compound C1.2-a)
To a mixture of tert-butyl N-[2-[7-[2-(6-methyl-2-pyridyl)-2-oxo-acetyl]quinoxalin-2-yl] oxyethyl]carbamate (A2) (300 mg, 687 umol, 1.0 equiv) and tert-butyl N-(2-fluorophenyl)-N-(2- oxoethyl)carbamate (C1.1-d) (261 mg, 1.03 mmol, 1.5 equiv) in THF (5 mL) was added NH4OAc (265 mg, 3.44 mmol, 5.0 equiv), and then stirred at 35°C for 12 h. The reaction mixture was quenched by addition H2O (10 mL) at 20 °C, and then extracted with EtOAc (10 mL x 3). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 1/1) to give compound tert-butyl N-[[5-[3-[2-(tert-butoxycarbonylamino) ethoxy]quinoxalin-6-yl]-4-(6-methyl-2- pyridyl)-1H-imidazol-2-yl]methyl]-N-(2-fluorophenyl)carbamate (C1.2-a) (300 mg, 448 umol, 65.17% yield) as a yellow solid. 1H NMR (MeOD, 400MHz) δ8.44 (s, 1H), 7.97 (d, J = 1.2 Hz, 1H), 7.92 (d, J = 8.8 Hz, 1H), 7.67 (d, J = 7.6 Hz, 1H), 7.61 (t, J = 7.6 Hz, 1H), 7.34-7.28 (m, 3H), 7.21-7.13 (m, 3H), 4.95 (s, 2H), 4.51 (t, J = 5.2 Hz, 2H), 3.51 (t, J = 5.6 Hz, 2H), 2.51 (s, 3H), 1.41 (s, 18H) Step 2: Preparation of tert-butyl N-[[5-[3-[2-(tert-butoxycarbonylamino)ethoxy] quinoxalin-6-yl]-4-(6-methyl-2-pyridyl)-1H-imidazol-2-yl]methyl]-N-(2-fluorophenyl) carbamate (Compound C1.2)
To a mixture of tert-butyl N-[[5-[3-[2- (tert-butoxycarbonylamino)ethoxy]quinoxalin-6- yl]-4-(6-methyl-2-pyridyl)-1H-imidazol-2-yl]methyl]-N-(2-fluorophenyl)carbamate (C1.2-a) (300 mg, 448 umol, 1.0 equiv) in EtOAc (5 mL) was added HCl/EtOAc (4 M 10 mL), and then stirred at 20°C for 1h. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase HPLC(column: YMC-Actus Triart C18100 x 30mm x 5um;mobile phase: [water(0.04%HCl)-ACN];B%: 10%-40%,7min) to give compound N-[[5-[3-(2-aminoethoxy)quinoxalin-6-yl]-4-(6-methyl-2-pyridyl)-1H-imidazol-2-
yl]methyl]-2-fluoro-aniline (Compound C1.2) (101 mg, 164 umol, 36.64% yield, 4HCl) as a yellow solid. 1H NMR (MeOD, 400MHz) δ8.66 (s, 1H), 8.15 (d, J = 8.4 Hz, 1H), 8.12 (d, J = 1.6 Hz, 1H), 8.01 (t, J = 8.0 Hz, 1H), 7.75 (dd, J = 2.0 Hz, J = 8.8 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.53 (d, J = 8.0 Hz, 1H), 7.09-7.01 (m, 2H), 6.84 (t, J = 8.4 Hz, 1H), 6.79-6.73 (m, 1H), 4.86 (s, 2H), 4.78-4.74 (m, 2H), 3.48 (t, J = 4.8 Hz, 2H), 2.74 (s, 3H) HPLC: 96.99% (220 nm), 96.53% (254 nm) MS (ESI): mass calcd. For C26H24FN7O 469.20, m/z found 470.1 [M+H]+. Compounds C1.3-C1.66, as illustrated in Table 16, could be prepared in a manner similar to that described for Compound 1.1 and Compound 1.2 using appropriate intermediates according to Method A as described herein. TABLE 16 COMPOUND C1.3–C1.66
ALK5 INHIBITORS OF FORMULA (C-I)—SYNTHETIC METHOD B Synthesis of 2-[7-[2- [2-(2- fluorophenyl) ethyl]-4-(6-methyl-2-pyridyl)-1H-imidazol-5-yl] quinoxalin-2-yl]oxyethanamine (Compound C2.1) METHOD B
Step 1: Preparation of tert- 2-[7-[2-[2-(2-fluorophenyl)ethyl]-4-(6-methyl-2-
imidazol-5-yl]quinoxalin-2-yl]oxyethyl]carbamate (Compound C2.1-a)
To a mixture of tert-butyl N-[2-[7-[2-(6-methyl-2-pyridyl)-2-oxo-acetyl]quinoxalin-2-yl] oxyethyl]carbamate (A2) (0.15 g, 343 umol, 1.0 equiv) and 3-(2-fluorophenyl)propanal (104 mg, 687 umol, 2.0 equiv) in THF (20 mL) was added NH4OAc (132 mg, 1.72 mmol, 5.0 equiv) at 25°C and then stirred at 35°C for 12 hours. The mixture was added water (50 mL) and extracted with ethyl acetate (30 mL x 3). The combined organic phase was washed with brine (30 mL), dried with anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=30/1 to 0:1). Compound tert- butyl N-[2-[7- [2-[2-(2-fluorophenyl)ethyl]-4-(6-methyl-2-pyridyl)-1H-imidazol-5-yl] quinoxalin-2-yl]oxyethyl]carbamate (C2.1-a) (0.14 g, 246 umol, 71.64% yield) was obtained as a yellow solid. Step 2: Preparation of tert-butyl N-[2-[7-[2-[2-(2-fluorophenyl)ethyl]-4-(6-methyl-2- pyridyl)-1H-imidazol-5-yl]quinoxalin-2-yl]oxyethyl]carbamate (Compound C2.1)
A solution of tert-butyl N-[2- [7-[2- [2-(2- fluorophenyl) ethyl]-4- (6-methyl-2-pyridyl)- 1H-imidazol-5-yl] quinoxalin-2-yl] oxyethyl] carbamate (C2.1-a) (0.14 g, 246 umol, 1 equiv) in HCl/EtOAc (4 M, 20 mL) was stirred at 25°C for 1 hour. The mixture was concentrated under reduced pressure at 45°C. Compound 2-[7-[2-[2-(2-fluorophenyl) ethyl]-4-(6-methyl-2-pyridyl)- 1H-imidazol-5-yl]quinoxalin-2-yl]oxyethanamine (Compound C2.1) (90 mg, 182 umol, 73.78% yield, 94.56% purity) was obtained as a yellow solid. 1H NMR (MeOD, 400 MHz) δ8.68 (s, 1H), 8.18 (d, J = 8.6 Hz, 1H), 8.10 (d, J = 1.8 Hz, 1H), 7.82-7.70 (m, 2H), 7.41 (d, J = 7.8 Hz, 1H), 7.36-7.25 (m, 3H), 7.21-7.07 (m, 2H), 4.78 (t, J = 5.2 Hz, 2H), 3.49-3.51 (m, 2H), 3.39-3.41 (m, 2H), 3.34-3.32 (m, 2H), 2.65 (s, 3H) HPLC: 94.56% (220 nm), 96.23% (254 nm) LCMS (ESI): mass calcd. for C27H25N6OF 468.21, m/z found 469.1 [M+H]+.
Compound C2.2, as illustrated in Table 17, could be prepared in a manner similar to that described for Compound C2.1 using appropriate intermediates according to Method B as described herein. TABLE 17 COMPOUND C2.2
ALK5 INHIBITORS OF FORMULA (C-1)—SYNTHETIC METHOD C Synthesis of 2-[7-[2- fluorophenoxy)methyl]-4-(6-methyl-2-pyridyl)- midazol -5-yl]
quinoxalin-2-yl]oxyethanamine (Compound C3.1) METHOD C
Synthetic Scheme
Step 1: Preparation of 1-(2,2-diethoxyethoxy)-2-fluoro-benzene (Compound
To a mixture of 2-fluorophenol (C3.1-a) (3 g, 26.7 mmol, 2.48 mL, 1.0 equiv) and 2- bromo-1,1-diethoxy-ethane (6.33 g, 32.1 mmol, 4.8 mL, 1.2 equiv) in DMF (20 mL) was added K2CO3 (5.55 g, 40.1 mmol, 1.5 equiv) at 25°C, and then stirred at 100°C for 12h. The reaction mixture was quenched by addition H2O (40 mL) at 20°C, and then extracted with EtOAc (20 mL x 3). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered
and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 3/1) to give compound 1-(2,2- diethoxyethoxy)-2-fluoro-benzene (C3.1-b) (3.2 g, 14.0 mmol, 52.39% yield) as a yellow oil. 1H NMR (CDCl3, 400MHz) δ7.10-6.97 (m, 3H), 6.94-6.87 (m, 1H), 4.85 (t, J = 5.2 Hz, 1H), 4.07 (d, J = 5.2 Hz, 2H), 3.83-3.73 (m, 2H), 3.70-3.62 (m, 2H), 1.27-1.24 (m, 6H) Step 2: Preparation of 2-(2-fluorophenoxy) acetaldehyde (Compound 3.1-c)
To a mixture of 1-(2, 2-diethoxyethoxy)-2-fluoro-benzene (C3.1-b) (1.40 g, 6.13 mmol, 1.0 equiv) in THF (20 mL) and H2O (15 mL) was added HCl (12 M, 14.9 mL, 29.2 equiv), and then stirred at 20°C for 12 hr. The reaction mixture was quenched by addition of H2O (20 mL), diluted with aqueous of NaHCO3 (1 M) until pH to between 8 and 9. The reaction mixture was extracted with EtOAc (20 mL x 3). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 3/1) to give compound 2-(2-fluorophenoxy) acetaldehyde (C3.1-c) (700 mg, 4.54 mmol, 74.0% yield) as yellow oil. 1H NMR (CDCl3, 400 MHz) δ9.95-9.86 (m, 1H), 7.18-6.89 (m, 4H), 4.65 (d, J = 0.8 Hz, 2H)
Step 3: Preparation of tert-butyl N-[2-[7-[2-[(2-fluorophenoxy)methyl]-4-(6-methyl-2- pyridyl)-1H-imidazol-5-yl]quinoxalin-2-yl]oxyethyl]carbamate (Compound 3.1-d)
To a mixture of tert-butyl N-[2-[7-[2-(6-methyl-2-pyridyl)-2-oxo-acetyl]quinoxalin-2- yl]oxyethyl]carbamate (A2) (150 mg, 343 umol, 1.0 equiv) and 2-(2- fluorophenoxy)acetaldehyde (3.1-c) (79 mg, 515 umol, 1.5 equiv) in THF (3 mL) was added NH4OAc (132 mg, 1.72 mmol, 5.0 equiv), and then stirred at 35°C for 12h. The reaction mixture was quenched by addition H2O (10 mL) at 20°C, and then extracted with EtOAc (10 mL x 3). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase HPLC(column: Phenomenex Luna C18200 x 40mm x 10um;mobile phase: [water(0.1%TFA)-ACN];B%: 18%-48%,8min) to give compound tert-butyl N-[2-[7-[2-[(2- fluorophenoxy)methyl]-4-(6-methyl-2-pyridyl)-1H-imidazol-5-yl]quinoxalin-2-yl]oxyethyl] carbamate (C3.1-d) (150 mg, 146 umol, 42.51% yield, TFA salt) as a yellow solid. 1H NMR (MeOD, 400MHz) δ8.57 (s, 1H), 8.13 (d, J = 8.4 Hz, 1H), 8.08 (d, J = 2.0 Hz, 1H), 8.03 (t, J = 8.0 Hz, 1H), 7.75 (dd, J = 2.0, 8.4 Hz, 1H), 7.56 (dd, J = 8.0, 12.4 Hz, 2H), 7.31 (t, J = 8.4 Hz, 1H), 7.21-7.13 (m, 2H), 7.07-7.00 (m, 1H), 5.41 (s, 2H), 4.53 (t, J = 5.2 Hz, 2H), 3.52 (t, J = 5.6 Hz, 2H), 2.78 (s, 3H), 1.42 (s, 9H)
Step 4: Preparation of 2-[7-[2-[(2-fluorophenoxy)methyl]-4-(6-methyl-2-pyridyl)-1H- imidazol -5-yl]quinoxalin-2-yl]oxyethanamine (Compound C3.1)
A mixture of tert-butyl N-[2-[7-[2-[(2-fluorophenoxy)methyl]-4-(6-methyl-2-pyridyl)- 1H-imidazol-5-yl]quinoxalin-2-yl]oxyethyl]carbamate (C3.1-d) (150 mg, 263 umol, 1.0 equiv) in HCl/EtOAc (4 M, 10 mL) was stirred at 20°C for 1h. The reaction mixture was concentrated under reduced pressure to give a residue. Compound 2-[7-[2-[(2- fluorophenoxy) methyl]-4-(6- methyl-2-pyridyl)-1H-imidazol -5-yl]quinoxalin-2-yl] oxyethanamine (Compound C3.1) (74 mg, 120 umol, 45.67% yield, HCl salt) was obtained as a yellow solid. 1H NMR (MeOD, 400MHz) δ8.67 (s, 1H), 8.17 (d, J = 8.4 Hz, 1H), 8.14 (s, 1H), 8.10 (t, J = 8.0 Hz, 1H), 7.81 (d, J = 8.4 Hz, 1H), 7.65 (d, J = 7.6 Hz, 1H), 7.59 (d, J = 8.0 Hz, 1H), 7.36-7.31 (m, 1H), 7.21-7.14 (m, 2H), 7.09-7.02 (m, 1H), 5.45 (s, 2H), 4.80-4.75 (m, 2H), 3.52- 3.46(m, 2H), 2.80 (s, 3H) HPLC: 94.71% (220 nm), 96.21% (254 nm) MS (ESI): mass calcd. For C26H23FN6O2470.19, m/z found 471.1 [M+H]+. Compounds C3.2 and C3.3, as illustrated in Table 18, could be prepared in a manner similar to that described for Compound C3.1 using appropriate intermediates according to Method C as described herein.
ALK5 INHIBITORS OF FORMULA (C-1)—SYNTHETIC METHOD D Synthesis of N-[[5-[3-(2-aminoethoxy)quinoxalin-6-yl]-4-(6-methyl-2-pyridyl)-1H-imidazol -2- yl]methyl]-5-fluoro-pyridin-2-amine (Compound C4.1)
Step 1: Preparation of tert-butyl N-[2-[7- [2-(dimethoxymethyl)- 4-(6-methyl-2-pyridyl)- 1H-imidazol-5-yl]quinoxalin-2-yl]oxyethyl]carbamate (Compound C4.1-a)
To a mixture of tert-butyl N-[2-[7-[2-(6-methyl-2-pyridyl)-2-oxo-acetyl]quinoxalin-2-yl] oxyethyl]carbamate (A2) (2 g, 4.58 mmol, 1.0 eq) and 2,2-dimethoxyacetaldehyde (954 mg, 9.16 mmol, 830 uL, 2.0 eq) in THF (20 mL) was added NH4OAc (1.77 g, 22.9 mmol, 5.0 eq), and then stirred at 35 °C for 12h. The reaction mixture was poured into ice-water (20 mL) and stirred for 10 min. The aqueous phase was extracted with ethyl acetate (20 mL x 2). The combined organic phase was washed with brine (20 mL), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=2/1 to 1/1) to give compound tert-butyl N-[2-[7- [2- (dimethoxymethyl)-4-(6-methyl-2-pyridyl)-1H-imidazol-5-yl]quinoxalin-2- yl]oxyethyl]carbamate (C4.1-a) (1.8 g, 3.46 mmol, 75.46% yield) as a yellow solid. 1H NMR (CDCl3, 400 MHz) δ8.47 (s, 1H), 8.16 (s, 1H), 8.02 (d, J = 8.4 Hz, 1H), 7.89 (d, J = 8.0 Hz, 1H), 7.46-7.42 (m, 1H), 7.38-7.31 (m, 1H), 7.02 (d, J = 7.6 Hz, 1H), 5.61 (s, 1H), 5.01 (s, 1H), 4.54 (t, J = 5.2 Hz, 2H), 3.65-3.60 (m, 2H), 3.50 (s, 6H), 2.60 (s, 3H), 1.46 (s, 9H) Step 2: Preparation of 5-[3- (2-aminoethoxy)quinoxalin-6-yl]-4-(6-methyl-2-pyridyl)- 1H-imidazole-2-carbaldehyde (Compound C4.1-b)
To a solution of tert-butyl N-[2-[7- [2-(dimethoxymethyl)-4-(6-methyl-2-pyridyl)-1H- imidazol-5-yl]quinoxalin-2-yl]oxyethyl]carbamate (C4.1-a) (1.8 g, 3.46 mmol, 1.0 eq) in DCM (20 mL) was added TFA (5.12 mL, 20 eq) dropwise at 25°C and then stirred for 12 hours at this temperature. The mixture was concentrated in vacuum to give 5-(3-(2-aminoethoxy)quinoxalin- 6-yl)-4-(6-methylpyridin-2-yl)-1H-imidazole-2-carbaldehyde (C4.1-b) (1.5 g crude) as yellow oil. Step 3: Preparation of tert-butyl N-[2-[7-[2-formyl-4-(6-methyl-2-pyridyl)-1H-imidazol- 5-yl]quinoxalin-2-yl]oxyethyl]carbamate (Compound C4.1-c)
To a solution of 5-[3-(2-aminoethoxy)quinoxalin-6-yl]-4-(6-methyl-2-pyridyl) -1H- imidazole-2-carbaldehyde (C4.1-b) (1.3 g, 3.47 mmol, 1 eq) in THF (20 mL) was added NaHCO3 (584 mg, 6.94 mmol, 2 eq) in H2O (2 mL) and Boc2O (909 mg, 4.17 mmol, 957 uL, 1.2 eq) at 25°C, and then stirred at 25°C for 1 hour. The mixture was added water (50 mL) and extracted with ethyl acetate (30 mL x 3). The combined organic phase was washed with brine (30 mL), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The crude product was triturated with MTBE (20 mL) at 25oC for 30 min. Compound tert-butyl N-[2-[7- [2- formyl-4- (6-methyl-2-pyridyl)- 1H-imidazol-5-yl]quinoxalin-2-yl]oxyethyl]carbamate (C4.1-c) (1.4 g, 2.95 mmol, 84.97% yield) was obtained as a yellow solid. 1H NMR (CDCl3,400 MHz) δ9.86 (s, 1H), 8.52 (s, 1H), 8.17 (d, J = 1.2 Hz, 1H), 8.10 (d, J = 8.4 Hz, 1H), 7.91-7.84 (m, 1H), 7.51-7.45 (m, 1H), 7.38 (d, J = 7.6 Hz, 1H), 7.12 (d, J = 7.6 Hz, 1H), 4.56 (t, J = 5.2 Hz, 2H), 3.64-3.61 (m, 2H), 2.63 (s, 3H), 1.54 (s, 9H)
Step 4: Preparation of tert-butyl N-[2-[7-[2-[[(5-fluoro-2-pyridyl)amino]methyl]-4-(6- methyl-2-pyridyl)-1H-imidazol-5-yl]quinoxalin-2-yl]oxyethyl]carbamate (Compound C4.1-d)
To a solution of tert-butyl N-[2- [7-[2-formyl- 4-(6-methyl- 2-pyridyl)- 1H-imidazol- 5- yl]quinoxalin-2-yl]oxyethyl]carbamate (C4.1-c) (0.13 g, 273 umol, 1.0 eq) and 5-fluoropyridin- 2-amine (92.1 mg, 821 umol, 3.0 eq) in DCE (20 mL) was added AcOH (16.4 mg, 273 umol, 15.6 uL, 1.0 eq) at 25°C and then stirred at 70°C for 12 hours. The mixture was cooled to 0°C, MeOH (2 mL) and NaBH3CN (25.8 mg, 410 umol, 1.5 eq) were added, and it was stirred at 0°C for 1 hour. The pH of the reaction mixture was adjusted to ~7 with aq NaHCO3, and then extracted with DCM (30 mL x 3). The combined organic phase was washed with brine (30 mL), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by prep-HPLC (column: Phenomenex luna C18100 x 40mm x 5 um; mobile phase: [water (0.1%TFA)-ACN]; B%: 25%-38%, 8 min). Compound tert-butyl N-[2-[7-[2-[[(5-fluoro-2- pyridyl)amino]methyl]-4-(6-methyl-2-pyridyl)-1H-imidazol-5-yl]quinoxalin-2- yl]oxyethyl]carbamate (C4.1-d) (0.1 g, 175 umol, 63.97% yield) was obtained as a yellow solid. 1H NMR (MeOH, 400 MHz) δ8.56 (s, 1H), 8.10 (d, J = 8.4 Hz, 1H), 8.04 (s, 1H), 7.92- 7.94 (m, 1H), 7.74-7.67 (m, 2H), 7.42-7.34 (m, 2H), 7.27 (d, J = 7.6 Hz, 1H), 6.75 (dd, J = 3.2, 9.4 Hz, 1H), 4.87 (s, 2H), 4.53 (t, J = 5.4 Hz, 2H), 3.52 (t, J = 5.4 Hz, 2H), 2.64 (s, 3H), 1.41 (s, 9H)
Step 5: Preparation of N-[[5-[3-(2-aminoethoxy)quinoxalin-6-yl]-4-(6-methyl-2-pyridyl)- 1H-imidazol-2-yl]methyl]-5-fluoro-pyridin-2-amine (Compound C4.1)
To a solution of tert-butyl N-[2 [7-[2- [[(5-fluoro- 2-pyridyl) amino]methyl]- 4-(6- methyl-2-pyridyl)-1H-imidazol-5-yl]quinoxalin-2-yl]oxyethyl]carbamate (C4.1-d) (0.1 g, 175 umol, 1.0 eq) in EtOAc (10 mL) was added HCl/EtOAc (4 M, 10 mL) at 25°C and then stirred for 1 hour. The mixture was concentrated under reduced pressure at 45°C. Compound N-[[5-[3- (2-aminoethoxy)quinoxalin-6-yl]-4-(6-methyl-2-pyridyl)-1H-imidazol -2-yl]methyl]-5-fluoro- pyridin-2-amine (Compound C4.1) (56 mg, 116 umol, 66.67% yield, 98.16% purity, 4HCl) was obtained as a yellow solid. 1H NMR (MeOD, 400 MHz) δ8.65 (s, 1H), 8.16 (d, J = 8.6 Hz, 1H), 8.11 (d, J = 1.8 Hz, 1H), 8.01-8.03 (m, 1H), 7.96 (t, J = 8.0 Hz, 1H), 7.75-7.79 (m, 2H), 7.54 (d, J = 8.0 Hz, 1H), 7.47 (d, J = 8.0 Hz, 1H), 7.11 (dd, J = 4.0, 9.8 Hz, 1H), 4.93 (s, 2H), 4.76 (t, J = 4.8 Hz, 1H), 3.48-3.50 (m, 2H), 2.75 (s, 3H) HPLC: 98.16% (220 nm), 99.39% (254 nm) MS (ESI): mass calcd. For C25H23FN8O 470.20, m/z found 471.1 [M+H]+. Compounds C4.2–C4.9, as illustrated in Table 19, could be prepared in a manner similar to that described for Compound C4.1 using appropriate intermediates according to Method D as described herein.
ALK5 INHIBITOROF FORMULA (C-1)—SYNTHETIC METHOD E Synthesis of 2-[7-[2-tert-butyl-4-(6-methyl-2-pyridyl)-1H-imidazol-5-yl]quinoxalin-2- yl] oxyethanamine (Compound C5.1)
Synthetic Scheme
imidazol-5-yl] quinoxalin-2-yl] oxyethyl] carbamate (Compound C5.1-a)
To a solution of tert-butyl N-[2-[7-[2-(6-methyl-2-pyridyl)-2-oxo-acetyl] quinoxalin-2- yl] oxyethyl]carbamate (A2) (150 mg, 344 umol, 1.0 eq) in THF (9 mL) was added 2,2- dimethylpropanal (59.2 mg, 687 umol, 76.0 uL, 2.0 eq) and NH4OAc (132 mg, 1.72 mmol, 5.0 eq), and then stirred at 35°C for 12 hr. The mixture was poured into ice-water (w/w = 1/1) (20 mL). The aqueous phase was extracted with ethyl acetate (25 mL*3). The combined organic phase was washed with brine (25 mL), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 1/1). Compound tert-butyl N-[2-[7-[2-tert-butyl-4-(6-methyl-2-pyridyl)-1H- imidazol-5-yl] quinoxalin-2-yl] oxyethyl] carbamate (C5.1-a) (115 mg, 229 umol, 66.6% yield) was obtained as yellow oil.
1H NMR (MeOD, 400 MHz) δ8.44 (s, 1H), 7.98 (d, J = 1.6 Hz, 1H), 7.91 (d, J = 8.4 Hz, 1H), 7.71-7.57 (m, 2H), 7.29-7.17 (m, 2H), 4.52 (t, J = 5.6 Hz, 2H), 3.51 (t, J = 5.6 Hz, 2H), 2.54 (s, 3H), 1.49 (s, 9H), 1.42 (s, 9H) Step 2: Preparation of 2-[7-[2-tert-butyl-4-(6-methyl-2-pyridyl)-1H-imidazol-5-yl] quinoxalin-2-yl] oxyethanamine (Compound C5.1)
To a solution of tert-butyl N-[2-[7-[2-tert-butyl-4-(6-methyl-2-pyridyl)-1H-imidazol-5- yl] quinoxalin-2-yl]oxyethyl]carbamate (C5.1-a) (115 mg, 229 umol, 1.0 eq) in EtOAc (5 mL) was added HCl/EtOAc (4 M, 20 mL), and then stirred at 25°C for 30 min. The mixture was concentrated in vacuum. The residue was purified by Prep-HPLC (column: Phenomenex Synergi C18150*25*10um; mobile phase: [water (0.1%TFA)-ACN]; B%: 1%-30%, 8min). Compound 2-[7-[2-tert-butyl-4-(6-methyl-2-pyridyl)-1H-imidazol-5-yl] quinoxalin-2-yl] oxyethanamine (Compound C5.1) (37.1 mg, 42.0 umol, 18.4% yield, 97.2% purity, 4TFA) was obtained as yellow oil. 1H NMR (MeOD, 400MHz) δ8.65 (s, 1H), 8.14 (d, J = 8.8 Hz, 1H), 8.10 (d, J = 1.6 Hz, 1H), 7.78-7.70 (m, 2H), 7.39 (d, J = 8.0 Hz, 1H), 7.27 (d, J = 8.0 Hz, 1H), 4.75 (t, J = 5.6 Hz, 2H), 3.48 (t, J = 5.6 Hz, 2H), 2.65 (s, 3H), 1.61 (s, 9H) HPLC: 97.18% (220 nm), 98.37% (254 nm) Compound C5.2, as illustrated in Table 20, could be prepared in a manner similar to that described for Compound C5.1 using appropriate intermediates according to Method E as described herein.
TABLE 20 COMPOUND C5.2
EXAMPLE 4 Preparation of ALK5 Inhibitors of Formula (D-I) SYNTHESIS OF COMPOUNDS OF FORMULA (D-I) ACCORDING TO METHOD A Example 4-1A Synthesis of a TFA salt of 4-((6-(2-(6-methylpyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinolin-3-yl)oxy)butan-1-amine (Compound D-3.1)
Step 1: Preparation of benzyl (4-((6-bromoquinolin-3-yl)oxy)butyl)carbamate
To a solution of 6-bromoquinolin-3-ol (500.0 mg, 2.2 mmol, 1.0 equiv.) in tetrahydrofuran (10.0 mL) with an inert atmosphere of nitrogen, was added benzyl N-(4- hydroxybutyl)carbamate (491.2 mg, 2.2 mmol, 1.0 equiv.), triphenylphosphine (864.6 mg, 3.3 mmol, 1.5 equiv.), diisopropyl azodicarboxylate (666.6 mg, 3.3 mmol, 1.5 equiv.) with stirring at 0°C. The resulting solution was stirred for 5 h at 50°C. The reaction mixture was cooled to 25°C with a water/ice bath, diluted with 100 mL of water and extracted with 3x100 mL of dichloromethane and the organic layers were combined, washed with 3x100 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm. 400 mg (42%) of benzyl (4-((6-bromoquinolin- 3-yl)oxy)butyl)carbamate was obtained as a white solid. MS m/z [M+H]+ (ESI): 429. Step 2: Preparation of (4-((6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinolin-3- yl)oxy)butyl)carbamate
To a solution of benzyl benzyl (4-((6-bromoquinolin-3-yl)oxy)butyl)carbamate (100.0 mg, 0.23 mmol, 1.0 equiv.) in 1,4-dioxane (4.0 mL) with an inert atmosphere of nitrogen, was added bis(pinacolato)diboron (71.1 mg, 0.28 mmol, 1.2 equiv.), [1,1'-bis(diphenylphosphino) ferrocene]dichloropalladium(II) (33.6 mg, 0.046 mmol, 0.2 equiv.) and potassium acetate (67.6 mg, 0.69 mmol, 3.0 equiv.). The resulting solution was stirred for 2 h at 110°C. The reaction
mixture was cooled to 25°C with a water/ice bath and diluted with 20 mL of water. The resulting mixture was extracted with ethyl acetate 3x 20 mL and the organic layers were combined, washed with 3x20 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product without further purification was used at next step directly. 85 mg (77%) of (4-((6-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)quinolin-3-yl)oxy)butyl)carbamate was obtained as a yellow solid. MS m/z [M+H]+ (ESI): 477. Step 3: Preparation of ethyl 3-(6-methylpyridin-2-yl)-3-oxopropanoate
To a solution of methyl 6-methylpyridine-2-carboxylate (3.0 g, 19.8 mmol, 1.0 equiv.) in ethyl acetate (45.0 mL) with an inert atmosphere of nitrogen, was added sodium hydride (1.6 g, 39.6 mmol, 2.0 equiv.). The resulting solution was stirred for 12 h at 55°C and then quenched by the addition of ice/water. The resulting solution was extracted with 2x50 mL of ethyl acetate and the organic layers were combined and dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm. 2.4 g (58%) of ethyl 3-(6-methylpyridin-2-yl)-3-oxopropanoate was obtained as a colorless oil.MS m/z [M+H]+ (ESI): 208. Step 4: Preparation of ethyl (E)-3-(6-methylpyridin-2-yl)-3-((2-oxopyrrolidin-1- yl)imino)propanoate
To a solution of ethyl 3-(6-methylpyridin-2-yl)-3-oxopropanoate (1.5 g, 7.2 mmol, 1.0 equiv.) in pyridine (30.00 mL) with an inert atmosphere of nitrogen, was added 1- aminopyrrolidin-2-one hydrochloride (1.2 g, 8.6 mmol, 1.2 equiv.). The resulting solution was stirred for 20 h at 25°C. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 25 min); Detector, UV 254 nm. 1.4 g (67%) of ethyl (E)-3-(6- methylpyridin-2-yl)-3-((2-oxopyrrolidin-1-yl)imino)propanoate was obtained as a white oil. MS m/z [M+H]+ (ESI): 290. Step 5: Preparation of 2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole-3- carboxylic acid
To a solution of ethyl (E)-3-(6-methylpyridin-2-yl)-3-((2-oxopyrrolidin-1-yl)imino) propanoate (1.0 g, 3.5 mmol, 1.0 equiv.) in toluene (20.0 mL) with an inert atmosphere of nitrogen, was added sodium ethoxide (476.0 mg, 7.0 mmol, 2.0 equiv.). The resulting solution was stirred for 24 h at 80°C. LCMS showed the reaction was completed. It was followed by the addition of 1mol/L hydrochloric acid dropwise with stirring adjust pH to 4. The resulting solution was stirred for 3 h at 25oC. The pH of the reaction was adjusted to pH 8 with saturated aqueous sodium bicarbonate solution. The resulting solution was diluted with 100 mL of water and extracted with 3x200 mL of dichloromethane and the organic layers were combined and dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm. 520 mg (62%) of 2-(6-methylpyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazole-3-carboxylic acid was obtained as a yellow oil. MS m/z [M+H]+ (ESI): 244.
Step 6: Preparation of 3-bromo-2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole
To a solution of 2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole-3- carboxylic acid (200.0 mg, 0.82 mmol, 1.0 equiv.) in N,N-dimethylformamide (5.0 mL) with an inert atmosphere of nitrogen, was added N-bromosuccinimide (445.0 mg, 2.5 mmol, 3.0 equiv.). The resulting solution was stirred for 16 h at 25°C. The resulting solution was diluted with 20 mL of water and extracted with 3x20 mL of dichloromethane and the organic layers were combined, washed with 3x20 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by Flash-Prep- HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm. 170 mg (75%) of 3-bromo-2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole was obtained as a yellow solid. MS m/z [M+H]+ (ESI): 278. Step 7: Preparation of benzyl (4-((6-(2-(6-methylpyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinolin-3-yl)oxy)butyl)carbamate
To a solution of 3-bromo-2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b] pyrazole (100.0 mg, 0.36 mmol, 1.0 equiv.) in 1,4-dioxane/water (5.0 mL/0.5 mL) with an inert atmosphere of nitrogen, was added (4-((6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinolin- 3-yl)oxy)butyl)carbamate (205.0 mg, 0.43 mmol, 1.2 equiv.), [1,1'-bis(diphenylphosphino) ferrocene]dichloropalladium(II) (52.6 mg, 0.072 mmol, 0.2 equiv.) and sodium carbonate (116.6 mg, 1.1 mmol, 3.0 equiv.). The resulting solution was stirred for 4 h at 80oC. The reaction
mixture was cooled to 25 °C with a water/ice bath and diluted with 10 mL of water and extracted with 3x10 mL of dichloromethane and the organic layers were combined, washed with 3x10 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm. 110 mg (56%) of benzyl (4-((6-(2- (6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)quinolin-3-yl)oxy)butyl) carbamate was obtained as a yellow solid. MS m/z [M+H]+ (ESI): 548. Step 8: Preparation of a TFA salt of 4-((6-(2-(6-methylpyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinolin-3-yl)oxy)butan-1-amine (Compound D-3.1
To a solution of benzyl (4-((6-(2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3- yl)quinolin-3-yl)oxy)butyl)carbamate (60.0 mg, 0.11 mmol, 1.0 equiv.) trifluoroacetic acid (2.0 mL) with inert atmosphere of nitrogen, the resulting solution was stirred for 4 h at 50°C. The reaction mixture was cooled to 25°C with a water/ice bath, diluted with 10 mL of dichloromethane and concentrated under vacuum. The crude product was purified by Flash-Prep- HPLC with the following conditions (IntelFlash-1): C18 Column; mobile phase, water (with 0.5% trifluoroacetic acid) and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm. 19 mg (43%) of 4-((6-(2-(6-methylpyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinolin-3-yl)oxy)butan-1-amine TFA salt (Compound D-3.1) was obtained as a yellow solid. MS m/z [M+H]+ (ESI): 414. 1H NMR (300 MHz, Methanol-d4) δ: 1.87-1.96 (m, 4H), 2.46 (s, 3H), 2.67-2.83 (m, 2H), 2.95-3.00 (m, 2H), 3.10-3.20 (m, 2H), 4.18- 4.22 (m, 2H), 4.27-4.35 (m, 2H), 7.25-7.39 (m, 2H), 7.40-7.54 (m, 1H), 7.62-7.68 (m, 1H), 7.69- 7.82 (m, 2H), 7.83-7.85 (m, 1H), 8.50 (s, 1H).
Example 4-1B Synthesis of a TFA salt of N-(2-aminoethyl)-6-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro- 4H-pyrrolo[1,2-b]pyrazol-3-yl)quinoline-3-carboxamide (Compound D-3.15)
Compound D-3.15 was prepared according to synthetic Method A for compounds of Formula (D-1) with the appropriate starting materials, as shown below. Step 1: Preparation of benzyl (2-(6-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinoline-3-carboxamido)ethyl)carbamate
To solution of 3-bromo-2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazole (160.0 mg, 0.5 mmol, 1.0 equiv.) in 1,4-dioxane/water (5.0 mL/0.5 mL) with an inert atmosphere of nitrogen, was added benzyl (2-(6-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)quinoline-3-carboxamido)ethyl)carbamate (285.2 mg, 0.6 mmol, 1.2 equiv.), [1,1'-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (73.1 mg, 0.1 mmol, 0.2 equiv.) and sodium carbonate (159.0 mg, 1.5 mmol, 3.0 equiv.). The resulting solution was stirred for 10 h at 80oC. The reaction mixture was cooled to 25oC with a water/ice bath and diluted with 50 mL of water and extracted with 3x50 mL of dichloromethane and the organic layers were combined, washed with 3x50 mL of saturated sodium chloride, dried over anhydrous sodium sulfate,
filtered and concentrated under reduced pressure. The crude product was purified by Flash-Prep- HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm. 140 mg (47%) of benzyl (2-(6-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinoline-3-carboxamido)ethyl)carbamate was obtained as a yellow solid. MS m/z [M+H]+ (ESI): 583. Step 2: Preparation of N-(2-aminoethyl)-6-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinoline-3-carboxamide TFA salt (Compound D-3.15)
To a solution of benzyl (2-(6-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinoline-3-carboxamido)ethyl)carbamate (116.0 mg, 0.2 mmol, 1.0 equiv.) trifluoroacetic acid (2.0 mL) with inert atmosphere of nitrogen, the resulting solution was stirred for 4 h at 50oC. The reaction mixture was cooled to 25oC with a water/ice bath, diluted with 10 mL of dichloromethane and concentrated under vacuum. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): C18 Column; mobile phase, water (with 0.5% trifluoroacetic acid) and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm. 55 mg (62%) of N-(2-aminoethyl)-6-(2-(6-(difluoromethyl)pyridin- 2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)quinoline-3-carboxamide TFA salt (Compound D-3.15) was obtained as a yellow solid. MS m/z [M+H]+ (ESI): 449; 1H NMR (300 MHz, Methanol-d4) δ: 2.71-2.81 (m, 2H), 3.12-3.16 (m, 2H), 3.21-3.25 (m, 2H), 3.74-3.78 (m, 2H), 4.31 (t, J = 7.5 Hz, 2H), 6.19-6.57 (m, 1H), 7.59 (d, J = 6.9 Hz, 1H), 7.89-8.00 (m, 3H), 8.04-8.16 (m, 2H), 8.94 (d, J = 2.1 Hz, 1H), 9.30 (d, J = 2.1 Hz, 1H).
Example 4-1C Synthesis of a TFA salt of 2-(7-(2-(6-(trifluoromethyl)pyridin-2-yl)-5,6-dihydro-4H-pyrrolo [1,2-b]pyrazol-3-yl)quinoxalin-2-yl)ethan-1-amine (Compound D-3.22)
Compound 3.22 was prepared according to synthetic Method A for compounds of Formula (D-1) with the appropriate starting materials, as shown below. Step 1: Preparation of benzyl (2-(7-(2-(6-(trifluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinoxalin-2-yl)ethyl)carbamate To a solution of 3-bromo-2-(6-(trifluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazole (208 mg, 0.48 mmol, 1.2 equiv.) in 1,4-dioxane/water (5.0 mL/0.5 mL) with an inert atmosphere of nitrogen, was added benzyl (2-(7-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)quinoxalin-2-yl)ethyl)carbamate (130.0 mg, 0.4 mmol, 1.0 equiv.), [1,1'-Bis (diphenylphosphino)ferrocene]dichloropalladium(II) (58.0 mg, 0.08 mmol, 0.2 equiv.), sodium carbonate (127.2 mg, 1.2 mmol, 3 equiv.). The resulting solution was stirred for 6 h at 80oC. The reaction mixture was cooled to 25oC with a water/ice bath and diluted with 50 mL of water and extracted with 3x50 mL of dichloromethane and the organic layers were combined, washed with 3x50 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by Flash-Prep-HPLC with
the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm. 102 mg (47%) of benzyl (2-(7-(2-(6-(trifluoromethyl)pyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3- yl)quinoxalin-2-yl)ethyl)carbamate was obtained a yellow solid. MS m/z [M+H]+ (ESI): 559. Step 2: Preparation of 2-(7-(2-(6-(trifluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinoxalin-2-yl)ethan-1-amine TFA salt (Compound D-3.22)
To a solution of benzyl (2-(7-(2-(6-(trifluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)quinoxalin-2-yl)ethyl)carbamate (102.0 mg, 0.18 mmol, 1.0 equiv.) in trifluoroacetic acid (3.0 mL) with inert atmosphere of nitrogen, the resulting solution was stirred for 4 h at 50oC. The reaction mixture was cooled to 25oC with a water/ice bath, diluted with 20 mL of dichloromethane and concentrated under vacuum. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): C18 Column; mobile phase, water (with 0.5% trifluoroacetic acid) and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm. 55 mg (71%) of 2-(7-(2-(6-(trifluoromethyl)pyridin-2-yl)-5,6- dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)quinoxalin-2-yl)ethan-1-amine TFA salt (Compound D-3.22) was obtained as a yellow solid. MS m/z [M+H]+ (ESI): 425; 1H NMR (300 MHz, Methanol-d4) δ: 2.72-2.82 (m, 2H), 3.13-3.17 (m, 2H), 3.43-3.47 (m, 2H), 3.56-3.60 (m, 2H), 4.30-4.35 (m, 2H), 7.63-7.69 (m, 1H), 7.71-7.83 (m, 1H), 7.98-8.09 (m, 3H), 8.10-8.13 (m, 1H), 8.82 (s, 1H). Examples (4-1D)–(4-1V) The compounds in Table 21 were prepared according to synthetic Method A in a manner similar to that described within for Examples (4-1A)–(4-1C).
TABLE 21
SYNTHESIS OF COMPOUNDS OF FORMULA (D-I) ACCORDING TO METHOD B Example 4-2A Synthesis of a TFA salt of 4-((6-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)quinolin-3-yl)oxy) butan-1-amine (Compound D-4.1)
Compound D-4.1 was prepared according to synthetic Method B with the appropriate starting materials, as shown below. Step 1: Preparation of (E)-3-(dimethylamino)-1-(6-methylpyridin-2-yl)prop-2-en-1-one
To a solution of 1-(6-methylpyridin-2-yl)ethanone (270.0 mg, 2.0 mmol, 1.0 equiv.) in (dimethoxy- methyl)dimethylamine (262.2 mg, 2.2 mmol, 1.1 equiv.) with an inert atmosphere of nitrogen, The resulting solution was stirred for 7 h at 105oC. The reaction mixture was cooled to 25oC with a water/ice bath, diluted with 100 mL of water and extracted with 3x100 mL of dichloromethane and the organic layers were combined, washed with 3x100 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm. 305 mg (80%) of (E)-3-(dimethylamino)-1-(6- methylpyridin-2-yl)prop-2-en-1-one was obtained as a yellow solid. MS m/z [M+H]+ (ESI): 191.
Step 2: Preparation of 2-methyl-6-(1H-pyrazol-3-yl)pyridine
To a solution of (E)-3-(dimethylamino)-1-(6-methylpyridin-2-yl)prop-2-en-1-one (190.0 mg, 1.0 mmol, 1.0 equiv.) in ethanol (5.0 mL) with an inert atmosphere of nitrogen, was added hydrazine hydrate (2.0 mL). The resulting solution was stirred for 20 h at 25oC and concentrated under reduced pressure. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm. 150 mg (90%) of 2-methyl-6-(1H- pyrazol-3-yl)pyridine was obtained as a yellow solid. MS m/z [M+H]+ (ESI):160. Step 3: Preparation of 2-(4-bromo- pyrazol-3-yl)-6-methylpyridine
To a solution of 2-methyl-6-(1H-pyrazol-3-yl)pyridine (318 mg, 2.0 mmol, 1.0 equiv.) in dichloromethane (6.0 mL) with an inert atmosphere of nitrogen, was added N-bromosuccinimide (391.6 mg, 2.2 mmol, 1.1 equiv.). The resulting solution was stirred for 4 h at 25oC. The resulting solution was diluted with 20 mL of water and extracted with 3x20 mL of dichloromethane and the organic layers were combined, washed with 3x20 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm. 430 mg (90%) 2-(4-bromo-1H-pyrazol-3-yl)-6- methylpyridine was obtained as a yellow solid. MS m/z [M+H]+ (ESI): 238.
Step 4: Preparation of tert-butyl 4-bromo-3-(6-methylpyridin-2-yl)-1H-pyrazole-1-carboxylate
To a solution of 2-(4-bromo-1H-pyrazol-3-yl)-6-methylpyridine (474.0 mg, 2.0 mmol, 1.0 equiv.) in tetrahydrofuran (8.0 mL) with an inert atmosphere of nitrogen, was added di-tert- butyl pyrocarbonate (523.7 mg, 2.4 mmol, 1.2 equiv.), triethylamine (303.0 mg, 3.0 mmol, 1.5 equiv.). The resulting solution was stirred for 4 h at 25oC. The resulting solution was diluted with 50 mL of water and extracted with 3x50 mL of dichloromethane and the organic layers were combined, washed with 3x50 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm. 600 mg (89%) of tert-butyl 4-bromo-3-(6-methylpyridin-2-yl)-1H-pyrazole-1-carboxylate was obtained as a yellow solid. MS m/z [M+H]+ (ESI): 338. Step 5: Preparation of tert-butyl 4-(3-(4-(((benzyloxy)carbonyl)amino)butoxy)quinolin-6-yl)-3- (6-methylpyridin-2-yl)-1H-pyrazole-1-carboxylate
To a solution of tert-butyl 4-bromo-3-(6-methylpyridin-2-yl)-1H-pyrazole-1-carboxylate (202.0 mg, 0.6 mmol, 1.0 equiv.) in 1,4-dioxane/water (5.0 mL/0.5 mL) with an inert atmosphere of nitrogen, was added (4-((6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinolin-3- yl)oxy)butyl)carbamate (343.0 mg, 0.72 mmol, 1.2 equiv.), [1,1'-Bis(diphenylphosphino) ferrocene]dichloropalladium(II) (87.7 mg, 0.12 mmol, 0.2 equiv.), sodium carbonate (190.8 mg,
1.8 mmol, 3.0 equiv.). The resulting solution was stirred for 2 h at 80oC. The reaction mixture was cooled to 25oC with a water/ice bath and diluted with 20 mL of water and extracted with 3x20 mL of dichloromethane and the organic layers were combined, washed with 3x20 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm. 230 mg (63%) of tert-butyl 4-(3-(4- (((benzyloxy)carbonyl)amino)butoxy)quinolin-6-yl)-3-(6-methylpyridin-2-yl)-1H-pyrazole-1- carboxylate was obtained as a yellow solid. MS m/z [M+H]+ (ESI): 608. Step 6: Preparation of 4-((6-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)quinolin-3-yl)oxy)butan-
To a solution of (4-((6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinolin-3- yl)oxy)butyl)carbamate (200.0 mg, 0.33 mmol, 1.0 equiv.) in trifluoroacetic acid (5.0 mL) with an inert atmosphere of nitrogen, the resulting solution was stirred for 3 h at 50oC. The resulting solution was diluted with 10 mL of dichloromethane and concentrated under vacuum. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): C18 Column; mobile phase, water (with 0.5% trifluoroacetic acid) and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm. 80 mg (65%) of 4-((6-(3-(6-methylpyridin-2- yl)-1H-pyrazol-4-yl)quinolin-3-yl)oxy)butan-1-amine TFA salt was obtained as a yellow solid. MS m/z [M+H]+ (ESI): 374.1H NMR (300 MHz, Methanol-d4) δ: 1.91-1.99 (m, 4H), 2.82 (s, 3H), 3.04-3.09 (m, 2H), 4.23-4.27 (m, 2H), 7.56 (d, J = 8.7 Hz, 1H), 7.65 (d, J = 8.7 Hz, 1H), 7.74 (d, J = 7.8 Hz, 1H), 7.88-7.91 (m, 1H), 8.01-8.04 (m, 2H), 8.17-8.22 (m, 2H), 8.69 (s, 1H).SYNTHESIS OF COMPOUNDS OF FORMULA (D-I) ACCORDING TO METHOD C
Example 4-3A Synthesis of a TFA salt of 2-(4-(5-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)-2-fluorophenyl)-1H-pyrazol-1-yl)ethan-1-amine (Compound D-6.3)
Compound D-6.3 was prepared according to synthetic Method C with the appropriate starting materials, as shown below. Step 1: Preparation of (2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b] pyrazol-3-yl)boronic acid
To a solution of 3-bromo-2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazole (100.0 mg, 0.3 mmol, 1.0 equiv.) in tetrahydrofuran (10.0 mL) with an inert atmosphere of nitrogen, was added 2-isopropoxy-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (296.2 mg, 1.6 mmol, 5.0 equiv.). The reaction mixture was cooled to -78oC by the addition of n- butyllithium (51.0 mg, 0.8 mmol, 2.5 equiv.) to the reaction at -78oC. The resulting solution was stirred for 3 h at -78oC. The reaction was then quenched by the addition of water/ice. The resulting solution was diluted with 50 mL of water and extracted with 3x50 mL of dichloromethane and the organic layers were combined, washed with 3x50 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by Flash-Prep-HPLC with the following conditions
(IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm. 60 mg (68%) of (2-(6-(difluoromethyl)pyridin- 2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)boronic acid was obtained as a white solid. MS m/z [M+H]+ (ESI): 280. Step 2: Preparation of benzyl (2-(4-(5-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)-2-fluorophenyl)-1H-pyrazol-1-yl)ethyl)carbamate
To a solution of benzyl (2-(4-(5-bromo-2-fluorophenyl)-1H-pyrazol-1-yl)ethyl)carbamate (220.0 mg, 0.5 mmol, 1.0 equiv.) in 1,2-dimethoxyethane (5.0 mL) with an inert atmosphere of nitrogen, was added (2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol- 3-yl)boronic acid (220.2 mg, 0.8 mmol, 1.5 equiv.), Tetrakis(triphenylphosphine)palladium (60.8 mg, 0.05 mmol, 0.1 equiv.), Cesium fluoride (159.8 mg, 1.1 mmol, 2.0 equiv.). The resulting solution was stirred for 2 h at 80℃. The reaction mixture was cooled to 25oC with a water/ice bath, diluted with 50 mL of water and extracted with 3x50 mL of dichloromethane and the organic layers were combined, washed with 3x50 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm. 174 mg (58%) of benzyl (2-(4-(5-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6- dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)-2-fluorophenyl)-1H-pyrazol-1-yl)ethyl)carbamate was obtained as a white solid. MS m/z [M+H]+ (ESI): 573.
Step 3: Preparation of a TFA salt of 2-(4-(5-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)-2-fluorophenyl)-1H-pyrazol-1-yl)ethan-1-amine (Compound D-6.3)
To a solution of benzyl (2-(4-(5-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)-2-fluorophenyl)-1H-pyrazol-1-yl)ethyl)carbamate (174.0 mg, 0.304 mmol, 1.0 equiv.) in trifluoroacetic acid (3 mL), the resulting solution was stirred for 4 h at 50℃. The reaction mixture was cooled to 25oC with a water/ice bath, diluted with 10 mL of dichloromethane and concentrated under vacuum. The crude product was purified by Flash-Prep- HPLC with the following conditions (IntelFlash-1): C18 Column; mobile phase, water (with 0.5% trifluoroacetic acid) and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm. 87 mg (65%) of 2-(4-(5-(2-(6-(difluoromethyl)pyridin-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)-2-fluorophenyl)-1H-pyrazol-1-yl)ethan-1-amine TFA salt (Compound D-6.3) was obtained as a white solid. MS m/z [M+H]+ (ESI): 439; 1H NMR (400 MHz, Methanol-d4) δ: 2.69-2.77 (m, 2H), 3.05-3.09 (m, 2H), 3.47 (t, J = 5.6 Hz, 2H), 4.27 (t, J = 7.2 Hz, 2H), 4.49 (t, J = 6.0 Hz, 2H), 6.35-6.62 (m, 1H), 7.09-7.20 (m, 2H), 7.57-7.61 (m, 2H), 7.75-7.77 (m, 1H), 7.86 (s, 1H), 7.93-7.97 (m, 1H), 8.06 (s, 1H). Examples (4-3B)–(4-3D) The compounds in Table 22 were prepared according to synthetic Method C in a manner similar to that described within for Examples 4-3A.
TABLE 22
SYNTHESIS OF COMPOUNDS OF FORMULA (D-I) ACCORDING TO METHOD D Example 4-4A Synthesis of 2-(4-(2-fluoro-5-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)-1H-pyrazol-1- yl)ethan-1-amine (Compound D-7.2)
Compound D-7.2 was prepared according to synthetic Method D with the appropriate starting materials, as shown below. Step 1: Preparation of 2-(2-(4-(2-fluoro-5-(3-(6-methylpyridin-2-yl pyrazol-4-yl)phenyl)-
1H-pyrazol-1-yl)ethyl)isoindoline-1,3-dione
To a solution of 2-(4-[2-fluoro-5-[3-(6-methylpyridin-2-yl)-1H-pyrazol-4- yl]phenyl]pyrazol-1-yl)ethanol (600.0 mg, 1.65 mmol, 1.0 equiv.) in tetrahydrofuran (10.0 mL) with an inert atmosphere of nitrogen, was added phthalimide (485.1 mg, 3.3 mmol, 2.0 equiv.), triphenyl phosphine (657.5 mg, 2.5 mmol, 1.5 equiv.), diisopropyl azodiformate (505.0 mg, 2.5 mmol, 1.5 equiv.) with stirring at 0oC. The resulting solution was stirred for 4 h at 25oC. The resulting solution was diluted with 50 mL of water and extracted with 3x50 mL of dichloromethane and the organic layers were combined, washed with 3x50 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced
pressure. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 80.0% in 20 min); Detector, UV 254 nm. 450 mg (55%) 2-(2-(4-(2-fluoro-5-(3-(6- methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)-1H-pyrazol-1-yl)ethyl)isoindoline-1,3-dione was obtained as a yellow solid. MS m/z [M+H]+ (ESI): 493. Step 2: Preparation of 2-(4-(2-fluoro-5-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)-1H- pyrazol-1-yl)ethan-1-amine
To a solution of 2-(2-(4-(2-fluoro-5-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)- 1H-pyrazol-1-yl)ethyl)isoindoline-1,3-dione (450.0 mg, 0.9 mmol, 1.0 equiv.) in ethanol (4.0 mL) with an inert atmosphere of nitrogen, was added hydrazine hydrate (2.0 mL). The resulting solution was stirred for 1 h at 70oC in an oil bath, cooled to 25oC with a water/ice bath and concentrated under reduced pressure. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 100.0% in 20 min); Detector, UV 254 nm. 240 mg (73%) of 2-(4-(2-fluoro-5-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)-1H-pyrazol-1-yl)ethan-1- amine (Compound D-7.2) was obtained as a white solid. MS m/z [M+H]+ (ESI): 363; 1H NMR (300 MHz, Methanol-d4) δ: 2.52 (s, 3H), 3.07-3.11 (m, 2H), 4.23 (t, J = 6.0 Hz, 2H), 7.05-7.17 (m, 2H), 7.21-7.27 (m, 2H), 7.59-7.67 (m, 2H), 7.74-7.82 (m, 2H), 8.01 (s, 1H). Examples (4-4B)–(4-4J) The compounds in Table 23 were prepared according to synthetic Method D in a manner similar to that described within for Example (4-4A.)
TABLE 23
SYNTHESIS OF COMPOUNDS OF FORMULA (D-I) ACCORDING TO METHOD E Example 4-5A Synthesis of a TFA salt of (S)-4-amino-5-((2-(4-(2-fluoro-5-(3-(6-methylpyridin-2-yl
pyrazol-4-yl)phenyl)-1H-pyrazol-1-yl)ethyl) -5-oxopentanoic acid (Compound D-7.14)
Compound D-7.14 was prepared according to synthetic Method E with the appropriate starting materials, as shown below.
Step 1: Preparation of methyl (S)-4-((tert-butoxycarbonyl)amino)-5-((2-(4-(2-fluoro-5-(3-(6- methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)-1H-pyrazol-1-yl)ethyl)amino)-5-oxopentanoate
To a solution of 2-(4-(2-fluoro-5-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)-1H- pyrazol-1-yl)ethan-1-amine (150.0 mg, 0.41 mmol, 1.0 equiv.) in tetrahydrofuran (3.0 mL) with an inert atmosphere of nitrogen, was added N,N-carbonyldiimidazole (79.4 mg, 0.49 mmol, 1.2 equiv.), methyl (4S)-4-(benzylcarbamoyl)-4-[(tert-butoxycarbonyl)amino]butanoate (107.0 mg, 0.41 mmol, 1.0 equiv.). The resulting solution was stirred for 6 h at 25oC. The resulting solution was diluted with 50 mL of water and extracted with 3x50 mL of dichloromethane and the organic layers were combined, washed with 3x50 mL of saturated sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, water and acetonitrile (10.0% acetonitrile up to 80.0% in 20 min); Detector, UV 254 nm. 120 mg (48%) of methyl (S)-4-((tert-butoxycarbonyl)amino)-5-((2-(4-(2- fluoro-5-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)-1H-pyrazol-1-yl)ethyl)amino)-5- oxopentanoate was obtained as a yellow solid. MS m/z [M+H]+ (ESI): 606.
Step 2: Preparation of a TFA salt of methyl (S)-4-amino-5-((2-(4-(2-fluoro-5-(3-(6- methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)-1H-pyrazol-1-yl)ethyl)amino)-5-oxopentanoate T
To a solution of methyl (S)-4-((tert-butoxycarbonyl)amino)-5-((2-(4-(2-fluoro-5-(3-(6- methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)-1H-pyrazol-1-yl)ethyl)amino)-5-oxopentanoate (138.0 mg, 0.23 mmol, 1.0 equiv.) in dichloromethane (4.0 mL) with an inert atmosphere of nitrogen, was added trifluoroacetic acid (2 mL). The resulting solution was stirred for 2 h at 25oC. The reaction mixture was diluted with 10 mL of dichloromethane and concentrated under vacuum. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): C18 Column; mobile phase, water (with 0.5% trifluoroacetic acid) and acetonitrile (10.0% acetonitrile up to 100.0% in 15 min); Detector, UV 254 nm. 102 mg (89%) of methyl (S)-4-amino-5-((2-(4-(2-fluoro-5-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)- 1H-pyrazol-1-yl)ethyl)amino)-5-oxopentanoate TFA salt (Compound D-7.13) was obtained as a yellow semi-solid. MS m/z [M+H]+ (ESI): 506; 1H NMR (400 MHz, Methanol-d4) δ: 2.02-2.09 (m, 2H), 2.35-2.39 (m, 2H), 2.78 (s, 3H), 3.59-3.64 (m, 4H), 3.82-3.87 (m, 2H), 4.33-4.41 (m, 2H), 7.20-7.22 (m, 2H), 7.53-7.66 (m, 1H), 7.70-7.72 (m, 2H), 7.88 (s, 1H), 8.02 (s, 1H), 8.10- 8.15 (m, 2H). Step 3: Preparation of (S)-4-amino-5-((2-(4-(2-fluoro-5-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4- yl)phenyl)-1H-pyrazol-1-yl)ethyl)amino)-5-oxopentanoic acid TFA salt (Compound D-7.14)
To a solution of Compound D-7.13 (190.0 mg, 0.38 mmol, 1.0 equiv.) in tetrahydrofuran/water (2.0 mL/2.0 mL) with an inert atmosphere of nitrogen, was added lithium hydroxide monohydrate (31.9 mg, 0.76 mmol, 2.0 equiv.). The resulting solution was stirred for 6 h at 25oC. The pH of the reaction was adjusted to 7 with trifluoroacetic acid. The resulting mixture was concentrated under vacuum and purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): C18 Column; mobile phase, water (with 0.5% trifluoroacetic acid) and acetonitrile (10.0% acetonitrile up to 50.0% in 15 min); Detector, UV 254 nm. 70 mg (38%) of (S)-4-amino-5-((2-(4-(2-fluoro-5-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)phenyl)-1H- pyrazol-1-yl)ethyl)amino)-5-oxopentanoic acid TFA salt (Compound D-7.14) was obtained as a yellow semi-solid. MS m/z [M+H]+ (ESI): 492; 1H NMR (400 MHz, Methanol-d4) δ: 2.02-2.09 (m, 2H), 2.36-2.39 (m, 2H), 2.79 (s, 3H), 3.61-3.65 (m, 1H), 3.79-3.87 (m, 2H), 4.39-4.43 (m, 2H), 7.19-7.22 (m, 2H), 7.55 (d, J = 8.0 Hz, 1H), 7.66-7.72 (m, 2H), 7.89 (s, 1H), 8.03 (s, 1H), 8.11-8.18 (m, 2H). Examples (4-5C)–(4-5D) The compounds in Table 24 were prepared according to synthetic Method E in a manner similar to that described within for Examples (4-5A) and (4-5B). TABLE 24
EXAMPLE 5 Preparation of 4-((2S,5S)-23-(((2R,3R,4R,5R,6R)-3-acetamido-4,5-dihydroxy-6- (hydroxymethyl)tetrahydro-2H-pyran-2-yl)oxy)-13,13-bis((3-((4-(((2R,3R,4R,5R,6R)-3- acetamido-4,5-dihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl)oxy)butyl)amino)-3- oxopropoxy)methyl)-5-isopropyl-4,7,11,18-tetraoxo-2-(3-ureidopropyl)-15-oxa-3,6,12,19- tetraazatricosanamido)benzyl ((5-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-4-(6-methylpyridin-2-yl)- 1H-imidazol-2-yl)methyl)(2,6-difluorophenethyl)carbamate (Compound 3.1).
The title compound was prepared according to Scheme 2.
Scheme 5-1
Step 1: Synthesis of 4-((S)-2-((S)-2-amino-3-methylbutanamido)-5- ureidopentanamido)benzyl ((5-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-4-(6-methylpyridin-2- yl)-1H-imidazol-2-yl)methyl)(2,6-difluorophenethyl)carbamate (Intermediate 3).
To a stirred solution of 1 N-((5-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-4-(6-methylpyridin-2- yl)-1H-imidazol-2-yl)methyl)-2-(2,6-difluorophenyl)ethan-1-amine (Compound 60, 199.5 mg, 0.448 mmol) in DMF (4 mL) were added Fmoc-vc-PAB-PNP (327 mg, 0.448 mmol) and DIEA (165 mg, 1.28 mmol). The mixture was stirred at 40°C for 12 h. Once the reaction was deemed complete by LCMS, the reaction mixture was cooled to RT and 10 equiv. DBU was added to the reaction mixture. After 1 h the deprotection of the Fmoc protecting group was deemed complete by LCMS and the crude reaction mixture was purified via direct injection on a RP-HPLC (Teledyne ISCO, C18Aq, 0Æ45% acetonitrile in water (0.1% TFA)). Pure fractions were pooled, frozen and lyophilized. The product was obtained as an off-white solid (63 mg, 14% yield). LCMS (M+H) = 851.9.
Step 2: Preparation of tert-butyl 3-[2-amino-3-(3-tert-butoxy-3-oxo-propoxy) -2-[(3-tert- butoxy-3-oxo-propoxy)methyl]propoxy]propanoate. To a mixture of 2-amino-2-(hydroxymethyl)propane-1,3-diol (30 g, 247.66 mmol, 35.71 mL, 1 eq) in DMSO (83 mL) was added dropwise NaOH (5 M, 4.96 mL, 0.1 eq) at 0-15°C over 5 min. After the addition was complete, the mixture was stirred at 0-15°C for 5 min, and then tert-butyl prop-2-enoate (126.97 g, 990.63 mmol, 143.79 mL, 4 eq) was added dropwise at 20°C. The resulting mixture was stirred at 15°C for 12 h. LCMS showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was dissolved in EtOAc (200 mL), quenched by addition H2O (200 mL), and extracted with EtOAc (200 mL x 3). The combined organic layers were washed with brine (300 mL x 2), dried over Na2SO4, filtered and concentrated under reduced pressure to give tert-butyl 3-[2-amino-3- (3-tert-butoxy-3-oxo-propoxy)-2 -[(3-tert-butoxy-3-oxo-propoxy)methyl]propoxy]propanoate (100 g, 197.77 mmol, 79.86% yield) as a colorless oil. 1H NMR (DMSO-d6, 400MHz) δ 3.55(t, J = 6.0 Hz, 6H), 3.21-3.17 (m, 5H), 2.40 (t, J = 6.0 Hz, 6H), 1.99 (s, 27H). Step 3: Preparation of benzyl 5-[[2-(3-tert-butoxy-3-oxo-propoxy)-1,1-bis[(3-tert- butoxy- 3-oxo-propoxy)methyl]ethyl]amino]-5-oxo-pentanoate. To a solution of 5-benzyloxy-5-oxo-pentanoic acid (4.40 g, 19.78 mmol, 1 eq) in DMF (20 mL) was added dropwise HATU (9.02 g, 23.73 mmol, 1.2 eq) at 15°C. After the addition was complete, the mixture was stirred at 15°C for 5 min and then tert-butyl 3-[2-amino-3-(3-tert- butoxy-3-oxo-propoxy)-2-[(3-tert-butoxy-3-oxo-propoxy)methyl]propoxy]propanoate (10 g, 19.78 mmol, 1 eq) and Et3N (3.00 g, 29.67 mmol, 4.13 mL, 1.5 eq) were added at 15°C. The resulting mixture was stirred at 15°C for 12 h. LCMS showed the reaction was complete. The reaction mixture was quenched by the addition or H2O (200 mL) at 0°C, and extracted with EtOAc (100 mL x 3). The combined organic layers were washed with brine (200 mL x 2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 4/1) to give benzyl 5-[[2-(3-tert-butoxy-3-oxo-propoxy)-1,1-bis[(3-tert-butoxy-3-oxo- propoxy)methyl]ethyl]amino]-5-oxo-pentanoate (10 g, crude) as a colorless oil. 1H NMR (DMSO-d6, 400MHz) δ 7.39-7.30 (m, 5H), 6.97 (s, 1H), 5.08 (s, 2H), 3.57-3.48 (m, 12H), 2.38
(t, J = 6.0 Hz, 6H), 2.36-2.31 (m, 2H), 2.10 (t, J= 7.2 Hz, 2H), 1.72 (q, J = 7.6 Hz, 2H), 1.39 (s, 27H). Step 4: Preparation of 3-[2-[(5-benzyloxy-5-oxo-pentanoyl)amino]-3-(2-carboxyethoxy)- 2-(2-carboxyethoxymethyl)propoxy]propanoic acid. To a solution of benzyl 5-[[2-(3-tert-butoxy-3-oxo-propoxy)-1,1-bis [(3-tert-butoxy-3- oxo-propoxy)methyl]ethyl]amino]-5-oxo-pentanoate (10 g, 14.09 mmol, 1 eq) in DCM (100 mL) was added TFA (15.40 g, 135.06 mmol, 10 mL, 9.59 eq). The mixture was stirred at 15°C for 12 h. LCMS showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was washed with petroleum ether (100 mL) to give 3-[2-[(5-benzyloxy-5-oxo-pentanoyl)amino] -3-(2-carboxyethoxy)-2-(2- carboxyethoxymethyl)propoxy]propanoic acid (7 g, 12.93 mmol, 91.76% yield) as a white solid. 1H NMR (DMSO-d6, 400MHz) δ 12.14 (br s, 3H), 7.56-7.25 (m, 5H), 6.98 (s, 1H), 5.08 (s, 2H), 3.61-3.47 (m, 12H), 2.41 (t, J = 6.4 Hz, 6H), 2.37-2.31 (m, 2H), 2.09 (t, J = 7.2 Hz, 2H), 1.77- 1.67 (m, 2H). Step 5: Preparation of benzyl 5-oxo-5-[[2-[3-oxo-3-(2,3,4,5,6-pentafluorophenoxy) propoxy]-1,1-bis[[3-oxo-3-(2,3,4,5,6-pentafluorophenoxy)propoxy]methyl]ethyl]amino] pentanoate. To a solution of 3-[2-[(5-benzyloxy-5-oxo-pentanoyl)amino]-3-(2-carboxyethoxy) -2-(2- carboxyethoxymethyl)propoxy]propanoic acid (2.5 g, 4.62 mmol, 1 eq) in DMF (40 mL) was added DIEA (4.77 g, 36.93 mmol, 6.43 mL, 8 eq) and (2,3,4,5,6-pentafluorophenyl)-2,2,2- trifluoroacetate (5.17 g, 18.47 mmol, 4 eq). The mixture was stirred at 15°C for 12 h. The reaction mixture was quenched by addition of satd. aq. NaHCO3 (50 mL) at 0°C, and was extracted with EtOAc (50 mL x 3). The combined organic layers were washed with satd. aq. NH4Cl (50 mL) and brine (100 m), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=5/1 to 3/1) to give benzyl 5-oxo-5-[[2-[3-oxo-3-(2,3,4,5,6- pentafluorophenoxy)propoxy]-1,1-bis[[3-oxo-3-(2,3,4,5,6- pentafluorophenoxy)propoxy]methyl]ethyl]amino]pentanoate (3.3 g, 3.17 mmol, 68.75% yield)
was obtained as a yellow solid. 1H NMR (DMSO-d6, 400MHz) δ 7.45-7.26 (m, 5H), 7.07 (s, 1H), 5.05 (s, 2H), 3.69 (t, J = 5.6 Hz, 6H), 3.63 (s, 5H), 3.02-2.92 (m, 6H), 2.33 (t, J = 7.6 Hz, 2H), 2.10 (t, J = 7.2 Hz, 2H), 1.71 (q, J = 7.2 Hz, 2H). Step 6: Preparation of 5-[[2-[3-[4-[(2R,3R,4R,5R,6R)-3-acetamido-4,5-diacetoxy-6- (acetoxymethyl)tetrahydropyran-2-yl]oxybutylamino]-3-oxo-propoxy]-1,1-bis[[3-[4- [(2R,3R,4R,5R,6R)-3-acetamido-4,5-diacetoxy-6-(acetoxymethyl)tetrahydropyran-2- yl]oxybutylamino]-3-oxo-propoxy]methyl]ethyl]amino]-5-oxo-pentanoic acid. To a solution of [5-acetamido-3,4-diacetoxy-6-[4-(benzyloxycarbonylamino) butoxy]tetrahydropyran-2-yl]methyl acetate (850.36 mg, 1.54 mmol, 3.2 eq) and benzyl 5-oxo-5- [[2-[3-oxo-3-(2,3,4,5,6-pentafluorophenoxy)propoxy]-1,1-bis[[3-oxo-3-(2,3,4,5,6- pentafluorophenoxy)propoxy]methyl]ethyl]amino]pentanoate (0.5 g, 480.91 umol, 1 eq) in THF (20 mL) was added Pd(OH)2/C (10%, 0.1g) under N2 atmosphere. The suspension was degassed and purged with H2 three times. The mixture was stirred under H2 (15 psi) at 15°C for 12 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, DCM/MeOH = 10/1 to 0/1) to give 5- [[2-[3-[4-[(2R,3R,4R,5R,6R)-3-acetamido-4,5-diacetoxy-6-(acetoxymethyl)tetrahydropyran-2- yl]oxybutylamino]-3-oxo-propoxy]-1,1-bis[[3-[4-[(2R,3R,4R,5R,6R)-3-acetamido-4,5- diacetoxy-6-(acetoxymethyl)tetrahydropyran-2-yl]oxybutylamino]-3-oxo- propoxy]methyl]ethyl]amino]-5-oxo-pentanoic acid (0.56 g, crude) as a white solid. 1H NMR (DMSO-d6, 400MHz) δ 7.98-7.68 (m, 6H), 7.03 (s, 1H), 5.75 (s, 1H), 5.21 (d, J = 3.2 Hz, 3H), 4.96 (dd, J = 3.6, 11.2 Hz, 3H), 4.48 (d, J = 8.4 Hz, 3H), 4.02 (s, 9H), 3.94-3.83 (m, 3H), 3.78- 3.65 (m, 4H), 3.59-3.49 (m, 13H), 3.46-3.36 (m, 4H), 3.03 (d, J = 6.0 Hz, 8H), 2.28 (t, J = 6.4 Hz, 6H), 2.22-2.14 (m, 3H), 2.10 (s, 11H), 1.99 (s, 9H), 1.89 (s, 9H), 1.77 (s, 9H), 1.71-1.61 (m, 3H), 1.53-1.33 (m, 14H).
Step 7: Preparation of (2,3,4,5,6-pentafluorophenyl) 5-[[2-[3-[4-[(2R,3R,4R,5R,6R)-3- acetamido-4,5-diacetoxy-6-(acetoxymethyl)tetrahydropyran-2-yl]oxybutylamino]-3-oxo- propoxy]-1,1-bis[[3-[4-[(2R,3R,4R,5R,6R)-3-acetamido-4,5-diacetoxy-6- (acetoxymethyl)tetrahydropyran-2-yl]oxybutylamino]-3-oxo-propoxy]methyl]ethyl] amino]-5-oxo-pentanoate. To a solution of 5-[[2-[3-[4-[(2R,3R,4R,5R,6R)-3-acetamido-4,5-diacetoxy-6- (acetoxymethyl)tetrahydropyran-2-yl]oxybutylamino]-3-oxo-propoxy]-1,1-bis[[3-[4- [(2R,3R,4R,5R,6R)-3-acetamido-4,5-diacetoxy-6-(acetoxymethyl)tetrahydropyran-2- yl]oxybutylamino]-3-oxo-propoxy]methyl]ethyl]amino]-5-oxo-pentanoic acid (1.5 g, 907.61 umol, 1 eq) in DCM (30 mL) was added Et3N (275.52 mg, 2.72 mmol, 378.99 uL, 3 eq) and (2,3,4,5,6-pentafluorophenyl)-2,2,2-trifluoroacetate (762.59 mg, 2.72 mmol, 3 eq). The mixture was stirred at 20°C for 2 h. The reaction mixture was quenched by addition of H2O (20 mL), and extracted with DCM (50 mL x 2). The combined organic layers were washed with brine (100 mL x 2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Ethyl acetate/ EtOH=1/1) to give (2,3,4,5,6-pentafluorophenyl) 5-[[2-[3-[4-[(2R,3R,4R,5R,6R)-3-acetamido-4,5-diacetoxy-6- (acetoxymethyl) tetrahydropyran-2-yl]oxybutylamino]-3-oxo-propoxy]-1,1-bis[[3-[4- [(2R,3R,4R,5R,6R)-3-acetamido-4,5-diacetoxy-6-(acetoxymethyl)tetrahydropyran-2- yl]oxybutylamino]-3-oxo-propoxy]methyl]ethyl]amino]-5-oxo-pentanoate (1.5 g, 824.75 umol, 90.87% yield) was obtained as a white solid. 1H NMR (DMSO-d6, 400MHz) δ 7.82 (d, J = 8.4 Hz, 6H), 7.16 (s, 1H), 5.22 (d, J = 2.8 Hz, 3H), 4.97 (dd, J = 2.8, 11.4 Hz, 3H), 4.49 (d, J = 8.4 Hz, 3H), 4.03 (s, 10H), 3.93-3.83 (m, 3H), 3.76-3.67 (m, 3H), 3.61-3.50 (m, 14H), 3.48-3.39 (m, 4H), 3.12-2.98 (m, 7H), 2.79 (t, J = 7.2 Hz, 2H), 2.30 (t, J = 6.0 Hz, 7H), 2.22 (t, J = 7.0 Hz, 2H), 2.11 (s, 9H), 2.00 (s, 10H), 1.90 (s, 9H), 1.78 (s, 9H), 1.52-1.35 (m, 14H); MS (ESI): mass calcd. for C78H112F5N7O36.1818.71, m/z found 910.0[M/2+H]+. Step 8: Preparation of Intermediate 4. To a solution of (2,3,4,5,6-pentafluorophenyl) 5-[[2-[3-[4-[(2R,3R,4R,5R,6R)-3- acetamido-4,5-diacetoxy-6- (acetoxymethyl) tetrahydropyran-2-yl]oxybutylamino]-3-oxo- propoxy]-1,1-bis[[3-[4-[(2R,3R,4R,5R,6R)-3-acetamido-4,5-diacetoxy-6-
(acetoxymethyl)tetrahydropyran-2-yl]oxybutylamino]-3-oxo-propoxy]methyl]ethyl]amino]-5- oxo-pentanoate (81 mg, 44.5 umol, 1.0 equiv) and 4-((S)-2-((S)-2-amino-3-methylbutanamido)- 5-ureidopentanamido)benzyl ((5-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-4-(6-methylpyridin-2-yl)- 1H-imidazol-2-yl)methyl)(2,6-difluorophenethyl)carbamate (Intermediate 3) (37.5 mg, 42.2 μmol, 1.0 equiv) in DMF (4 mL) was added DIEA (4.0 equiv) via syringe. The resultant solution was allowed to stir at ambient temperature for 60 min. Upon confirmation the reaction had proceeded to completion (by LCMS), the reaction mixture was purified directly via prep- HPLC (column: Teledyne RediSep Prep C18, 100 Å, 5 μM (length: 150 mm; ID: 20 mm); mobile phase: [water (0.1% TFA)-AcN (0.1% TFA)]; B%: 10%-80%, 30 min). Fractions containing the desired product were dried via lyophilization. Intermediate 4 (16.2 mg, 6.5 μmol, 15% yield) was obtained as an off white solid. MS (ESI): mass calcd. for C115H159F2N19O40 2485.6, m/z found 1243.8 [M/2+H]+. Step 9: Preparation of Compound 3.1. To a solution of Intermediate 4 (16.2 mg, 6.5 umol, 1.0 equiv) in MeOH (5 mL) was added NaOMe (6.9 mg, 0.13 mmol, 20 equiv), the mixture was stirred at 20°C for 1 h. LCMS showed the reaction was complete. The reaction mixture was adjusted to pH~5-6 using acetic acid, and then was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Teledyne RediSep Prep C18, 100 Å, 5 μM (length: 150 mm; ID: 20 mm); mobile phase: [water (0.1% TFA)-AcN (0.1% TFA)]; B%: 10%-60%, 30 min). Compound 4-((2S,5S)-21-(((2R,3R,4R,5R,6R)-3-acetamido-4,5-dihydroxy-6- (hydroxymethyl)tetrahydro-2H-pyran-2-yl)oxy)-13,13-bis((3-((2-(((3R,4R,5R,6R)-3-acetamido- 4,5-dihydroxy-6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl)oxy)ethyl)amino)-3- oxopropoxy)methyl)-5-isopropyl-4,7,11,18-tetraoxo-2-(3-ureidopropyl)-15-oxa-3,6,12,19- tetraazahenicosanamido)benzyl ((5-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-4-(6-methylpyridin-2-yl)- 1H-imidazol-2-yl)methyl)(2,6-difluorophenethyl)carbamate (6.3 mg, 2.9 μmol, 45% yield) was obtained as a white solid. MS (ESI): mass calcd. for C97H141F2N19O312107.3, m/z found 1054.6 [M/2+H]+.
The compounds in Table 25 were prepared in a manner similar to that described for Compound 3.1. TABLE 25
BIOLOGICAL EXAMPLE 1: In vitro ALK5 Inhibitor-GalNAc Conjugate Screening The resultant conjugates were tested via a cell reporter assay. HEK293 SBE-LUC reporter cells (i.e., containing luciferase under the control of the SMAD-binding element (SBE), a TGFβ responsive transcriptional element) transfected to stably express full length human ASGR1 were seeded in 96 well plates at about 4 x 105 cells/well in an assay media of MEM
containing 0.5% FBS, 1% NEAA, 1% NaPyr, and 1% Pen/Strep. ALK5 Inhibitor-GalNAc conjugates and vehicle controls were added to wells containing either HEK293 SBE-LUC or ASGR1-HEK293 SBE-LUC cells in a dose titration ranging from about 5 μM to about 0.06 nM. After 24 hours of culture at 37°C in a 5% CO2 environment, human TGFβ1 was added (PeproTech Inc.) to a final concentration of 1.6 ng/ml followed by an additional 18 hour incubation. Luciferase Steady Glo reagent (Promega Corporation) was added to each well, incubated for 10 minutes with shaking, and then luciferase activity was determined by measuring luminescence with an Envision Plate Reader (Perkin-Elmer Inc.). Using Prism Software v7.04 (GraphPad Inc.), data were fit with a four-parameter non-linear regression to calculate IC50 values. By way of background, the mammalian asialoglycoprotein receptor (ASGR) is located on the sinusoidal membrane of hepatocytes where it binds and endocytoses galactose-terminated glycoproteins, such as N-acetylgalactosamine (GalNAc) (Braun et al., J. Biol. Chem. 271:21160, 1996). GalNAc is a well-defined liver-targeted moiety having high affinity to ASGR1. HEK293 transfected with ASGR1 is known model for detecting uptake of GalNAc conjugates (see, e.g., Tanowitz et al., Nucleic Acid Res. 45:12388, 2017). The results for exposure of the ASGR1- positive cells (Conjugate X + ASGR1) and ASGR1-negative cells (Conjugate X) as compared to DMSO control are shown in Figures 1A (Conjugate 3.1), 1B (Conjugate 3.2), and 1C (Conjugate 3.3). These data indicate that GalNAc-ALK5 inhibitor conjugates are unable to be taken up by HEK cells that do not express ASGR1. In contrast, HEK cells expressing ASGR1 show increased uptake of the GalNAc-ALK5 inhibitor conjugates, with release of the ALK5 inhibitor and ALK5 activity inhibition, which is reflected by the reduced expression of the luciferase reporter that is under the control of the SMAD-binding element (SBE). BIOLOGICAL EXAMPLE 2: TGFE Reporter Assay TGFE/SMAD Signaling Pathway SBE reporter cell line are obtained from BPS Bioscience. Cells are passed, expanded, and stored in liquid nitrogen as per the supplier's instructions with the exception that growth media is changed to DMEM-C with Geneticin
(DMEM supplemented with 10% fetal bovine serum, 1X NEAA, 1 mM Pyruvate, 2 mM glutamine, 50 μg/mL penicillin, 50 U/mL streptomycin and 400 μg/mL of Geneticin). The assay media is MEM supplemented with 0.5% fetal bovine serum, 1X NEAA, 1mM Pyruvate, 50 μg/mL penicillin and 50 U/mL streptomycin. Compounds are assayed to measure their activity as ALK5 inhibitors. Enzyme Inhibition Assay ALK5 enzyme inhibition assays were performed by Reaction Biology Corp (Malvern, PA). 1 mg/mL of peptide subtrate (casein) and 10 uM ATP were prepared in a mixture of fresh reaction buffer. The kinase was delivered into the substrate solution which was gently mixed. Compounds in 100% DMSO were added to the kinase reaction mixture by Acoustic technology (Echo550; nanoliter range) and the mixture was incubated for 20 min at room temperature. 33P- ATP (Specific activity 10 uCi/uL) was added into the reaction mixture to initiate the reaction and the reaction mixture was incubated for 2 hours at room temperature. Radioactivity was detected by filter-binding method and kinase activity data were expressed as the percent remaining kinase activity in test samples compared to vehicle (dimethyl sulfoxide) reactions. IC50 values and curve fits were obtained using Prism (GraphPad Software). Results are reported in Table 26. TABLE 26
Table 26 Key: Compounds having an IC50 value between 0.1 nM and 50 nM are denoted 50 nM and 100 nM as +++, 100 nM and 1000 nM as ++, and 1000 nM to 10,000 nM as +.
The data in Table 26 shows that the ALK5 inhibitors of this disclosure are capable of diffusing into the cells and inhibiting ALK5 serine/threonine kinase activity at nanomolar and sub-nanomolar concentrations. TGFβ Reporter Assay The HEK293 SBE-LUC reporter cells from Example 1 were grown in DMEM-C with Geneticin (DMEM supplemented with 10% fetal bovine serum, 1X NEAA, 1mM Pyruvate, 2mM glutamine, 50 μg/mL penicillin, 50 U/mL streptomycin and 400 ug/mL of Geneticin). The assay media was MEM supplemented with 0.5% fetal bovine serum, 1X NEAA, 1 mM Pyruvate, 50 μg/mL penicillin and 50 U/mL streptomycin. The HEK293 SBE-LUC reporter cells were harvested from the tissue culture flasks by incubation in small quantity of Versene at room temperature for three to five minutes after the media in the flask is removed and cells rinsed with PBS. Cells were counted and diluted in the assay media at ~0.8 x 106 cells/mL then 50 uL/well were added to 96-well assay plate. Test samples (at desired concentrations diluted in assay media) were added to assay plate containing the 50 uL/well of cells (or media only), 50 uL per well, and incubated for 5-6 hours at 37°C in a 5% CO2 humidified incubator. After that time, 15 uL of TGFβ diluted to 12.5ng/mL in the assay media was added to the plate. Controls included TGFβ titration (from 25 to 0 ng/mL) without inhibitors, and media only (without cells, inhibitor or TGFβ). Plates were incubated at 37°C in a 5% CO2 humidified incubator for 18h. Luciferase substrate solution was subsequently added at 75uL per well, incubated in dark with shaking at room temperature for 10 min, and luminescence
Table 27 Key: Compounds having an IC50 value between 0.1 nM and 10 nM are denoted as ++++, 10 nM and 100 nM as +++, 100 nM and 1000 nM as ++, and 1000 nM to 10,000 nM as +. These data from the TGFbβ reporter assay demonstrate that ALK5 inhibitors are capable of diffusing into the cells and indirectly reducing expression of the luciferase reporter that is under the control of the SMAD-binding element (SBE) by inhibiting ALK5 activity. While aspects of the present disclosure have been shown and described herein, it will be apparent to those skilled in the art that such aspects are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the disclosure. It should be understood that various alternatives to the aspects of the disclosure described herein may be employed in practicing the disclosure. It is intended that the following claims define the scope of the disclosure and that methods and structures within the scope of these claims and their equivalents be covered thereby. The various embodiments described above can be combined to provide further embodiments. All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification and/or listed in the Application Data Sheet, including U.S. patent application Serial No. 62/912,783, filed October 9, 2019, and U.S. patent application Serial No. 63/047,252, filed July 2, 2020, are incorporated herein by reference, in their entirety. Aspects of the embodiments can be modified, if necessary to employ concepts of the various patents, applications and publications to provide yet further embodiments.
Claims
CLAIMS: 1. A conjugate of Formula (I):
wherein Region G comprises at least one GalNAc moiety; Region L3 is a connector that connects Region G to Region Inh; Region Inh comprises a TGFβR1 inhibitor; z is 1, 2, 3, 4, or 5; or a salt thereof. 2. The conjugate of claim 1, wherein the TGFβR1 inhibitor is a compound of Formula (A-I): wherein
one of M1 and M2 is
and the other of M1 and M2 is selected from:
R1 and R2 are, at each occurrence, independently selected from hydrogen, halogen, −OR11, −SR11, −N(R11)2, −NO2, −CN, phenyl, and −C1-C6 alkyl, wherein said −C1-C6 alkyl is optionally substituted with one or more substituents independently selected from halogen, −OR11, −SR11, −S(O)R10, −S(O)2R11, −S(O)2N(R11)2 −N(R11)2, −C(O)R10, −C(O)N(R11)2, −N(R11)C(O)R10, −C(O)OR11, −OC(O)R10, −NO2, and −CN;
R3 is, at each occurrence, independently selected from halogen, −C1-C3 alkyl, −C1-C3 haloalkyl, −OH, −NO2, −CN, −OC1-C3 alkyl, and −OC1-C3 haloalkyl; each R4 is, at each occurrence, independently selected from hydrogen and C1-C3 alkyl or two R4 join together with atoms to which they are attached to form a 5- or 6-membered heterocycle optionally substituted with one or more substituents independently selected from halogen, C1-C3 alkyl, −OH, OC1-C3 alkyl, and −OC1-C3 haloalkyl; R5 is hydrogen, halogen, −OR61, −SR61, −N(R61)2, −NO2, −CN, and −C1-C6 alkyl, wherein said −C1-C6 alkyl is optionally substituted with one or more substituents independently selected from halogen, −OR61, −SR61, −N(R61)2, −NO2, and −CN; R6 is, at each occurrence, independently selected from: halogen, −OR21, −SR21, −N(R21)2, −C(O)R20, −C(O)N(R21)2, −N(R21)C(O)R20 , −C(O)OR21, −OC(O)R21, −S(O)R20, −S(O)2R21, −S(O)2N(R21)2, −OC(O)OR21, −OC(O)N(R21)2, −NR21C(=O)OR21, −N(R21)C(O)N(R21)2, −NO2, and −CN; C1-C10 alkyl, C2-C10 alkenyl, and C2-C10 alkynyl, each of which is optionally substituted with one or more substituents independently selected from halogen, −OR21, −SR21, −N(R21)2, −C(O)R20, −C(O)N(R21)2, −N(R21)C(O)R20 , −C(O)OR21, −OC(O)R21, −S(O)R20, −S(O)2R21, −S(O)2N(R21)2, −OC(O)OR21, −OC(O)N(R21)2, −NR21C(=O)OR21, −N(R21)C(O)N(R21)2, −NO2, =O, =S, =N(R21), −CN, a C3-C10 carbocycle, and a 3- to 10- membered heterocycle wherein said C3-C10 carbocycle and said 3- to 10-membered heterocycle are optionally substituted with one or more substituents independently selected from RX; and a C3-C10 carbocycle and a 3- to 10-membered heterocycle, each of which is optionally substituted with one or more substituents independently selected from halogen, −OR20, −OH, −SR20, −SH, −N(R21)2, −C(O)R20, −C(O)N(R21)2, −N(R21)C(O)R20 , −C(O)OR21, −OC(O)R21, −S(O)R20, −S(O)2R21, −S(O)2N(R21)2, −OC(O)OR21, −OC(O)N(R21)2, −NR21C(=O)OR21, −N(R21)C(O)N(R21)2, −NO2, =O, =S, =N(R21), −CN, −C2-C6 alkenyl, −C2-C6 alkynyl and C1-C6 alkyl wherein said C1-C6 alkyl is optionally substituted with one or more substituents independently selected from RY; R7 and R8 are independently selected from hydrogen, halogen, C1-C3 alkyl, −OH, OC1-C3 alkyl, and −OC1-C3 haloalkyl, or R7 and R8 join together with the atoms to which they are attached to form a C5-C6 carbocycle or 5- or 6- membered heterocycle each of which is
optionally substituted with one or more substituents independently selected from halogen, −OR31, −SR31, −N(R31)2, −NO2, −CN and −C1-C6 alkyl wherein said C1-C6 alkyl is optionally substituted with one or more substituents independently selected from halogen, −OR31, −SR31, −N(R31)2, −NO2, and −CN; Y is selected from −O− and −N(R9)− and R9 is, at each occurrence, independently selected from: hydrogen; and −C1-C6 alkyl optionally substituted with one or more substituents independently selected from halogen, −OR41, −SR41, −S(O)R40, −S(O)2R41, −S(O)2N(R41)2, −N(R41)2, −C(O)R40, −C(O)N(R41)2, −N(R41)C(O)R40 , −C(O)OR41, −OC(O)R40, −NO2, and −CN; each R10, R20, and R40 is independently selected at each occurrence from: −C1-C10 alkyl, −C2-C10 alkenyl, and −C2-C10 alkynyl, each of which is optionally substituted with one or more substituents independently selected from RY; and a C3-C12 carbocycle and a 3- to 12-membered heterocycle, each of which is optionally substituted with one or more substituents independently selected from RX; each R11, R21, R31, R41, and R61 is independently selected at each occurrence from: hydrogen; −C1-C10 alkyl, −C2-C10 alkenyl, and −C2-C10 alkynyl, each of which is optionally substituted with one or more substituents independently selected from RY; and a C3-C12 carbocycle and a 3- to 12-membered heterocycle, each of which is optionally substituted with one or more substituents independently selected from RX, or two R11, R21, R31, R41, or R61 on the same N atom are taken together with the N atom to which they are attached to form a N-containing heterocycle optionally substituted with RX; each RX is independently selected at each occurrence from: halogen, −OR51, −SR51, −N(R51)2, −C(O)R50, −C(O)N(R51)2, −N(R51)C(O)R50 , −C(O)OR51, −OC(O)R51, −S(O)R50, −S(O)2R51, −S(O)2N(R51)2, −OC(O)OR51, −OC(O)N(R51)2, −NR51C(=O)OR51, −N(R51)C(O)N(R51)2, −NO2, =O, =S, =N(R51), −CN, −C2-C6 alkenyl, −C2-C6 alkynyl, and C1-C6 alkyl, wherein said C1-C6 alkyl is optionally substituted with one or more substituents independently selected from −OR51, −SR51, −N(R51)2, −C(O)R50, −C(O)N(R51)2,
−N(R51)C(O)R50 , −C(O)OR51, −OC(O)R51, −S(O)R50, −S(O)2R51, −S(O)2N(R51)2, −OC(O)OR51, −OC(O)N(R51)2, −NR51C(=O)OR51, −N(R51)C(O)N(R51)2, and =O; each RY is independently selected at each occurrence from: halogen, −OR51, −SR51, −N(R51)2, −C(O)R50, −C(O)N(R51)2, −N(R51)C(O)R50, −C(O)OR51, −OC(O)R51, −S(O)R50, −S(O)2R51, −S(O)2N(R51)2, −OC(O)OR51, −OC(O)N(R51)2, −NR51C(=O)OR51, −N(R51)C(O)N(R51)2, −NO2, =O, =S, =N(R51), and −CN; each R50 is independently selected at each occurrence from: −C1-C10 alkyl, −C2-C10 alkenyl, and −C2-C10 alkynyl, each of which is optionally substituted with one or more substituents independently selected from halogen, −OH, −CN, −NO2, −NH2, =O, =S, −O−C1-C10 alkyl, C3-C12 carbocycle, and a 3- to 12-membered heterocycle; and a C3-C12 carbocycle and a 3- to 12-membered heterocycle, each of which is optionally substituted with one or more substituents independently selected from halogen, −OH, −CN, −NO2, −NH2, =O, =S, −C1-C10 alkyl, −O−C1-C10 alkyl, and −C1-C10 haloalkyl; each R51 is independently selected at each occurrence from: hydrogen; −C1-C10 alkyl, −C2-C10 alkenyl, and −C2-C10 alkynyl, each of which is optionally substituted with one or more substituents independently selected from halogen, −OH, −CN, −NO2, −NH2, =O, =S, −O−C1-C10 alkyl, C3-C12 carbocycle, and a 3- to 12-membered heterocycle; and a C3-C12 carbocycle and a 3- to 12-membered heterocycle, each of which is optionally substituted with one or more substituents independently selected from halogen, −OH, −CN, −NO2, −NH2, =O, =S, −C1-C10 alkyl, −O−C1-C10 alkyl, and −C1-C10 haloalkyl; Z1, Z2, Z3, and Z4 are each independently selected from N or C(H); n is selected from 1, 2, and 3; m is 0, 1, or 2; s is selected from 0 and 1; and w is selected from 0, 1,
2, 3, 4, and 5; or a salt thereof.
5. The conjugate of claim 1, wherein the the TGFβR1 inhibitor is a compound selected from: Cmpd. Cmpd. Structure Structure No. No. 1 46 2 47 3 48 4 49
6. The conjugate of claim 1, wherein the TGFβR1 inhibitor is a compound of Formula (B-I):
wherein: M1 and M2 are independently selected from
and
; R1 and R2 are independently selected at each occurrence from: a halogen, −OR10, −SR10, −N(R10)2, −C(O)R10, −C(O)N(R10)2, −N(R10)C(O)R10 , −C(O)OR10, −OC(O)R10, −S(O)R10, −S(O)2R10, −S(O)2N(R10)2, −P(O)(OR10)2, −OP(O)(OR10)2, −NO2, and −CN; −C1-C10 alkyl, −C2-C10 alkenyl, and −C2-C10 alkynyl, each of which is optionally substituted at each occurrence with one or more substituents independently selected from a halogen, −OR10, −SR10, −N(R10)2, −C(O)R10, −C(O)N(R10)2, −N(R10)C(O)R10 , −C(O)OR10, −OC(O)R10, −S(O)R10, −S(O)2R10, −S(O)2N(R10)2, −P(O)(OR10)2, −OP(O)(OR10)2, −NO2, =O, =S, =N(R10), −CN, a C3-C10 carbocycle, and a 3- to 10-membered heterocycle; and a C3-C10 carbocycle and a 3- to 10-membered heterocycle, each of which is optionally substituted with one or more substituents independently selected from a halogen, −OR10, −SR10, −N(R10)2, −C(O)R10, −C(O)N(R10)2, −N(R10)C(O)R10 , −C(O)OR10, −OC(O)R10, −S(O)R10, −S(O)2R10, −S(O)2N(R10)2, −P(O)(OR10)2, −OP(O)(OR10)2, −NO2, =O, =S, =N(R10), −CN, −C1-C6 alkyl, −C2-C6 alkenyl, and −C2-C6 alkynyl; R3 is selected from hydrogen and −C1-C10 alkyl optionally substituted with one or more substituents independently selected from a halogen, −NO2, =O, =S, =N(R10), −CN, −OR10, −SR10, −N(R10)2, −C(O)R10, −C(O)N(R10)2, −N(R10)C(O)R10 , −C(O)OR10, and −OC(O)R10; n and m are independently selected from 0, 1, 2, 3, and 4;
Q is selected from a bond, −(CR10 2)p−, −(CR10 2)qC(=O)(CR10 2)q−, −(CR10 2)qC(=S)(CR10 2)q−, −(CR10 2)qC(=NR10)(CR10 2)q−, −(CR10 2)qO(CR10 2)q−, −(CR10 2)qS(CR10 2)q−, −(CR10 2)qN(R10)(CR10 2)q−, −(CR10 2)qOC(=O)O(CR10 2)q−, −(CR10 2)qC(=O)N(R10)(CR10 2)q−, −(CR10 2)qN(R10)C(=O)(CR10 2)q−, and −(CR10 2)qN(R10)SO2(CR10 2)q−; p is selected from 1, 2, 3, 4, and 5; q is independently selected at each occurrence from 0, 1, 2, 3, 4, and 5; T is selected from an optionally substituted saturated C3-C7 carbocycle, an optionally substituted C5-12 bicyclic carbocycle, and an optionally substituted 4- to 12-membered heterocycle, wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13; R13 is independently selected at each occurrence from: a halogen, −OR10, −SR10, −N(R10)2, −C(O)R10, −C(O)N(R10)2, −N(R10)C(O)R10 , −C(O)OR10, −OC(O)R10, −S(O)R10, −S(O)2R10, −S(O)2N(R10)2, −P(O)(OR10)2, −OP(O)(OR10)2, −NO2, =O, =S, =N(R10), and −CN; −C1-C10 alkyl, −C2-C10 alkenyl, and −C2-C10 alkynyl, each of which is optionally substituted at each occurrence with one or more substituents independently selected from a halogen, −OR10, −SR10, −N(R10)2, −C(O)R10, −C(O)N(R10)2, −N(R10)C(O)R10 , −C(O)OR10, −OC(O)R10, −S(O)R10, −S(O)2R10, −S(O)2N(R10)2, −P(O)(OR10)2, −OP(O)(OR10)2, −NO2, =O, =S, =N(R10), −CN, a C3-C10 carbocycle, and a 3- to 10-membered heterocycle; and a C3-C10 carbocycle and a 3- to 10-membered heterocycle, each of which is optionally substituted with one or more substituents independently selected from a halogen, −OR10, −SR10, −N(R10)2, −C(O)R10, −C(O)N(R10)2, −N(R10)C(O)R10 , −C(O)OR10, −OC(O)R10, −S(O)R10, −S(O)2R10, −S(O)2N(R10)2, −P(O)(OR10)2, −OP(O)(OR10)2, −NO2, =O, =S, =N(R10), −CN, −C1-C6 alkyl, −C2-C6 alkenyl, and −C2-C6 alkynyl; and R10 is independently selected at each occurrence from: hydrogen; −C1-C10 alkyl, −C2-C10 alkenyl, and −C2-C10 alkynyl, each of which is optionally substituted at each occurrence with one or more substituents independently selected
from a halogen, −OH, −CN, −NO2, −NH2, =O, =S, −O−C1-C10 alkyl, C3-C12 carbocycle, and a 3- to 12-membered heterocycle; and a C3-C12 carbocycle and a 3- to 12-membered heterocycle, each of which is optionally substituted at each occurrence with one or more substituents independently selected from a halogen, −OH, −CN, −NO2, −NH2, =O, =S, −C1-C10 alkyl, −O−C1-C10 alkyl, and −C1-C10 haloalkyl; or a salt thereof.
8. The conjugate of claim 6 or claim 7, wherein: Q is selected from −CH2−, −CH2NH−, −CH2NHCH2−, and −CH2NHCH2CH2−;
T is selected from: ,
wherein T is optionally substituted with one or more substituents independently selected at each occurrence from R13, wherein R13 is independently selected at each occurrence from halogen, −OH, −NH2, and −C1-C3 alkyl.
10. The conjugate of claim 1, wherein the TGFβR1 inhibitor is a compound of Formula (C-I):
(C-I) or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, tautomer, isotope, or salt thereof, wherein: L is −[CR′2]p−L’−[CH2]q− L’ is absent, −S−, −O−, or −NH−; A is absent, carbocycle, or heterocycle; Q3 is N or CR3;
Q4 is N or CR4; Q5 is N or CR5; Q6 is N or CR6; R1 is hydrogen, C1–3 alkyl, or C1–3 haloalkyl; R2 is, at each occurrence, independently halo, C1–3 alkyl, C1-3 alkoxy, C1–3 haloalkyl, or C1-3 haloalkoxy; R3 is H, halo, C1–3alkyl, C1–3haloalkyl, or C1–3 alkoxy; R4 is H, halo, C1–3 alkyl, C1–3haloalkyl, or C1–3 alkoxy; R5 is H, halo, C1–3 alkyl, C1–3haloalkyl, or C1–3 alkoxy; R6 is H, halo, C1–3 alkyl, C1–3haloalkyl, or C1–3 alkoxy; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to a GalNAc moiety; R9 is, at each occurrence, independently halo, −OR′, −SR′, −N(R′)2, −C(O)R′, −C(O)N(R′)2, −N(R′)C(O)R′, −C(O)OR′, −OC(O)R′, −S(O)R′, −S(O)2R′, −S(O)2OR′, −P(O)(OR′)2, −OP(O)(OR′)2, −NO2, =O, −CN, C1–4 alkyl, C2–5 alkenyl, C2–5 alkynyl, C1-4 haloalkyl, C2–5 haloalkenyl, C2–5 haloalkynyl, carbocycle, or heterocycle; wherein R1, R2, R3, R4, R5, R6, and R9 are each substituted with 0–3 R10; R10 is, at each occurrence, independently C1–3 alkoxy or C1–3 haloalkoxy; R′ is, at each occurrence, independently hydrogen, C1–6 alkyl, C2–6 alkenyl, C2–6 alkynyl, or C1–6 haloalkyl; m is 0–3; n is 0–5; p is 1-3; and q is 0-3.
11. The conjugate of claim 10, wherein the compound of Formula (C-I) is a compound of Formula (C-II), (C-II-A), (C-II-B), (C-II-C), (C-III-A), (C-III-C), (C-III-C), (C-IV-A), (C-IV-B), (C-IV-C), (C-V-A), (C-V-B), (C-V-C), (C-IV-A), (C-VI-B), OR (C-VI-C):
13. The conjugate of claim 1, wherein the TGFβR1 inhibitor is a compound of Formula (D-I):
(D-I) or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope, or salt thereof, wherein: QA is CRA or N; QB is CRB; Q3 is N or CR3; Q4 is N or CR4; Q5 is N or CR5;
RA is H, halo, C1-3 alkyl, or C1-3 haloalkyl and RB is A B
, or R and R , together with the atoms to which they are attached, form a heterocyclic ring; Ra and Rb are each H, or Ra and Rb, together with the atoms to which they are attached, form a heterocyclic ring; ring B is carbocycle or heterocycle; R1 is H, C1-3 alkyl, or C1-3 haloalkyl; R2 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; R3 is H, halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; R4 is H, halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; R5 is H, halo, C1-3 alkyl, C1-3 alkoxy, C1-3 haloalkyl, or C1-3 haloalkoxy; wherein R1, R2, R3, R4, and R5 are, at each occurrence, independently substituted with 0-3 R10; R7 is a reactive moiety capable of attachment to a linker or a reactive moiety capable of attachment to an antibody, an antibody construct, or a targeting moiety; R9 is, at each occurrence, independently halo, C1-3 alkyl, C1-3 haloalkyl, C1-3 alkoxy, or C1-3 haloalkoxy; R10 is, at each occurrence, independently C1-3 alkoxy or C1-3 haloalkoxy; m is 0–3; and n is 0–5.
14. The conjugate of claim 13, wherein the compound of Formula (D-I) is a compound of Formula (D-II), (D-III), (D-IV), (D-V), (D-VI), (D-IV-A), (D-VI-B), (D-VII), (D-VIII), (D-VIII-A), (D-VIII-B), (D-IX), (D-IX-A), OR (D-IX-B), or a salt thereof:
16. The conjugate of any one of claims 1 to 15, wherein Region G comprises a structure of Formula (V):
wherein n is 1, 2, or 3; SP is a spacer, wherein each SP is independently a heteroalkylene, heteroalkenylene, or heteroalkynylene comprising 5 to 30 components in the longest linear chain, wherein the components are selected from −CH2−, −CH(C1-4alkyl), −C(C1-4alkyl)2, −CH=CH−, −C≡C−, −C(O)−, −O−, −NH−, −N(C1-4alkyl), −S−, −S(O)−, −S(O)2−, and −P(O)(O−)−; and DE is a branched Display Element, wherein the asterisk (*) is the position of connection to the rest of the conjugate.
17. The conjugate of claim 16, wherein n is 2 or 3.
18. The conjugate of claim 16, wherein n is 3.
19. The conjugate of any one of claims 16 to 18, wherein one or more SP units comprise an ethylene glycol or poly(ethylene glycol) region, an amide bond, or an unsubstituted ethylene or propylene unit.
20. The conjugate of any one of claims 16 to 19, each SP is −(CH2)2−C(O)−NH−(CH2)x−CH2− (where x is 1, 2, 3, 4, 5, or 6, or x is 1, 2, or 3, or x is 1, or x is 3, or x is 4, or x is 5); −CH2−(CH2)a−CH2−NH−C(O)−(CH2)b−, where a is 0, 1, 2, or 3 (or a is 1 or 2, or a is 1); and b is 1, 2, 3, 4, 5, or 6 (or b is 3, 4, or 5, or b is 5); −C(O)−NH−(CH2)(a+2)−NH−C(O)−(CH2)b−, where a is 0, 1, 2, or 3 (or a is 1) and b is 1, 2, 3, 4,
5, or 6 (or b is 3, 4, or 5, or b is 5); or −C(O)−NH−(CH2)p− where p is 2, 3, 4, 5, or 6 (or p is 2, or p is 4, or p is 6)..
21. The conjugate of any one of claims 16 to 20, wherein the Display Element (DE) comprises a central atom unit and two or three Arm Elements (AE) attached to the central atom unit that connect DE to each SP.
23. The conjugate of claim 21 or 22, wherein each AE is independently −(CH2)1-3O−, −(CH2)1-3NR−, −NR−, −(CH2)1-3−, or a bond, wherein each CH2 is optionally substituted with one or more C1-4alkyl or fluoro substituents, and R is H or C1-4alkyl.
25. The conjugate of claim 16, wherein Region G has the structure:
27. The conjugate of claim 16, wherein Region G has the structure:
28. The conjugate of any one of claims 1 to 27, wherein Region L3 comprises a dipeptide.
29. The conjugate of claim 28, wherein the dipeptide is Val−Cit.
30. The conjugate of any one of claims 1 to 29, wherein Region L3 comprises a para-aminobenzyl moiety.
31. The conjugate of any one of claims 1 to 30, wherein Region L3 comprises −NHC(O)−(CH2)p−C(O)− where p is 2, 3, 4, 5, or 6.
32. The conjugate of claim 31, wherein Region L3 is −NHC(O)−(CH2)p−C(O)−Val−Cit−PAB−O−C(O)−.
33. A conjugate selected from:
34. A pharmaceutical composition comprising the conjugate of any one of claims 1 to 33 or a pharmaceutically acceptable salt thereof.
35. A method of treating a disease mediated by TGFβR1 activity in a subject in need thereof comprising administering to the subject an effective amount of a conjugate of any one of claims 1 to 34 or a pharmaceutically acceptable salt thereof.
36. The method of claim 35, wherein the disease is a viral infection, Hepatitis B, Hepatatis C, cancer, liver cancer, hepatocellular carcinoma, fibrosis, liver fibrosis, scleroderma, systemic fibrosis, steatohepatitis, or non-alcoholic steatohepatitis.
37. A compound of Formula (XX): [Inh]−Z (XX) wherein Region Inh comprises a TGFβR1 inhibitor; and Z is the residual portion of a released, cleavable linker or comprises a non- cleavable linker and a residual portion of a degraded Region G; or a pharmaceutically acceptable salt thereof.
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201962912783P | 2019-10-09 | 2019-10-09 | |
US62/912,783 | 2019-10-09 | ||
US202063047252P | 2020-07-02 | 2020-07-02 | |
US63/047,252 | 2020-07-02 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2021072330A1 true WO2021072330A1 (en) | 2021-04-15 |
Family
ID=73040303
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2020/055159 WO2021072330A1 (en) | 2019-10-09 | 2020-10-09 | Galnac-tgfbr1 inhibitor conjugates for the treatment of liver diseases |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2021072330A1 (en) |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2022006340A1 (en) * | 2020-07-01 | 2022-01-06 | Silverback Therapeutics, Inc. | Alk5 inhibitors, conjugates, and uses thereof |
CN114380823A (en) * | 2022-01-26 | 2022-04-22 | 中国药科大学 | Imidazole-2-methylamine derivative and medical application thereof |
WO2024152886A1 (en) * | 2023-01-17 | 2024-07-25 | 中国科学院上海药物研究所 | Conjugate of asialoglycoprotein receptor ligand and cytotoxin and use thereof |
Citations (12)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5846514A (en) | 1994-03-25 | 1998-12-08 | Isotechnika, Inc. | Enhancement of the efficacy of nifedipine by deuteration |
US6334997B1 (en) | 1994-03-25 | 2002-01-01 | Isotechnika, Inc. | Method of using deuterated calcium channel blockers |
US8450467B2 (en) | 2007-12-04 | 2013-05-28 | Alnylam Pharmaceuticals, Inc. | Carbohydrate conjugates as delivery agents for oligonucleotides |
US20130309256A1 (en) | 2012-05-15 | 2013-11-21 | Seattle Genetics, Inc. | Self-stabilizing linker conjugates |
WO2015095755A1 (en) | 2013-12-19 | 2015-06-25 | Seattle Genetics, Inc. | Methylene carbamate linkers for use with targeted-drug conjugates |
US9399775B2 (en) | 2011-11-18 | 2016-07-26 | Alnylam Pharmaceuticals, Inc. | RNAi agents, compositions and methods of use thereof for treating transthyretin (TTR) associated diseases |
US10077433B2 (en) | 2006-04-03 | 2018-09-18 | Promega Corporation | Permuted and nonpermuted luciferase biosensors |
WO2018170179A1 (en) | 2017-03-15 | 2018-09-20 | Silverback Therapeutics, Inc. | Benzazepine compounds, conjugates, and uses thereof |
US20190038765A1 (en) | 2016-02-08 | 2019-02-07 | Synaffix B.V. | Improved sulfamide linkers for use in bioconjugates |
WO2019195278A1 (en) * | 2018-04-02 | 2019-10-10 | Silverback Therapeutics, Inc. | Alk5 inhibitors, conjugates, and uses thereof |
WO2020181040A1 (en) * | 2019-03-06 | 2020-09-10 | Silverback Therapeutics, Inc. | Cyclic amino-pyrazinecarboxamide compounds and uses thereof |
WO2021011834A1 (en) * | 2019-07-16 | 2021-01-21 | Silverback Therapeutics, Inc. | Alk5 inhibitors, conjugates, and uses thereof |
-
2020
- 2020-10-09 WO PCT/US2020/055159 patent/WO2021072330A1/en active Application Filing
Patent Citations (12)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5846514A (en) | 1994-03-25 | 1998-12-08 | Isotechnika, Inc. | Enhancement of the efficacy of nifedipine by deuteration |
US6334997B1 (en) | 1994-03-25 | 2002-01-01 | Isotechnika, Inc. | Method of using deuterated calcium channel blockers |
US10077433B2 (en) | 2006-04-03 | 2018-09-18 | Promega Corporation | Permuted and nonpermuted luciferase biosensors |
US8450467B2 (en) | 2007-12-04 | 2013-05-28 | Alnylam Pharmaceuticals, Inc. | Carbohydrate conjugates as delivery agents for oligonucleotides |
US9399775B2 (en) | 2011-11-18 | 2016-07-26 | Alnylam Pharmaceuticals, Inc. | RNAi agents, compositions and methods of use thereof for treating transthyretin (TTR) associated diseases |
US20130309256A1 (en) | 2012-05-15 | 2013-11-21 | Seattle Genetics, Inc. | Self-stabilizing linker conjugates |
WO2015095755A1 (en) | 2013-12-19 | 2015-06-25 | Seattle Genetics, Inc. | Methylene carbamate linkers for use with targeted-drug conjugates |
US20190038765A1 (en) | 2016-02-08 | 2019-02-07 | Synaffix B.V. | Improved sulfamide linkers for use in bioconjugates |
WO2018170179A1 (en) | 2017-03-15 | 2018-09-20 | Silverback Therapeutics, Inc. | Benzazepine compounds, conjugates, and uses thereof |
WO2019195278A1 (en) * | 2018-04-02 | 2019-10-10 | Silverback Therapeutics, Inc. | Alk5 inhibitors, conjugates, and uses thereof |
WO2020181040A1 (en) * | 2019-03-06 | 2020-09-10 | Silverback Therapeutics, Inc. | Cyclic amino-pyrazinecarboxamide compounds and uses thereof |
WO2021011834A1 (en) * | 2019-07-16 | 2021-01-21 | Silverback Therapeutics, Inc. | Alk5 inhibitors, conjugates, and uses thereof |
Non-Patent Citations (15)
Title |
---|
"Curr., Pharm. Des.", vol. 6, 2000, article "Recent Advances in the Synthesis and Applications of Radiolabeled Compounds for Drug Discovery and Development", pages: 110 |
"Encyclopedia of Reagents for Organic Synthesis", 1995 |
CAS, no. 356560-84-4 |
CHENG HUA JIN ET AL: "4-([1,2,4]Triazolo[1,5-a]pyridin-6-yl)-5(3)-(6-methylpyridin-2-yl)imidazole and -pyrazole derivatives as potent and selective inhibitors of transforming growth factor-[beta] type I receptor kinase", BIOORGANIC & MEDICINAL CHEMISTRY, vol. 22, no. 9, 1 May 2014 (2014-05-01), NL, pages 2724 - 2732, XP055571926, ISSN: 0968-0896, DOI: 10.1016/j.bmc.2014.03.022 * |
CHENG HUA JIN ET AL: "Discovery of N -((4-([1,2,4]Triazolo[1,5- a ]pyridin-6-yl)-5-(6-methylpyridin-2-yl)-1 H -imidazol-2-yl)methyl)-2-fluoroaniline (EW-7197): A Highly Potent, Selective, and Orally Bioavailable Inhibitor of TGF-[beta] Type I Receptor Kinase as Cancer Immunotherapeutic/Antifibrotic Agent", JOURNAL OF MEDICINAL CHEMISTRY, vol. 57, no. 10, 31 May 2014 (2014-05-31), US, pages 4213 - 4238, XP055475279, ISSN: 0022-2623, DOI: 10.1021/jm500115w * |
EVANS, E. ANTHONY: "Synthesis of radiolabeled compounds", J. RADIOANAL. CHEM., vol. 64, no. 1-2, 1981, pages 9 - 32 |
GEORGE W.VARMA, RAJENDER S: "The Synthesis of Radiolabeled Compounds via Organometallic Intermediates", TETRAHEDRON, vol. 45, no. 21, 1989, pages 6601 - 21 |
HARRISONHARRISON ET AL.: "Compendium of Synthetic Organic Methods", 1971, JOHN WILEY AND SONS |
KIM MIN-JIN ET AL: "TGF-[beta] Type I Receptor Kinase Inhibitor EW-7197 Suppresses Cholestatic Liver Fibrosis by Inhibiting HIF1[alpha]-Induced Epithelial Mesenchymal Transition", CELLULAR PHYSIOLOGY AND BIOCHEMISTRY, vol. 38, no. 2, 1 January 2016 (2016-01-01), CH, pages 571 - 588, XP055776646, ISSN: 1015-8987, Retrieved from the Internet <URL:https://www.karger.com/Article/Pdf/438651> DOI: 10.1159/000438651 * |
L. FIESERM. FIESER, FIESER AND FIESER'S REAGENTS FOR ORGANIC SYNTHESIS, 1994 |
MARIKE MARJOLIJN VAN BEUGE ET AL: "Enhanced Effectivity of an ALK5-Inhibitor after Cell-Specific Delivery to Hepatic Stellate Cells in Mice with Liver Injury", PLOS ONE, vol. 8, no. 2, 18 February 2013 (2013-02-18), pages 1 - 9, XP055604603, DOI: 10.1371/journal.pone.0056442 * |
PARK SANG-A ET AL: "EW-7197 inhibits hepatic, renal, and pulmonary fibrosis by blocking TGF-[beta]/Smad and ROS signa", CMLS CELLULAR AND MOLECULAR LIFE SCIENCES, BIRKHAUSER VERLAG, HEIDELBERG, DE, vol. 72, no. 10, 9 December 2014 (2014-12-09), pages 2023 - 2039, XP035485829, ISSN: 1420-682X, [retrieved on 20141209], DOI: 10.1007/S00018-014-1798-6 * |
R. LAROCK, COMPREHENSIVE ORGANIC TRANSFORMATIONS, 1989 |
T. W. GREENEP. G. M. WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS |
T. W. GREENEP. G. M. WUTS: "Protective Groups in Organic Synthesis,", 1991 |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2022006340A1 (en) * | 2020-07-01 | 2022-01-06 | Silverback Therapeutics, Inc. | Alk5 inhibitors, conjugates, and uses thereof |
CN114380823A (en) * | 2022-01-26 | 2022-04-22 | 中国药科大学 | Imidazole-2-methylamine derivative and medical application thereof |
WO2024152886A1 (en) * | 2023-01-17 | 2024-07-25 | 中国科学院上海药物研究所 | Conjugate of asialoglycoprotein receptor ligand and cytotoxin and use thereof |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP6883518B2 (en) | Conjugated and therapeutic hemiasterin derivatives | |
RU2489423C2 (en) | Water-soluble analogues cc-1065 and their conjugates | |
AU2018288029B2 (en) | Antibody-drug conjugate having acidic self-stabilization junction | |
RU2512541C2 (en) | 8-oxy-quinoline derivatives as bradykinin b2 receptor modulators | |
TWI582093B (en) | Fgf receptor (fgfr) agonist dimeric compounds, process for the preparation thereof and therapeutic use thereof | |
WO2021072330A1 (en) | Galnac-tgfbr1 inhibitor conjugates for the treatment of liver diseases | |
JP2021532162A (en) | 1H-pyrazolo [4,3-d] pyrimidine compounds as toll-like receptor 7 (TLR7) agonists and methods and uses thereof. | |
JP2006523237A5 (en) | ||
JP2019512492A (en) | Process for preparing PEGylated drug linkers and intermediates thereof | |
KR20190074292A (en) | Targeted delivery of remedial pathway inhibitors of nicotinamide adenine dinucleotides | |
JP2021517113A (en) | Trialkin binder and how to use | |
JP7165270B2 (en) | Compounds for retinal diseases | |
TW202017570A (en) | Integrin ligands and uses thereof | |
JP2022552757A (en) | Camptothecin class drugs and their antibody conjugates | |
CN110461836B (en) | Selective kinase inhibition compound and application thereof | |
KR20230040957A (en) | Drug delivery systems for topical delivery of therapeutic agents and uses thereof | |
US9556192B2 (en) | Functionalized 9-bromo-camptothecin derivatives | |
KR20200051733A (en) | Manufacturing process of tubulinic acid and its intermediates | |
SK17112002A3 (en) | Pyridine-2-yl-aminoalkyl carbonyl glycyl-beta-alanine and derivatives thereof | |
AU2020482223A1 (en) | Trop2 targeting antibody-drug conjugate, and preparation method and use therefor | |
JP2022526450A (en) | Inhibitor of CD40-CD154 binding | |
JP7177495B2 (en) | Compositions for treating hypertension and/or fibrosis | |
US20180036312A1 (en) | Novel Scaffolds for Intracellular Compound Delivery for the Detection of Cancer Cells | |
CN112074290B (en) | Integrin targeting ligands and uses thereof | |
TW200401644A (en) | Novel azepane derivatives |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 20800451 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 20800451 Country of ref document: EP Kind code of ref document: A1 |