WO2025076284A1 - Compounds, pharmaceutical compositions thereof, and methods of using the same - Google Patents
Compounds, pharmaceutical compositions thereof, and methods of using the same Download PDFInfo
- Publication number
- WO2025076284A1 WO2025076284A1 PCT/US2024/049873 US2024049873W WO2025076284A1 WO 2025076284 A1 WO2025076284 A1 WO 2025076284A1 US 2024049873 W US2024049873 W US 2024049873W WO 2025076284 A1 WO2025076284 A1 WO 2025076284A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- ring
- iii
- cancer
- formula
- compound
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D401/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
- C07D401/14—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D239/00—Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
- C07D239/02—Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
- C07D239/24—Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
- C07D239/28—Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
- C07D239/46—Two or more oxygen, sulphur or nitrogen atoms
- C07D239/47—One nitrogen atom and one oxygen or sulfur atom, e.g. cytosine
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D401/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
- C07D401/02—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
- C07D401/04—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D405/00—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
- C07D405/14—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D417/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
- C07D417/14—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D471/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
- C07D471/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
- C07D471/04—Ortho-condensed systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D487/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
- C07D487/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
- C07D487/04—Ortho-condensed systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D513/00—Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
- C07D513/02—Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
- C07D513/04—Ortho-condensed systems
Definitions
- the cancer is a Wnt-mediated cancer. In some embodiments, the cancer is a p53-mediated cancer. In some embodiments, the cancer is an adenomatous polyposis coli (APC)-mediated cancer. In some embodiments, the cancer is selected from colorectal cancer (CRC) (e.g., APC colorectal cancer), small intestine (small bowel) cancer, thyroid cancer, brain cancer, pancreatic cancer, bile duct cancer, hepatoblastoma, primary effusion lymphoma (PEL), myelodysplastic syndrome (MDS), and acute myeloid lymphoma (AML).
- CRC colorectal cancer
- PEL primary effusion lymphoma
- MDS myelodysplastic syndrome
- AML acute myeloid lymphoma
- a substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
- Animal refers to any member of the animal kingdom. In some embodiments, “animal” refers to humans, at any stage of development. In some embodiments, “animal” refers to non-human animals, at any stage of development.
- Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl.
- Heterocycle As used herein, the terms “heterocycle”, “heterocyclyl”, “heterocyclic radical”, and “heterocyclic ring” are used interchangeably and refer to a stable 5- to 7-membered monocyclic or 7- to 10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above.
- nitrogen includes a substituted nitrogen.
- the nitrogen may be N (as in 3,4–dihydro–2H–pyrrolyl), NH (as in pyrrolidinyl), or +NR (as in N–substituted pyrrolidinyl).
- a heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted.
- materials which can serve as pharmaceutically-acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ring
- compositions A “pharmaceutically acceptable derivative” means any non-toxic salt, ester, salt of an ester or other derivative of a compound disclosed herein that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound disclosed herein or an active metabolite or residue thereof.
- Pharmaceutically acceptable salt As used herein, the term “pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M.
- Subject refers to any organism to which a compound or composition is administered in accordance with the present disclosure e.g., for experimental, diagnostic, prophylactic and/or therapeutic purposes. Typical subjects include animals (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans; insects; worms; etc.) and plants.
- animals e.g., mammals such as mice, rats, rabbits, non-human primates, and humans; insects; worms; etc.
- a subject is a mouse.
- subject is a rat.
- a subject is a non-human primate.
- a subject is a human.
- a subject is a patient.
- an individual who is susceptible to a disease, disorder, and/or condition is predisposed to have that disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition may not have been diagnosed with the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition may exhibit symptoms of the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition may not exhibit symptoms of the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition will develop the disease, disorder, and/or condition.
- Treat refers to any method used to partially or completely alleviate, ameliorate, relieve, inhibit, prevent, delay onset of, reduce severity of, and/or reduce incidence of one or more symptoms or features of a disease, disorder, and/or condition.
- Treatment may be administered to a subject who does not exhibit signs of a disease, disorder, and/or condition.
- treatment may be administered to a subject who exhibits only early signs of the disease, disorder, and/or condition, for example for the purpose of decreasing the risk of developing pathology associated with the disease, disorder, and/or condition.
- Unsaturated means that a moiety has one or more units of unsaturation.
- Wild-type As used herein, the term “wild-type” has its art-understood meaning that refers to an entity having a structure and/or activity as found in nature in a “normal” (as contrasted with mutant, diseased, altered, etc.) state or context. Those of ordinary skill in the art will appreciate that wild type genes and polypeptides often exist in multiple different forms (e.g., alleles).
- structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the disclosure. Unless otherwise stated, all tautomeric forms of the compounds of the disclosure are within the scope of the disclosure.
- the present disclosure provides a compound of formula I’: I’ or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from a 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, a 10-membered bicyclic aryl ring, a 3- to 7-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, an 8- to 10-membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 9- to 10-membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; each R x is independently selected from oxo, halogen, -NO 2 , -(CH 2
- the present disclosure provides a compound of formula I”: or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from a 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, a 10-membered bicyclic aryl ring, a 3- to 7-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, an 8- to 10-membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 9- to 10-membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; each R x is independently selected from oxo, halogen, -NO 2 , -(CH 2 )
- Ring C is a bivalent group comprising or . In some embodiments, Ring C is a bivalent group comprising . In some embodiments, Ring C is a bivalent group comprising . In some embodiments, Ring C has the structure . In some embodiments, Ring C has the structure . In some embodiments, Ring C has the structure . In some such embodiments, Ring C has the structure . In some embodiments, Ring C has the structure . In some embodiments, Ring C has the structure . In some such embodiments, Ring C has the structure . In some embodiments, Ring C has the structure . In some embodiments, Ring C has the structure . In some embodiments, Ring C has the structure . In some embodiments, Ring C has the structure . In some embodiments, Ring C has the structure . In some embodiments, Ring C has the structure . In some embodiments, Ring C has the structure . In some embodiments, Ring C has the structure . In some embodiments, Ring C has the structure . In some embodiments, Ring C has the structure . In
- Ring A is selected from a 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, a 10-membered bicyclic aryl ring, a 3- to 7-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, an 8- to 10-membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 9- to 10-membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
- Ring A is selected from cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. [00052] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is phenyl. [00053] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 10- membered bicyclic aryl ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a naphthyl ring.
- Ring A is 2-naphthyl.
- Ring A is a 3- to 7- membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
- Ring A is a 5- to 6-membered saturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
- Ring A is a piperidinyl ring.
- Ring A is a 5- to 6-membered saturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of R x .
- Ring A is a 6-membered saturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of R x .
- Ring A is a 6-membered saturated monocyclic heterocyclic ring having 1 heteroatom selected from nitrogen, oxygen, and sulfur optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of R x .
- Ring A is a piperidinyl ring optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of R x .
- Ring A is a 5- to 6-membered partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of R x .
- Ring A is a 5- to 6-membered partially unsaturated monocyclic heterocyclic ring having 1 heteroatom selected from nitrogen, oxygen, and sulfur optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of R x .
- Ring A is a 3,6-dihydro-2H-pyranyl ring optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of R x .
- Ring A is an 8- to 10- membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
- Ring A is a 10-membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
- Ring A is a 9- to 10-membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 9-membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III- a, Ring A is a 9-membered bicyclic heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
- Ring A is a benzo[d]isoxazolyl, benzo[c][1,2,5]thiadiazolyl ring, a thiazolo[5,4-b]pyridinyl ring, or a benzo[d]thiazolyl ring.
- Ring A is a 9- membered bicyclic heteroaryl ring having 1-4 nitrogen atoms.
- Ring A is a 9-membered bicyclic heteroaryl ring having 1-3 nitrogen atoms. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 9- membered bicyclic heteroaryl ring having 1-2 nitrogen atoms. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 9-membered bicyclic heteroaryl ring having 2-3 nitrogen atoms.
- Ring A is a 10-membered bicyclic heteroaryl ring having 1 nitrogen atom.
- Ring A is a quinolinyl ring or an isoquinolinyl ring.
- Ring A is a 5,6,7,8-tetrahydroquinolinyl ring.
- Ring A is selected from:
- Ring A is . In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is . In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is . In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is . In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is . In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is . In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is . In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is .
- R x is -N(R) 2 . In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R x is -(CH 2 ) y R. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R x is -(CH 2 ) z Y. It will be appreciated that a R x group on Ring A can be independent of a R x group on Ring C. In some embodiments, a R x group on Ring A is different than a R x group on Ring C.
- each Y is selected from –OR, -C(O)R, -CO 2 R, -OC(O)R, -C(O)N(R) 2 , -N(R)C(O)R, -SO 2 R, -SO 2 N(R) 2 , and -N(R)SO 2 R; or two instances of R x , together with the atoms to which they are attached, form a 5- to 6-membered saturated, partially unsaturated or aromatic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
- Y is —CO 2 R. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Y is –OR. [00072] As defined generally above for formula I”, Y is selected from -OR, -N(R) 2 , - O(CH 2 ) 2 OR, -C(O)R, -CO 2 R, -OC(O)R, -C(O)N(R) 2 , -N(R)C(O)R, -SO 2 R, -SO 2 N(R) 2 , and - N(R)SO 2 R.
- In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is –N(R y )-.
- L is –CH 2 N(R y )- or –N(R y )CH 2 -. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is –N(R y )-. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is -OCH 2 - or -CH 2 O-. [00080] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is .
- In some embodiments of Formula I, I’, I”, II, II-a, III, or III- a, L is , wherein * denotes the attachment to Ring A. [00082] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is . In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is , wherein * denotes the attachment to Ring A. In some embodiments of Formula I, I’, I”, II, II-a, III, or III- a, L is , wherein * denotes the attachment to Ring A.
- R y is lower haloalkyl. In some such embodiments, R y is -CF 3 , -CH 2 F, or -CF 2 H.
- R is –CH 3 , -CH 2 CH 3 , -CH(CH 3 ) 2 , -CD(CH 3 ) 2 , -CH(CH 3 )CH 2 CH 3 , or – CF 3 .
- R is C 1-2 aliphatic optionally substituted with –(CH 2 ) 0-4 ORo, wherein Ro is hydrogen or C 1-6 aliphatic.
- R is an optionally substituted 5- to 6-membered saturated monocyclic carbocyclic ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted cyclopropyl or cyclobutyl ring. [00096] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted phenyl.
- R is a piperidinyl ring optionally substituted with –(CH 2 ) 0-4 Ro, –(CH 2 ) 0- 4 ORo, or –(CH 2 ) 0-4 CO 2 Ro, wherein Ro is C 1-6 aliphatic.
- Ro is substituted with –(CH 2 ) 0-2 OR ⁇ or –(CH 2 ) 0-2 OH, wherein R ⁇ is C 1-4 aliphatic.
- In some embodiments of Formula I, I’, I”, II, II-a, III, or III-am is 0. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, m is 1. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, m is 2. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, m is 3. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, m is 2. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, m is 0-1.
- y is 0-1. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, y is 1-2. [000101] As defined generally above, z is 0-1. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, z is 0. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, z is 1. [000102] In some embodiments, the compound is selected from a compound in Table 1. Table 1.
- compositions comprising a compound of this disclosure, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
- a composition of this disclosure is formulated for administration to a subject in need of such composition.
- a composition of this disclosure is formulated for oral administration to a subject.
- the compounds and compositions, according to the method of the present disclosure may be administered using any amount and any route of administration effective for treating or lessening the severity of a cancer, e.g., a cancer described herein.
- Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
- These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
- Other commonly used surfactants such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
- compositions of this disclosure may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
- Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches may also be used.
- Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
- provided pharmaceutically acceptable compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride.
- the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum.
- compositions of this disclosure may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents. [000115] Most preferably, pharmaceutically acceptable compositions of this disclosure are formulated for oral administration. [000116] The amount of compounds of the present disclosure that may be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration.
- compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a subject receiving these compositions.
- a specific dosage and treatment regimen for any particular subject will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated.
- the amount of a compound of the present disclosure in the composition will also depend upon the particular compound in the composition.
- the present disclosure provides a composition (e.g., a pharmaceutical composition) comprising a compound of this disclosure, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
- a composition of this disclosure is formulated for administration to a subject in need of such composition.
- a composition of this disclosure is formulated for oral administration to a subject.
- Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
- the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar--agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite
- the dosage form may also comprise buffering agents.
- Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
- the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
- Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
- the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
- inert diluents commonly used in the art such as, for example, water or other solvents,
- the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
- adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
- injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
- the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
- Injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
- compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this disclosure with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
- suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
- Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide- polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides).
- compositions of this disclosure can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated.
- the compounds of the disclosure may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
- a dose is from about 0.01 to about 1000 mg, from about 0.5 to about 100 mg, from about 1 to about 50 mg, or from about 5 to about 100 mg. Exact doses may depend upon routes of administration, forms in which compounds are administered, subjects (e.g., body weight, age, body surface area, etc.), conditions, disorders or diseases, and/or preferences and experiences of physicians.
- a fixed dose is administered.
- two or more doses are about the same amount.
- one or more doses are independently more than one or more other doses.
- one or more loading doses each independently of a higher amount are administered before one or more maintenance doses each independently of a lower amount.
- two or more or all loading doses are about the same amount.
- a loading dose is of a higher amount than another loading dose.
- two or more or all maintenance doses are about the same amount. In some embodiments, a maintenance dose is of a higher amount than another maintenance dose.
- Compounds of the disclosure are preferably formulated in dosage unit form for ease of administration and uniformity of dosage.
- dosage unit form refers to a physically discrete unit of agent appropriate for the subject to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present disclosure will be decided by the attending physician within the scope of sound medical judgment.
- kits comprising any of the compounds and compositions described herein.
- a kit comprises a solid composition.
- a kit comprises a tablet, capsule, or pill.
- a kit comprises a liquid composition.
- a kit comprises instructions for performing any of the methods described herein. 5. Characterization and Assessment [000132] As appreciated by those skilled in the art, various technologies may be utilized to characterize and/or assess provided technologies in accordance with the present disclosure. Certain useful technologies are described in the Examples. As demonstrated, among other things, the present disclosure describes various in vitro technologies suitable for assessing and characterizing provided technologies. In some embodiments, provided technologies are characterized and/or assessed, e.g., in in vitro systems, e.g., in cells. [000133] In some embodiments, provided technologies (e.g., compounds, compositions) are characterized and/or assessed using various in vitro assays known in the art.
- compounds are assessed using a HiBiT assay.
- the HiBiT assay reportedly can be used to provide quantitative measurement of proteins in a system, e.g., an in vitro system, e.g., a cell or population of cells.
- a HiBiT assay can be conducted in a high-throughput manner.
- the protein levels can reportedly be measured through the addition of the LgBiT component to form an active nanoluciferase enzyme.
- Luminescence generated by the nanoluciferase enzyme under assay conditions can then reportedly be quantitatively measured to provide quantitation of Small BiT-tagged protein levels.
- HiBiT system is described in, e.g., Schwinn et al. ACS Chem Biol.2018 Feb 16;13(2):467- 474.
- Small BiT-tagged CK1 ⁇ is used in a HiBiT assay.
- a HiBiT assay is used to assess level of targeted protein in a system, e.g., an in vitro system.
- a HiBit assay is used to assess level of CK1 ⁇ protein in a system, e.g., an in vitro system.
- a HiBiT assay is used to assess level of targeted protein degradation in a system, e.g., an in vitro system, by a provided compound or composition.
- a HiBit assay is used to assess level of CK1 ⁇ protein degradation in a system, e.g., an in vitro system, by a provided compound or composition.
- a HiBiT assay is conducted as described herein, e.g., in an example.
- compounds are assessed using an assay to assess level of CK1 ⁇ protein or level of CK1 ⁇ activity in a system.
- level of CK1 ⁇ protein is assessed by an immunoassay.
- level of CK1 ⁇ protein is assessed by Western blot. In some embodiments, level of CK1 ⁇ protein is assessed by capillary-based immunoassay. Various additional methods for assessing levels of CK1 ⁇ protein or activity are described in the art, including those described in WO 2021/222542. [000135] In some embodiments, compounds are assessed using a TOPflash assay.
- the TOPflash assay reportedly utilizes a reporter plasmid comprising multiple (e.g., two sets of three) copies of a wild-type TCF binding site upstream of a thymidine kinase minimal promoter and luciferase open reading frame (ORF).
- the TOPflash reporter plasmid is described in, e.g., Korinek et al. Science.1997 Mar 21;275(5307):1784-7.
- an assay can reportedly be performed to provide quantitative measurement (through measurement of luciferase-driven luminescence) of ⁇ -catenin/TCF activity in a system (e.g., an in vitro system, e.g., a cell), which activates the expression from the luciferase ORF.
- This quantitative measurement may reportedly be used to assess Wnt signaling pathway activation in a system.
- a TOPflash assay is used to assess level of Wnt signaling pathway activation in a system, e.g., an in vitro system, by a provided compound or composition.
- a TOPflash assay is used to assess level of ⁇ -catenin activity in a system, e.g., an in vitro system, by a provided compound or composition.
- a TOPflash assay is conducted as described herein, e.g., in an example.
- compounds are assessed using an assay to assess level of p53 protein or level of p53 activity in a system.
- level of p53 protein is assessed by an immunoassay.
- level of p53 protein is assessed by Western blot. In some embodiments, level of p53 protein is assessed by capillary-based immunoassay. Various additional methods for assessing levels of p53 protein or activity are described in the art, including those described in WO 2021/222542. [000137]
- a provided compound or composition is characterized or assessed in an in vivo system. In some embodiments, a provided compound or composition is characterized or assessed in an animal, e.g., a mouse, rat, pig, dog, non-human primate, e.g., a monkey. In some embodiments, a provided compound or composition is characterized or assessed in a human.
- a provided compound or composition is characterized in xenograft model, e.g., a xenograft mouse model.
- xenograft model e.g., a xenograft mouse model.
- Various xenograft models for assessment of anti-cancer and/or anti-tumor properties of compounds and compositions thereof are known in the art. [000138] Those skilled in the art reading the present disclosure will readily appreciate that other technologies, e.g., in vitro models (e.g., cell lines) for various conditions, disorders, or diseases, animals models for various conditions, disorders, or diseases, clinical trials, etc. may be designed and/or utilized to assess provided technologies (e.g., compounds, compositions, methods, etc.) in accordance with the present disclosure. 6.
- provided technologies are useful for many purposes.
- provided technologies e.g., compounds, compositions, methods, etc.
- provided technologies e.g., compounds, compositions, methods, etc.
- provided technologies e.g., compounds, compositions, methods, etc.
- the Wnt signaling pathway which may also be referred to the Wnt/ ⁇ -catenin signaling pathway, reportedly involves a variety of proteins and modulates gene expression in cells ( Komiya, Y. and Habas, R. Organogenesis.2008 Apr;4(2):68-75). Wnt proteins are reportedly secreted glycoproteins which bind to receptors belonging to the Frizzled (Fz) family of receptors on the outside of cells. Further, the interaction of a Wnt protein with a co-receptor may also be necessary; reported co-receptors include lipoprotein receptor-related protein (LRP) 5/6 and various receptor tyrosine kinases (RTKs).
- LRP lipoprotein receptor-related protein
- RTKs receptor tyrosine kinases
- the present disclosure provides a method of activating an oncogenic pathway, the method comprising contacting a biological sample with, or administering to a subject in need thereof, a compound of formula I, or a pharmaceutically salt thereof.
- the oncogenic pathway is a WNT- mediated pathway.
- the tumor suppressor gene, p53 reportedly regulates transcription of a variety of genes, including those involved in, e.g., DNA damage repair, cell cycle progression, and apoptosis. Further, p53 has been reported as the most frequently mutated gene in human cancers, and even in cancers without a p53 mutation, reduced levels of p53 activity have been reported.
- CK1 ⁇ involvement in p53/MDM2/MDMX pathways is discussed in, e.g., Jiang, S. et al. Cell Commun Signal.2018 May 24;16(1):23.
- Potential downregulation of p53 through the p53/MDM2/MDMX pathway has been reported in some cancers, including, e.g., acute myeloid leukemia (AML), wherein high levels of CK1 ⁇ is associated with suppression of p53 and decreased overall survival (Xu, W. et al. Oncol Rep. 2020 Nov;44(5):1895-1904).
- AML acute myeloid leukemia
- modulation of one or more proteins in a p53/MDM2/MDMX pathway may be conducted to increase or decrease level of p53 protein and/or activity and/or downstream gene expression.
- modulation of one or more proteins may modulate level of p53 protein in a system, e.g. a cell.
- modulation of one or more proteins may modulate level of p53 activity in a system, e.g. a cell.
- modulation of one or more proteins may modulate level of p53 binding activity in a system, e.g., a cell.
- modulation of one or more proteins may involve increasing or decreasing a level of one or more proteins. In some embodiments, modulation of one or more proteins may involve increasing or decreasing a level of activity of one or more proteins. In some embodiments, modulation of one or more proteins may involve degradation of the one or more proteins.
- the present disclosure provides a method of inhibiting an oncogenic pathway, the method comprising contacting a biological sample with, or administering to a subject in need thereof, a compound of formula I, or a pharmaceutically salt thereof. In some such embodiments, the oncogenic pathway is a p53-mediated pathway.
- Casein kinase 1 ⁇ refers to a protein encoded by, in humans, the CSNK1A1 gene. CK1 ⁇ may also be known as casein kinase 1 isoform alpha, CKI-alpha, or CK1. Various CK1 ⁇ sequences are readily available to those of skill in the art, including NCBI Reference Protein Sequences Accession Nos. NP_001020276.1, NP_001883.4, NP_001258670.1, and NP_001258671.1.
- CK1 ⁇ reportedly also phosphorylates APC, which in turn assists in the association of APC with ⁇ -catenin (Ferrarese, A. et al. Biochemistry.2007 Oct 23;46(42):11902-10).
- studies have suggested potential additional roles for CK1 ⁇ in cellular functioning due to widespread localization in the cell and ubiquitous expression of the protein in different types of cells and tissues (Jiang, S. et al. Cell Commun Signal.2018 May 24;16(1):23).
- a provided compound interacts with a CK1 ⁇ polypeptide or protein. In some embodiments, a provided compound binds a CK1 ⁇ polypeptide or protein. In some embodiments, a provided compound binds a CK1 ⁇ polypeptide or protein and binds an additional polypeptide or protein. In some embodiments, a provided compound binds a CK1 ⁇ polypeptide or protein and binds a CRBN polypeptide or protein.
- Cereblon refers to a protein encoded by, in humans, the CRBN gene. Various CRBN sequences are readily available to those of skill in the art, including NCBI Reference Protein Sequences Accession Nos.
- CRBN reportedly acts a substrate receptor as part of the E3 ubiquitin ligase complex, which also includes CUL4, RBX1, and DDB1. As the substrate receptor for the complex, CRBN directly binds to proteins targeted for ubiquitin-mediated degradation.
- provided technologies modulate level of CRBN- mediated degradation of CK1 ⁇ polypeptides or proteins.
- provided technologies increase level of CRBN-mediated degradation of CK1 ⁇ polypeptides or proteins.
- the Wnt signaling pathway has been previously implicated as an oncogenic in a variety of cancers, including cancers comprising a mutation of APC, e.g., APC mutant colorectal cancer. Mutations of APC in such cancers reportedly provide oncogenic activation of Wnt signaling pathway and oncogenic activation of target genes by increased ⁇ -catenin stability and/or activity.
- Wnt signaling pathway is an oncogenic pathway.
- further perturbation of proteins in the destruction complex e.g., Axin, CK1 ⁇
- the moiety of formula I comprises one or more atoms or groups that interact with the side chains of CK1 ⁇ Lys18, CK1 ⁇ Arg21, CRBN Glu377, and/or CRBN His353.
- Ring A or a substituent on Ring A i.e., an R x group
- L comprises at least one atom that interacts with CK1 ⁇ Lys18, CRBN Glu377, and/or CRBN His353.
- a method described herein comprises administering or delivering to a subject (or contacting a biological sample with) a compound selected from those in Table 1. In some embodiments, a method described herein comprises administering or delivering to a subject (or contacting a biological sample with) a compound selected from those in Tables 1 and 2. [000158] In some embodiments, the present disclosure provides a method of modulating association of CK1 ⁇ polypeptides and CRBN polypeptides in a system, comprising administering or delivering to the system an effective amount of a provided compound or composition thereof to the system.
- a system comprises an in vivo system. In some embodiments, a system is an in vivo system. [000164] In some embodiments, a system comprises a cell. In some embodiments, a system is a cell. In some embodiments, a cell is a tumor cell. In some embodiments, a cell is a cancer cell. In some embodiments, a system comprises a tissue. In some embodiments, a system is a tissue. In some embodiments, a system comprises a tumor. In some embodiments, a system is a tumor. In some embodiments, a system comprises an organ. In some embodiments, a system is an organ. In some embodiments, a system comprises an organism. In some embodiments, a system is an organism.
- a level is reduced by at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% compared to absence of a provided compound or composition and/or presence of a reference compound or composition.
- a method of treating a cancer comprises administering or delivering a compound or composition thereof described herein. In some embodiments, a method of treating a cancer comprises administering or delivering a therapeutically effective amount of a compound or composition thereof described herein. In some embodiments, a method of treating a cancer comprises administering or delivering a compound or composition thereof described herein and one or more cancer therapeutics, e.g., one or more cancer therapeutics described herein. In some embodiments, a method of treating a cancer comprises administering or delivering a therapeutically effective amount of a compound or composition thereof described herein and a therapeutically effective amount of one or more cancer therapeutics, e.g., one or more cancer therapeutics described herein.
- an additional treatment comprises or is radiation.
- an additional treatment comprises or is one or more additional therapeutic agents.
- an additional therapeutic agent comprises or is one or more cancer therapeutics described herein.
- an additional therapeutic agent comprises or is an antibody or antigen binding fragment thereof.
- an additional therapeutic agent comprises or is a cell therapy, e.g., a genetically modified cell therapy or an engineered cell therapy.
- an additional therapeutic agent comprises or is a chemotherapeutic.
- an additional therapeutic agent comprises or is a hormone.
- an additional therapeutic agent comprises or is radiation.
- an additional therapeutic agent comprises or is a vaccine.
- an additional treatment is capable of preventing, treating, ameliorating, or slowing the progression of a condition, disorder, or disease when administered alone. In some embodiments, an additional treatment is capable of preventing, treating, ameliorating, or slowing the progression of a cancer when administered alone. In some embodiments, an additional treatment is capable of preventing, treating, ameliorating, or slowing the progression of a condition, disorder, or disease when administered in combination with a provided compound or composition. In some embodiments, an additional treatment is capable of preventing, treating, ameliorating, or slowing the progression of a cancer when administered in combination with a provided compound or composition.
- one or more cancer therapeutics are used in combination with a compound or composition described herein. In some embodiments, one or more cancer therapeutics are administered or delivered to a subject in combination with a compound or composition described herein. In some embodiments, one or more cancer therapeutics are administered or delivered to a subject prior to administering or delivering a compound or composition described herein to the subject. In some embodiments, one or more cancer therapeutics are administered or delivered to a subject at the same time as administering or delivering a compound or composition described herein to the subject. In some embodiments, one or more cancer therapeutics are administered or delivered to a subject following administering or delivering a compound or composition described herein to the subject.
- a provided compound and an additional therapeutic agent are administered or delivered in the same composition. In some embodiments, a provided compound and an additional therapeutic are administered or delivered in separate compositions.
- Example 1.3 Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-7- methoxy-1,5-naphthyridine-3-carboxamide (I-7) [000212] Synthesis of diethyl 2-(((5-methoxypyridin-3-yl)amino)methylene)malonate [000213] Into a 100 mL round-bottom flask were added 5-methoxypyridin-3-amine (2.0 g, 16.110 mmol, 1 equiv), 1,3-diethyl 2-(ethoxymethylidene) propanedioate (3.5 g, 16.110 mmol, 1.0 equiv) and EtOH (60 mL) at room temperature.
- 5-methoxypyridin-3-amine 2.0 g, 16.110 mmol, 1 equiv
- reaction was stirred at -60°C for 0.5 h. Then, dimethylformamide (2.59 g, 35.481 mmol, 3 equiv) was added dropwise to the reaction. The reaction was stirred at -60 °C for 15 min. Then, the reaction was stirred at -30 °C for 15 min. The reaction was detected by LCMS and desired product was obtained. The reaction was quenched by the addition of saturated ammonium chloride aqueous (50 mL) at -30 °C. The resulting mixture was extracted with EA (3 x 100 mL). The combined organic layers were washed with saturated NaCl aqueous (3 x 50 mL), dried over anhydrous Na 2 SO 4 .
- Example 1.13 Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- [1,2,4]triazolo[4,3-b]pyridazine-6-carboxamide (I-30) [000280] The synthesis of Compound I-30 was completed following the procedure in Example 1.1 using [1,2,4]triazolo[4,3-b]pyridazine-6-carboxylic acid.
- Example 1.14 Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1,6-naphthyridine-2-carboxamide (I-14) [000284] The synthesis of Compound I-14 was completed following the procedure in Example 1.1 using 1,6-naphthyridine-2-carboxylic acid.
- Example 1.16 Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-methyl-1H-benzo[d]imidazole-5-carboxamide (I-29) [000292] The synthesis of Compound I-29 was completed following the procedure in Example 1.1 using 1-methyl-1H-benzo[d]imidazole-5-carboxylic acid.
- Example 1.21 Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1H-pyrazolo[4,3-b]pyridine-5-carboxamide (I-6) [000312] The synthesis of Compound I-6 was completed following the procedure in Example 1.1 using 1H-pyrazolo[4,3-b]pyridine-5-carboxylic acid.
- Example 1.24 Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 8-methoxy-2H-chromene-3-carboxamide (I-16) [000324] The synthesis of Compound 16 was completed following the procedure in Example 1.1 using 8-methoxy-2H-chromene-3-carboxylic acid.
- Example 1.25 Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 6-methoxy-2-naphthamide (I-15) [000328] The synthesis of Compound I-15 was completed following the procedure in Example 1.1 using 6-methoxy-2-naphthoic acid. [000329] LC-MS: (ES, m/z): [M+H] + : 444.10.
- the resulting mixture was stirred for 16h at 60°C under nitrogen atmosphere.
- the mixture was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C18 OBD Column 30*150mm 5 ⁇ m, n; Mobile Phase A: Water(0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 9% B to 29% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 10.43) to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1- methylpyrrolo[2,3-b]pyridine-5-carboxamide (51.4 mg, 15.21%) as an off-white solid.
- Example 1.33 Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-methyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxamide (I-158) [000378] Synthesis of 6-chloro-1-methyl-1H-pyrazolo[3,4-d]pyrimidine [000379] Into a 25mL round-bottom flask were added methylhydrazine sulfate (447.97 mg, 3.108 mmol, 1.1 equiv) and Et3N (314.48 mg, 3.108 mmol, 1.1 equiv) in MeOH at 0°C.
- the resulting mixture was stirred for 16h at 60°C under nitrogen atmosphere.
- the mixture was purified by Prep-HPLC with the following conditions (Column: XBridge Prep Phenyl OBD Column 19*250 mm, 5m; Mobile Phase A: Water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 11% B to 26 % B in 10 min; Wave Length: 254nm/220nm nm; RT1 (min): 16.917) to afford N-[2-(2,6- dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1-methylpyrazolo[3,4-d]pyrimidine-6- carboxamide (26.7 mg, 17.41%) as a yellow solid.
- Example 1.34 Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-1H-pyrazolo[3,4-d]pyrimidine-6-carboxamide (I-157) [ [000395] Into a 40 mL vial were added cyclobutyl-hydrazine hydrochloride (228.60 mg, 1.865 mmol, 1.1 equiv) and MeOH (10 mL) at room temperature. To the above mixture was added Et 3 N (566.06 mg, 5.593 mmol, 3.3 equiv) dropwise at 0°C.
- Example 1.37 Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-methyl-1H-pyrazolo[3,4-b]pyridine-5-carboxamide (I-156) [000425] Into a 40 mL vial were added 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6- dione (50 mg, 0.193 mmol, 1 equiv), 1-methylpyrazolo[3,4-b]pyridine-5-carboxylic acid (34.17 mg, 0.193 mmol, 1 equiv), DMF (2 mL), DIEA (149.55 mg, 1.158 mmol, 6 equiv) and HATU (109.99 mg, 0.289 mmol, 1.5 equiv) at room temperature.
- Example 1.40 Synthesis of (R)-N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5- yl)-3-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-150) [000439] Into a 25mL round-bottom flask were added 3-(5-amino-1-oxo-3H-isoindol-2-yl) piperidine-2,6-dione (200 mg, 0.771 mmol, 1 equiv), 3-methyl-1H-pyrrolo[2,3-b] pyridine-5- carboxylic acid (135.90 mg, 0.771 mmol, 1 equiv), DIEA (299.11 mg, 2.313 mmol, 3 equiv), DMF (10 mL) and HATU (439.98 mg, 1.157 mmol, 1.5 equiv) at room temperature.
- Example 1.43 Synthesis of 1-cyclopropyl-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-146) [000458] Synthesis of methyl 1-cyclopropyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate [000459] Into a 50mL round-bottom flask were added methyl 1H-pyrrolo[2,3-b] pyridine-5- carboxylate (200 mg, 1.135 mmol, 1 equiv), cyclopropylboronic acid (195.03 mg, 2.270 mmol, 2 equiv), Na 2 CO 3 (240.64 mg, 2.270 mmol, 2 equiv), Cu(OAc) 2 (206.20 mg, 1.135 mmol, 1 equiv), 2,2’-Bipyridine (I-146) [000
- Example 1.44 Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-148) [000469] Synthesis of methyl 1-cyclobutyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate [000470] In a 100 mL round-bottom flask were added methyl 1H-pyrrolo[2,3-b] pyridine-5- carboxylate (300 mg, 1.703 mmol, 1 equiv) in DMF (25 mL) and Cs 2 CO 3 (1664.46 mg, 5.109 mmol, 3 equiv) at 0 °C.
- Example 1.58 Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 2-hydroxybenzo[d]thiazole-6-carboxamide (I-159) [000551] Synthesis of 3-(tert-butoxycarbonyl)-2-oxo-2,3-dihydrobenzo[d]thiazole-6-carboxylic acid [000552] Into a 25mL round-bottom flask were added 2-hydroxy-1,3-benzothiazole-6- carboxylic acid (100 mg, 0.512 mmol, 1 equiv) in DMF (2 mL) and sodium hydride (60% in oil, 14.75 mg, 0.614 mmol, 1.2 equiv) at 0 oC.
- Example 1.60 Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 4-fluoro-1-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-131)
- Synthesis of methyl 4-fluoro-1-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate [000569] To a stirred mixture of methyl 4-fluoro-1H-pyrrolo[2,3-b]pyridine-5-carboxylate (150 mg, 0.773 mmol, 1 equiv) and K 2 CO 3 (160.15 mg, 1.159 mmol, 1.5 equiv) in DMF (6 mL) was added MeI (109.65 mg, 0.773 mmol, 1 equiv) dropwise at room temperature under argon atmosphere.
- Example 1.62 Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-methyl-3-(trifluoromethyl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-133) [000584] Synthesis of 5-bromo-1-methyl-3-(trifluoromethyl)-1H-pyrrolo[2,3-b]pyridine [000585] Into a 100 mL 3-necked round-bottom flask were added 5-bromo-3-(trifluoromethyl)- 1H-pyrrolo[2,3-b]pyridine (900 mg, 3.396 mmol, 1 equiv) and K 2 CO 3 (938.63 mg, 6.792 mmol, 2 equiv), MeI (723.00 mg, 5.094 mmol, 1.50 equiv) in DMF(10 mL) at 0°C under nitrogen atmosphere.
- the resulting mixture was stirred for overnight at 25°C under argon atmosphere. The reaction was monitored by LCMS. The reaction was quenched by the addition of Water (50 mL) at room temperature. The mixture was extracted with EtOAc (3x50mL) and the combined organic layers were dried with Na 2 SO 4 , filtered, and concentrated under vacuum. The resulting mixture was dissolved in DMF (5 mL).
- Desired product could be detected by LCMS.
- the residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH4HCO3), 35% to 55% gradient in 20 min; detector, UV 254 nm. This resulted in methyl 1-cyclobutylpyrazolo[3,4-b]pyrazine-5- carboxylate (60 mg, 61.37%) as an off-white solid.
- the reaction was monitored by LCMS. Desired product could be detected by LCMS.
- the resulting mixture was filtered, the filter cake was washed with EtOAc (2 x 5 mL).
- the filtrate was concentrated under reduced pressure.
- the residue was purified by silica gel column chromatography, eluted with PE / EA (0:1) to afford 1-cyclobutylpyrazolo[3,4-b]pyrazine-5 - carboxylic acid (45 mg, 79.82%) as a white solid.
- Example 1.68 Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-isopropyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-127) [000668] Synthesis of methyl 1-isopropyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate [000669] Into a 40 mL vial were added methyl 1H-pyrrolo[2,3-b]pyridine-5-carboxylate (500 mg, 2.838 mmol, 1 equiv), NaH (102.16 mg, 4.257 mmol, 1.5 equiv), DMF (5 mL) and 2- iodopropane (530.70 mg, 3.122 mmol, 1.1 equiv) at room temperature.
- the mixture was purged with nitrogen for 3 mins and then was pressurized to 40 atm. with carbon monoxide at 120°C for overnight.
- the reaction mixture was cooled to room temperature and filtered to remove insoluble solids.
- the residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water , 50% to 70% gradient in 10 min; detector, UV 254 nm.to afford as a methyl 3,4-dimethyl-1H-pyrrolo[2,3-b]pyridine-5- carboxylate (171 mg, 47.12%) as a white solid.
- Example 1.74 Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 4-hydroxy-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxamide (I-111) [000762] Synthesis of 4-fluoro-6-iodo-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidine [000763] Into a 50mL 3-necked round-bottom flask were added 4-fluoro-1- isopropylpyrazolo[3,4-d]pyrimidin-6-amine (50 mg, 0.256 mmol, 1 equiv) ,I 2 (65.01 mg, 0.256 mmol, 1 equiv) ,CH 2 I 2 (686.05 mg, 2.560 mmol, 10 equiv) ,THF (2 mL) ,(3-methylbutyl)
- Example 1.77 Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-(1-methylpiperidin-4-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-116) [000796] Synthesis of methyl 1-(1-methylpiperidin-4-yl)-1H-pyrrolo[2,3-b]pyridine-5- carboxylate [000797] Into a 40 mL vial were added methyl 1H-pyrrolo[2,3-b]pyridine-5-carboxylate (300 mg, 1.703 mmol, 1 equiv), 4-bromo-1-methylpiperidine (454.85 mg, 2.554 mmol, 1.5 equiv) and DMF (1 mL) at room temperature.
- the crude product was purified by Prep-HPLC with the following conditions (column, XBridge Prep; mobile phase, MeCN in Water (0.1% TFA), 10% to 50% gradient in 10 min; detector, UV 254 nm) to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1-(1-methylpiperidin-4- yl)pyrrolo[2,3-b]pyridine-5-carboxamide (2.8 mg, 4.77%) as a white solid.
- the crude product was purified by Prep-HPLC with the following conditions (Xselect CSH F-Phenyl OBD column; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm) to afford tert-butyl 4-(5- ⁇ [2- (2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]carbamoyl ⁇ pyrrolo[2,3-b]pyridin-1- yl)piperidine-1-carboxylate (9.5 mg, 54.42%) as a white solid.
- the crude product was purified by Prep-HPLC with the following conditions (Xselect CSH F-Phenyl OBD column; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm) to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1- (piperidin-4-yl)pyrrolo[2,3- b]pyridine-5-carboxamide (25.0 mg, 6.37%) as a white solid.
- Example 1.82 Synthesis of 1-(sec-butyl)-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-123) [000857] Synthesis of 1-(sec-butyl)-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid [000858] Into a 50 mL round-bottom flask were added methyl 1H-pyrrolo[2,3-b]pyridine-5- carboxylate (500 mg, 2.838 mmol, 1 equiv) and DMF (5 mL) at room temperature.
- the crude product (116 mg) was purified by Prep-HPLC with the following conditions (column, C18 silica gel; mobile phase, MeOH in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm) to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1-(sec-butyl)pyrrolo[2,3- b]pyridine-5-carboxamide (44.0 mg, 20.52%) as an off-white solid.
- Example 1.84 Synthesis of N-(1-cyclobutyl-1H-pyrrolo[2,3-b]pyridin-5-yl)-2- (2,6-dioxopiperidin-3-yl)-6-fluoro-1-oxoisoindoline-5-carboxamide (I-93) [000887] Synthesis of 2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1-oxoisoindoline-5-carboxylic acid [000888] A mixture of 3-(5-bromo-6-fluoro-1-oxo-3H-isoindol-2-yl)piperidine-2,6-dione (400 mg, 1.173 mmol, 1 equiv), Pd(OAc) 2 (88 mg, 0.392 mmol, 0.33 equiv), H2O (124 mg, 6.883 mmol, 5.87 equiv), DIEA (133 mg, 1.029
- the resulting mixture was stirred at 60°C for 2 h under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The reaction was quenched with Water/Ice at room temperature. The resulting mixture was extracted with CH 2 Cl 2 (3 x 70 mL). The combined organic layers were washed with brine (3x30 mL), dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure.
- the residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 0% to 40% gradient in 30 min; detector, UV 254 nm.
- the product was purified by Prep-HPLC to afford N-(4-cyclobutoxypyrimidin-2-yl)-2-(2,6- dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5-carboxamide (7.7 mg, 3.62%) as a white solid.
- the resulting mixture was stirred for 16 h at 60 °C under nitrogen atmosphere.
- the reaction was quenched with water (3 mL).
- the reaction mixture was extracted with EA (30 mL).
- the organic layer was washed with water (3 x 10 mL), dried over anhydrous Na 2 SO 4 . After filtrated, the filtrate was concentrated under reduced pressure.
- Example 1.94 Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(1-isopropyl-3-methyl- 1H-pyrazolo[3,4-d]pyrimidin-6-yl)-1-oxoisoindoline-5-carboxamide (I-102) [000986] Synthesis of 1-isopropyl-3-methyl-1H-pyrazolo[3,4-d]pyrimidin-6-amine [000987] Into a 8mL vial were added 6-chloro-1-isopropyl-3-methylpyrazolo[3,4-d]pyrimidine (100 mg, 0.475 mmol, 1 equiv) in NH3(g) in MeOH (2 mL).
- Example 1.96 Synthesis of N-(1-cyclobutyl-3-methyl-1H-pyrrolo[2,3-b]pyridin- 5-yl)-2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxamide (I-101) [0001007] Synthesis of 1-cyclobutyl-3-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine [0001008] Into a 40 mL vial were added 3-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine (300 mg, 1.693 mmol, 1 equiv), Cs 2 CO 3 (827.59 mg, 2.540 mmol, 1.5 equiv), DMF (5 mL) and iodocyclobutane (369.84 mg, 2.032 mmol, 1.2 equiv) at room temperature.
- Desired product could be detected by LCMS.
- the resulting mixture was filtered and the filter cake was washed with MeOH (2 x 10 mL). The filtrate was concentrated under reduced pressure.
- the residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH 4 HCO 3 ), 30% to 40% gradient in 10 min; detector, UV 254 nm. This resulted in 1-cyclobutyl-3-methylpyrrolo[2,3-b]pyridin-5-amine (240 mg, 91.92%) as a brown solid.
- the final reaction mixture was irradiated with microwave radiation at 0 °C for 1 h.
- the reaction was monitored by LCMS. About 70% product.
- the resulting mixture was concentrated under vacuum.
- the residue was purified by reversed-phase flash chromatography with the following conditions: column, C18; mobile phase, MeCN in Water (0.1% FA), 5% to 95% gradient in 25 min; detector, UV 220 nm.
- Step 1 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxylic acid
- Pd(OAc) 2 418 mg, 1.86 mmol
- DCC 2.6 g, 12.6 mmol
- triethylamine 2.6 g, 125 mmol
- Xantphos (1.08 g, 1.86 mmol) and formic acid (10 g, 217 mmol) and the reaction heated at 100 °C for 3 h.
- Step 3 N-(5-cyclobutoxypyrazin-2-yl)-2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline- 5-carboxamide
- TEA 73.5 mg, 0.73 mmol
- 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5- carbonyl chloride 74 mg, 0.24 mmol.
- Example 1.103 Synthesis of 5-(benzofuran-2-yl)-N-((4'-fluoro-[1,1'-biphenyl]-4- yl)methyl)pyrimidin-4-amine (I-165) [0001116] Step 1: 6-cyclobutoxypyridazin-3-amine [0001117] To a solution of Na (19.5 mg, 0.84 mmol) in cyclobutanol (2 mL) was added 6- chloropyridazin-3-amine (100 mg, 0.77 mmol) and TBAI (10 mg) and the reaction heated at 110 °C for 3 h.
- Step 2 5-(benzofuran-2-yl)-N-((4'-fluoro-[1,1'-biphenyl]-4-yl)methyl)pyrimidin-4- amine
- HATU 118.7 mg, 0.31 mmol
- DIEA 121.0 mg, 0.94 mmol
- Example 1.105 Synthesis of N-(4-cyclobutoxypyrimidin-2-yl)-3-(2,4- dioxotetrahydropyrimidin-1(2H)-yl)benzamide (I-231) [0001124] Step 1: 3-((2-carboxyethyl)amino)benzoic acid [0001125] To a solution of 3-aminobenzoic acid (5.0 g, 36.46 mmol) in toluene (100 mL) was added acrylic acid (3.42 g, 47.4 mmol) and the reaction heated at reflux for 48 h.
- Step 4 N-(4-cyclobutoxypyrimidin-2-yl)-3-(2,4-dioxotetrahydropyrimidin-1(2H)- yl)benzamide
- TEA 612 mg, 6.05 mmol
- DMAP 7.4 mg, 0.06 mmol
- 3-(2,4- dioxotetrahydropyrimidin-1(2H)-yl)benzoyl chloride 305 mg, 1.21 mmol.
- the reaction was stirred at room temperature for 2 h then the solvent was removed under vaccuum.
- Step 3 N-(4-cyclobutoxypyrimidin-2-yl)-2-(2,6-dioxopiperidin-3-yl)-1,3- dioxoisoindoline-5-carboxamide
- Step 3 4-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzoyl chloride
- a solution of 4-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzoic acid (150 mg, 0.64 mmol) in SOCl2 (3 mL) was heated at reflux for 4 h.
- the reaction was concentrated to afford 4- (2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzoyl chloride (160 mg, quant.) as a yellow oil which was used in next step without further purification.
- Step 1 4-chloro-6-methoxypyridin-2-amine [0001209] To a solution of 4,6-dichloropyridin-2-amine (3.0 g, 18.52 mmol) in NMP (30 mL) under a N2 atmosphere was added sodium methoxide (5.0 g, 92.6 mmol) and the reaction heated at 120 °C overnight. The mixture was diluted with water (50 mL) and extracted with EtOAc (50 mL ⁇ 2). The combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered and concentrated.
- Step 3 1-cyclobutyl-4-cyclopropyl-1H-pyrrolo[2,3-b]pyridin-5-amine
- 1-cyclobutyl-4-cyclopropyl-5-nitro-1H-pyrrolo[2,3-b]pyridine 100 mg, 0.39 mmol
- Fe power 163 mg, 2.92 mmol
- NH4Cl 31 mg, 0.58 mmol
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- General Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present disclosure provides compounds, pharmaceutically acceptable compositions thereof, and methods of using the same.
Description
COMPOUNDS, PHARMACEUTICAL COMPOSITIONS THEREOF, AND METHODS OF USING THE SAME BACKGROUND [0001] Treatment of cancer remains a considerable challenge despite significant research into the underpinnings of various cancers and a variety of existing cancer therapeutics. Currently available cancer therapeutics and treatment methods using said therapeutics largely focus on the inhibition of key cellular pathways which support, e.g., growth, proliferation, and metastasis of neoplastic cells. However, many cancers may not comprise, e.g., targeted mutations or may be resistant to currently available inhibition-based strategies. Moreover, such resistance may be developed during the course of treatment with available inhibitory cancer therapeutics. Accordingly, there is an ongoing and unmet need for novel cancer therapeutics. SUMMARY [0002] In some embodiments, the present disclosure provides a compound of formula I:
or a pharmaceutically acceptable salt thereof, wherein each of Ring A, Ring C, L, Rx, and m is as defined and described herein. [0003] In some embodiments, the present disclosure also provides a pharmaceutical composition comprising a compound of formula I or I’, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. [0004] In some embodiments, the present disclosure also provides a method of treating a cancer, the method comprising administering a compound of formula I or I’, or a pharmaceutically acceptable salt thereof, to a subject in need thereof. In some embodiments, the cancer is a CK1α-mediated cancer. In some embodiments, the cancer is a Wnt-mediated cancer. In some embodiments, the cancer is a p53-mediated cancer. In some embodiments, the cancer is an adenomatous polyposis coli (APC)-mediated cancer. In some embodiments, the cancer is selected from colorectal cancer (CRC) (e.g., APC colorectal cancer), small intestine (small bowel) cancer, thyroid cancer, brain cancer, pancreatic cancer, bile duct cancer, hepatoblastoma,
primary effusion lymphoma (PEL), myelodysplastic syndrome (MDS), and acute myeloid lymphoma (AML). [0005] In some embodiments, the present disclosure provides a method of modulating an oncogenic pathway, the method comprising contacting a biological sample with, or administering to a subject in need thereof, a compound of formula I or I’, or a pharmaceutically salt thereof. In some embodiments, the oncogenic pathway is a WNT-mediated pathway or a p53-mediated pathway. DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS 1. General Description of Provided Compounds [0006] In some embodiments, the present disclosure provides a compound of formula I”:
or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from a 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, a 10-membered bicyclic aryl ring, a 3- to 7-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, an 8- to 10-membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 9- to 10-membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; each Rx is independently selected from oxo, halogen, -NO2, -(CH2)yR, –CN, –OR, -N(R)2, -SR, and -(CH2)zY; or: two instances of Rx, together with the atoms to which they are attached, form a 5- to 6- membered saturated, partially unsaturated or aromatic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
Y is selected from -OR, -N(R)2, -O(CH2)2OR, -C(O)R, -CO2R, -OC(O)R, -C(O)N(R)2, - N(R)C(O)R, -SO2R, -SO2N(R)2, and -N(R)SO2R; L is a C1-4 aliphatic chain wherein one or more methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, -C(=O)-, -O-, - S(O)2-, and -CF2-; each Ry is selected from hydrogen, C1-3 alkyl and lower haloalkyl; Ring C is a bivalent group comprising
wherein Ring C is substituted with 0-3 instances of Rx; each R is independently hydrogen or an optionally substituted group selected from C1-6 aliphatic, a 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, a 3- to 7-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5- to 6- membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; m is 0-3; y is 0-2; and z is 0-1. 2. Compounds and Definitions [0007] Compounds of this disclosure include those described generally above, and are further illustrated by the classes, subclasses, and species disclosed herein. As used herein, the following definitions shall apply unless otherwise indicated. For purposes of this disclosure, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75th Ed. Additionally, general principles of organic chemistry are described in “Organic Chemistry”, Thomas Sorrell, University Science Books, Sausalito: 1999, and “March’s Advanced Organic Chemistry”, 5th Ed., Ed.: Smith, M.B. and March, J., John Wiley & Sons, New York: 2001, the entire contents of which are hereby incorporated by reference.
[0008] Aliphatic: The term “aliphatic” or “aliphatic group”, as used herein, means a straight- chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as “carbocycle” “cycloaliphatic” or “cycloalkyl”), that has a single point of attachment to the rest of the molecule. Unless otherwise specified, aliphatic groups contain 1-6 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-5 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms. In some embodiments, “cycloaliphatic” (or “carbocycle” or “cycloalkyl”) refers to a monocyclic C3-C6 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule. Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl. [0009] Alkylene: The term “alkylene” refers to a bivalent alkyl group. An “alkylene chain” is a polymethylene group, i.e., –(CH2)n–, wherein n is a positive integer, preferably from 1 to 6, from 1 to 4, from 1 to 3, from 1 to 2, or from 2 to 3. A substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group. [00010] Animal: As used herein, the term “animal” refers to any member of the animal kingdom. In some embodiments, “animal” refers to humans, at any stage of development. In some embodiments, “animal” refers to non-human animals, at any stage of development. In certain embodiments, the non-human animal is a mammal (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate and/or a pig). In some embodiments, animals include, but are not limited to, mammals, birds, reptiles, amphibians, fish and/or worms. In some embodiments, an animal may be a transgenic animal, a genetically-engineered animal and/or a clone. [00011] Aryl: The term “aryl” used alone or as part of a larger moiety as in “aralkyl”, “aralkoxy”, or “aryloxyalkyl”, refers to monocyclic and bicyclic ring systems having a total of
five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains three to seven ring members. The term “aryl” may be used interchangeably with the term “aryl ring”. In certain embodiments of the present disclosure, “aryl” refers to an aromatic ring system which includes, but not limited to, phenyl, biphenyl, naphthyl, anthracyl and the like, which may bear one or more substituents. Also included within the scope of the term “aryl”, as it is used herein, is a group in which an aromatic ring is fused to one or more non–aromatic rings, such as indanyl, phthalimidyl, naphthimidyl, phenanthridinyl, or tetrahydronaphthyl, and the like, wherein the point of attachment is on the aryl ring. [00012] Associated: Two events or entities are “associated” with one another, as that term is used herein, if the presence, level and/or form of one is correlated with that of the other. For example, a particular biological phenomenon (e.g., expression of a specific biomarker) is considered to be associated with a disease, disorder, and/or condition (e.g., a cancer, a specific type of cancer), if its presence correlates with incidence of and/or susceptibility of the disease, disorder, and/or condition (e.g., across a relevant population). [00013] Biological sample: The term “biological sample”, as used herein, includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from an animal, e.g., a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof. [00014] Cancer: The term “cancer” is used herein to generally refer to a disease or condition in which cells of a tissue of interest exhibit relatively abnormal, uncontrolled, and/or autonomous growth, so that they exhibit an aberrant growth phenotype characterized by a significant loss of control of cell proliferation. In some embodiments, cancer may comprise cells that are precancerous (e.g., benign), malignant, pre-metastatic, metastatic, and/or non-metastatic. [00015] Combination: As used herein, the term “combination,” “combined,” and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this disclosure. For example, a compound of the present disclosure may be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form. Accordingly, the present disclosure provides a single unit dosage form comprising a provided compound, an additional therapeutic agent, and a pharmaceutically acceptable carrier, adjuvant, or vehicle. [00016] Halogen: The term “halogen” means F, Cl, Br, or I.
[00017] Heteroaryl: The terms “heteroaryl” and “heteroar–”, used alone or as part of a larger moiety, e.g., “heteroaralkyl”, or “heteroaralkoxy”, refer to groups having 5 to 10 ring atoms, preferably 5, 6, or 9 ring atoms; having 6, 10, or 14 π electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms. The term “heteroatom” refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen. Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl. The terms “heteroaryl” and “heteroar–”, as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring. Nonlimiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, 5,6,7,8-tetrahydroquinolinyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H–quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, and phenoxazinyl. A heteroaryl group may be mono– or bicyclic. The term “heteroaryl” may be used interchangeably with the terms “heteroaryl ring”, “heteroaryl group”, or “heteroaromatic”, any of which terms include rings that are optionally substituted. The term “heteroaralkyl” refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted. [00018] Heteroatom: The term “heteroatom” means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR+ (as in N-substituted pyrrolidinyl)). [00019] Heterocycle: As used herein, the terms “heterocycle”, “heterocyclyl”, “heterocyclic radical”, and “heterocyclic ring” are used interchangeably and refer to a stable 5- to 7-membered monocyclic or 7- to 10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above. When used in reference to a ring atom of a heterocycle, the term “nitrogen” includes a substituted nitrogen. As an example, in a saturated or partially unsaturated
ring having 0–3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N (as in 3,4–dihydro–2H–pyrrolyl), NH (as in pyrrolidinyl), or +NR (as in N–substituted pyrrolidinyl). [00020] A heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted. Examples of such saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenyl pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and quinuclidinyl. The terms “heterocycle”, “heterocyclyl”, “heterocyclyl ring”, “heterocyclic group”, “heterocyclic moiety”, and “heterocyclic radical”, are used interchangeably herein, and also include groups in which a heterocyclyl ring is fused to one or more aryl, heteroaryl, or cycloaliphatic rings, such as indolinyl, 3H-indolyl, chromanyl, phenanthridinyl, 1,2,3,4-tetrahydroisoquinolinyl, or 1,2,3,4- tetrahydroquinolinyl, where the radical or point of attachment is on the heterocyclyl ring. A heterocyclyl group may be mono- or bicyclic. The term “heterocyclylalkyl” refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted. [00021] In vitro: As used herein, the term “in vitro” refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel, in cell culture, etc., rather than within an organism (e.g., animal, plant and/or microbe). [00022] In vivo: As used herein, the term “in vivo” refers to events that occur within an organism (e.g., animal, plant and/or microbe). [00023] Lower haloalkyl: The term “lower haloalkyl” refers to a C1-3 straight or branched alkyl group that is substituted with one or more halogen atoms. [00024] Optionally substituted: As described herein, compounds of the disclosure may contain “optionally substituted” moieties. In general, the term “substituted”, whether preceded by the term “optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an “optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. Combinations of substituents envisioned by this disclosure are preferably those that
result in the formation of stable or chemically feasible compounds. The term “stable”, as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein. [00025] Suitable monovalent substituents on a substitutable carbon atom of an “optionally substituted” group are independently halogen; –(CH2)0–4R°; –(CH2)0–4OR°; -O(CH2)0–4R°; –O– (CH2)0–4C(O)OR°; –(CH2)0–4CH(OR°)2; –(CH2)0–4SR°; –(CH2)0–4Ph, which may be substituted with R°; –(CH2)0–4O(CH2)0–1Ph which may be substituted with R°; –CH=CHPh, which may be substituted with R°; –(CH2)0–4O(CH2)0–1-pyridyl which may be substituted with R°; –NO2; –CN; –N3; -(CH2)0–4N(R°)2; –(CH2)0–4N(R°)C(O)R°; –N(R°)C(S)R°; –(CH2)0– 4N(R°)C(O)NR°2; -N(R°)C(S)NR°2; –(CH2)0–4N(R°)C(O)OR°; – N(R°)N(R°)C(O)R°; -N(R°)N(R°)C(O)NR°2; -N(R°)N(R°)C(O)OR°; –(CH2)0–4C(O)R°; – C(S)R°; –(CH2)0–4C(O)OR°; –(CH2)0–4C(O)SR°; -(CH2)0–4C(O)OSiR°3; –(CH2)0–4OC(O)R°; – OC(O)(CH2)0–4SR°, SC(S)SR°; –(CH2)0–4SC(O)R°; –(CH2)0–4C(O)NR°2; –C(S)NR°2; –C(S)SR°; –SC(S)SR°, -(CH2)0–4OC(O)NR°2; -C(O)N(OR°)R°; –C(O)C(O)R°; –C(O)CH2C(O)R°; – C(NOR°)R°; -(CH2)0–4SSR°; –(CH2)0–4S(O)2R°; –(CH2)0–4S(O)2OR°; –(CH2)0–4OS(O)2R°; – S(O)2NR°2; -(CH2)0–4S(O)R°; -N(R°)S(O)2NR°2; –N(R°)S(O)2R°; –N(OR°)R°; –C(NH)NR°2; – P(O)2R°; -P(O)R°2; -OP(O)R°2; –OP(O)(OR°)2; SiR°3; –(C1–4 straight or branched alkylene)O– N(R°)2; or –(C1–4 straight or branched alkylene)C(O)O–N(R°)2, wherein each R° may be substituted as defined below and is independently hydrogen, C1–6 aliphatic, –CH2Ph, –O(CH2)0– 1Ph, -CH2-(5-6 membered heteroaryl ring), or a 5–6–membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R°, taken together with their intervening atom(s), form a 3–12–membered saturated, partially unsaturated, or aryl mono– or bicyclic ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which may be substituted as defined below. [00026] Suitable monovalent substituents on R° (or the ring formed by taking two independent occurrences of R° together with their intervening atoms), are independently halogen, –(CH2)0–2R●, –(haloR●), –(CH2)0–2OH, –(CH2)0–2OR●, –(CH2)0– 2CH(OR●)2; -O(haloR●), –CN, –N3, –(CH2)0–2C(O)R●, –(CH2)0–2C(O)OH, –(CH2)0–2C(O)OR●, –
(CH2)0–2SR●, –(CH2)0–2SH, –(CH2)0–2NH2, –(CH2)0–2NHR●, –(CH2)0–2NRl2, –NO2, –SiRl3, – OSiR●3, -C(O)SR●, –( C1–4 straight or branched alkylene)C(O)OR●, or –SSR● wherein each R● is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently selected from C1–4 aliphatic, –CH2Ph, –O(CH2)0–1Ph, or a 5–6–membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of R° include =O and =S. [00027] Suitable divalent substituents on a saturated carbon atom of an “optionally substituted” group include the following: =O, =S, =NNR*2, =NNHC(O)R*, =NNHC(O)OR*, =NNHS(O)2R*, =NR*, =NOR*, –O(C(R*2))2–3O–, or –S(C(R*2))2–3S–, wherein each independent occurrence of R* is selected from hydrogen, C1–6 aliphatic which may be substituted as defined below, or an unsubstituted 5–6–membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an “optionally substituted” group include: –O(CR*2)2–3O–, wherein each independent occurrence of R* is selected from hydrogen, C1–6 aliphatic which may be substituted as defined below, or an unsubstituted 5–6– membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. [00028] Suitable substituents on the aliphatic group of R* include halogen, – R●, -(haloR●), -OH, –OR●, –O(haloR●), –CN, –C(O)OH, –C(O)OR●, –NH2, –NHR●, –NR●2, or –NO2, wherein each R● is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C1–4 aliphatic, –CH2Ph, –O(CH2)0–1Ph, or a 5–6– membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. [00029] Suitable substituents on a substitutable nitrogen of an “optionally substituted” group include –R†, –NR† 2, –C(O)R†, –C(O)OR†, –C(O)C(O)R†, –C(O)CH2C(O)R†, – S(O)2R†, -S(O)2NR† 2, –C(S)NR† 2, –C(NH)NR† 2, or –N(R†)S(O)2R†; wherein each R† is independently hydrogen, C1–6 aliphatic which may be substituted as defined below, unsubstituted –OPh, or an unsubstituted 5–6–membered saturated, partially unsaturated, or aryl ring having 0– 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R†, taken together with their intervening
atom(s) form an unsubstituted 3–12–membered saturated, partially unsaturated, or aryl mono– or bicyclic ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. [00030] Suitable substituents on the aliphatic group of R† are independently halogen, – R●, -(haloR●), –OH, –OR●, –O(haloR●), –CN, –C(O)OH, –C(O)OR●, –NH2, –NHR●, –NR● 2, or -NO2, wherein each R● is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C1–4 aliphatic, –CH2Ph, –O(CH2)0–1Ph, or a 5–6– membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. [00031] Partially unsaturated: As used herein, the term “partially unsaturated” refers to a ring moiety that includes at least one double or triple bond. The term “partially unsaturated” is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined. [00032] Pharmaceutically acceptable: As used herein, the phrase “pharmaceutically acceptable” refers 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. [00033] Pharmaceutically acceptable carrier: As used herein, the term “pharmaceutically acceptable carrier” means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the subject. Some examples of materials which can serve as pharmaceutically-acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic
saline; Ringer’s solution; ethyl alcohol; pH buffered solutions; polyesters, polycarbonates and/or polyanhydrides; and other non-toxic compatible substances employed in pharmaceutical formulations. [00034] Pharmaceutically acceptable derivative: A “pharmaceutically acceptable derivative” means any non-toxic salt, ester, salt of an ester or other derivative of a compound disclosed herein that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound disclosed herein or an active metabolite or residue thereof. [00035] Pharmaceutically acceptable salt: As used herein, the term “pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1–19, incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of this disclosure include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2–hydroxy–ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2–naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3–phenylpropionate, phosphate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p–toluenesulfonate, undecanoate, valerate salts, and the like. [00036] Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(C1–4alkyl)4 salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically
acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate. [00037] Subject: As used herein, the term “subject” or “test subject” refers to any organism to which a compound or composition is administered in accordance with the present disclosure e.g., for experimental, diagnostic, prophylactic and/or therapeutic purposes. Typical subjects include animals (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans; insects; worms; etc.) and plants. In some embodiments, a subject is a mouse. In some embodiments, subject is a rat. In some embodiments, a subject is a non-human primate. In some embodiments, a subject is a human. In some embodiments, a subject is a patient. In some embodiments, a subject may be suffering from and/or susceptible to a disease, disorder, and/or condition. In some embodiments, a subject may be suffering from and/or susceptible to a cancer. [00038] Suffering from: An individual who is “suffering from” a disease, disorder, and/or condition has been diagnosed with and/or displays one or more symptoms of a disease, disorder, and/or condition. [00039] Susceptible to: An individual who is “susceptible to” a condition, disorder, and/or disease is one who has a higher risk of developing the disease, disorder, and/or condition than does a member of the general public. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition is predisposed to have that disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition may not have been diagnosed with the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition may exhibit symptoms of the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition may not exhibit symptoms of the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition will develop the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition will not develop the disease, disorder, and/or condition. [00040] Therapeutically effective amount: As used herein, the term “therapeutically effective amount” means an amount of a substance (e.g., a therapeutic agent, composition, and/or formulation) that elicits a desired biological response when administered as part of a therapeutic
regimen. In some embodiments, a therapeutically effective amount of a substance is an amount that is sufficient, when administered to a subject suffering from or susceptible to a disease, disorder, and/or condition, to treat, diagnose, prevent, and/or delay the onset of the disease, disorder, and/or condition. As will be appreciated by those of ordinary skill in this art, the effective amount of a substance may vary depending on such factors as the desired biological endpoint, the substance to be delivered, the target cell or tissue, etc. For example, the effective amount of compound in a formulation to treat a disease, disorder, and/or condition is the amount that alleviates, ameliorates, relieves, inhibits, prevents, delays onset of, reduces severity of and/or reduces incidence of one or more symptoms or features of the disease, disorder, and/or condition. In some embodiments, a therapeutically effective amount is administered in a single dose; in some embodiments, multiple unit doses are required to deliver a therapeutically effective amount. [00041] Treat: As used herein, the term “treat,” “treatment,” or “treating” refers to any method used to partially or completely alleviate, ameliorate, relieve, inhibit, prevent, delay onset of, reduce severity of, and/or reduce incidence of one or more symptoms or features of a disease, disorder, and/or condition. Treatment may be administered to a subject who does not exhibit signs of a disease, disorder, and/or condition. In some embodiments, treatment may be administered to a subject who exhibits only early signs of the disease, disorder, and/or condition, for example for the purpose of decreasing the risk of developing pathology associated with the disease, disorder, and/or condition. [00042] Unsaturated: The term “unsaturated”, as used herein, means that a moiety has one or more units of unsaturation. [00043] Wild-type: As used herein, the term “wild-type” has its art-understood meaning that refers to an entity having a structure and/or activity as found in nature in a “normal” (as contrasted with mutant, diseased, altered, etc.) state or context. Those of ordinary skill in the art will appreciate that wild type genes and polypeptides often exist in multiple different forms (e.g., alleles). [00044] Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as
well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the disclosure. Unless otherwise stated, all tautomeric forms of the compounds of the disclosure are within the scope of the disclosure. Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this disclosure. Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present disclosure. 3. Description of Exemplary Compounds [00045] In some embodiments, the present disclosure provides a compound of formula I:
I or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from a 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, a 10-membered bicyclic aryl ring, a 3- to 7-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, an 8- to 10-membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 9- to 10-membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; each Rx is independently selected from oxo, halogen, -NO2, -(CH2)yR, –CN, –OR, -N(R)2, -SR, and -(CH2)zY; or: two instances of Rx, together with the atoms to which they are attached, form a 5- to 6- membered saturated, partially unsaturated or aromatic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
Y is selected from -C(O)R, -CO2R, -OC(O)R, -C(O)N(R)2, -N(R)C(O)R, -SO2R, -SO2N(R)2, and -N(R)SO2R; L is a C1-4 aliphatic chain wherein one or more methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, -C(=O)-, -O-, - S(O)2-, and -CF2-; each Ry is selected from hydrogen, C1-3 alkyl or lower haloalkyl; Ring C is a bivalent group comprising
each R is independently hydrogen or an optionally substituted group selected from C1-6 aliphatic, a 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, a 3- to 7-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5- to 6- membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; m is 0-3; y is 0-2; and z is 0-1. [00046] In some embodiments, the present disclosure provides a compound of formula I’:
I’ or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from a 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, a 10-membered bicyclic aryl ring, a 3- to 7-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, an 8- to 10-membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 9- to 10-membered
bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; each Rx is independently selected from oxo, halogen, -NO2, -(CH2)yR, –CN, –OR, -N(R)2, -SR, and -(CH2)zY; or: two instances of Rx, together with the atoms to which they are attached, form a 5- to 6- membered saturated, partially unsaturated or aromatic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; Y is selected from -C(O)R, -CO2R, -OC(O)R, -C(O)N(R)2, -N(R)C(O)R, -SO2R, -SO2N(R)2, and -N(R)SO2R; L is a C1-4 aliphatic chain wherein one or more methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, -C(=O)-, -O-, - S(O)2-, and -CF2-; each Ry is selected from hydrogen, C1-3 alkyl or lower haloalkyl; Ring C is a bivalent group comprising
, wherein Ring C is substituted with 0-3 instances of Rx; each R is independently hydrogen or an optionally substituted group selected from C1-6 aliphatic, a 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, a 3- to 7-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5- to 6- membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; m is 0-3; y is 0-2; and z is 0-1. [00047] In some embodiments, the present disclosure provides a compound of formula I”:
or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from a 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, a 10-membered bicyclic aryl ring, a 3- to 7-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, an 8- to 10-membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 9- to 10-membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; each Rx is independently selected from oxo, halogen, -NO2, -(CH2)yR, –CN, –OR, -N(R)2, -SR, and -(CH2)zY; or: two instances of Rx, together with the atoms to which they are attached, form a 5- to 6- membered saturated, partially unsaturated or aromatic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; Y is selected from -OR, -N(R)2, -O(CH2)2OR, -C(O)R, -CO2R, -OC(O)R, -C(O)N(R)2, - N(R)C(O)R, -SO2R, -SO2N(R)2, and -N(R)SO2R; L is a C1-4 aliphatic chain wherein one or more methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, -C(=O)-, -O-, - S(O)2-, and -CF2-; each Ry is selected from hydrogen, C1-3 alkyl and lower haloalkyl; Ring C is a bivalent group comprising
, wherein Ring C is substituted with 0-3 instances of Rx; each R is independently hydrogen or an optionally substituted group selected from C1-6 aliphatic, a 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, a 3- to 7-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5- to 6- membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
m is 0-3; y is 0-2; and z is 0-1. [00048] As defined generally above, Ring C is a bivalent group comprising
or
. In some embodiments, Ring C is a bivalent group comprising
. In some embodiments, Ring C is a bivalent group comprising
. In some embodiments, Ring C has the structure
. In some embodiments, Ring C has the structure
. In some embodiments, Ring C has the structure
. In some such embodiments, Ring C has the structure
. In some embodiments, Ring C has the structure
. In some embodiments, Ring C has the structure
. In some embodiments, Ring C has the structure
. In some such embodiments, Ring C has the structure
. In some embodiments, Ring C has the structure
. In some embodiments, Ring C has the structure
. In some embodiments, the present disclosure provides a compound of formula II:
or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides a compound of formula II-a:
or a pharmaceutically acceptable salt thereof. [00049] In some embodiments, the present disclosure provides a compound of formula III:
III or a pharmaceutically acceptable salt thereof. [00050] In some embodiments, the present disclosure provides a compound of formula III-a:
or a pharmaceutically acceptable salt thereof. [00051] As defined generally above, Ring A is selected from a 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, a 10-membered bicyclic aryl ring, a 3- to 7-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, an 8- to 10-membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 9- to 10-membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III- a, Ring A is a 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 3- to 7-membered saturated monocyclic carbocyclic ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 3- to 4-membered saturated monocyclic carbocyclic ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 4- to 5-membered saturated monocyclic carbocyclic ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 5- to 6-membered saturated monocyclic carbocyclic ring. In some embodiments of
Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 5- to 7-membered saturated monocyclic carbocyclic ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is selected from cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. [00052] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is phenyl. [00053] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 10- membered bicyclic aryl ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a naphthyl ring. In some such embodiments, Ring A is 2-naphthyl. [00054] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 3- to 7- membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 5- to 6-membered saturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a piperidinyl ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 3- to 7- membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of Rx. [00055] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 3- to 7- membered saturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of Rx. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 5- to 6-membered saturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of Rx. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 6-membered saturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of Rx. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 6-membered saturated monocyclic heterocyclic ring having 1 heteroatom selected from nitrogen, oxygen, and sulfur optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of Rx. In some embodiments of Formula I,
I’, I”, II, II-a, III, or III-a, Ring A is a piperidinyl ring optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of Rx. [00056] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 3- to 7- membered partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of Rx. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 5- to 6-membered partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of Rx. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 5- to 6-membered partially unsaturated monocyclic heterocyclic ring having 1 heteroatom selected from nitrogen, oxygen, and sulfur optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of Rx. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 3,6-dihydro-2H-pyranyl ring optionally fused to an aryl ring, wherein the bicyclic ring formed thereby is substituted with m instances of Rx. [00057] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is an 8- to 10- membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 10-membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 10-membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 10-membered saturated bicyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 10-membered saturated bicyclic heterocyclic ring having 1 heteroatom selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a tetrahydroisoquinolinyl ring is a 1,2,3,4-tetrahydroisoquinolinyl ring.
[00058] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 10- membered partially unsaturated bicyclic heterocyclic ring having 1 heteroatom selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 2H-chromenyl ring. [00059] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 5- to 6- membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 5-membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 5-membered monocyclic heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 5-membered monocyclic heteroaryl ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. [00060] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 6- membered monocyclic heteroaryl ring having 1-3 nitrogen atoms. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 6-membered monocyclic heteroaryl ring having 1-2 nitrogen atoms. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a pyridyl ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a pyrimidinyl ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a pyridazinyl ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a pyrazinyl ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 9- to 10-membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 9-membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III- a, Ring A is a 9-membered bicyclic heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 9-membered bicyclic heteroaryl ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 9-membered bicyclic heteroaryl ring having 2-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of
Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a benzo[d]isoxazolyl, benzo[c][1,2,5]thiadiazolyl ring, a thiazolo[5,4-b]pyridinyl ring, or a benzo[d]thiazolyl ring. [00061] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 9- membered bicyclic heteroaryl ring having 1-4 nitrogen atoms. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 9-membered bicyclic heteroaryl ring having 1-3 nitrogen atoms. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 9- membered bicyclic heteroaryl ring having 1-2 nitrogen atoms. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 9-membered bicyclic heteroaryl ring having 2-3 nitrogen atoms. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 9- membered bicyclic heteroaryl ring having 2-4 nitrogen atoms. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is selected from a 1H-pyrazolo[3,4-b]pyridinyl ring, a 1H- pyrazolo[4,3-b]pyridinyl ring, 2H-pyrazolo[4,3-b]pyridinyl ring, a 3H-imidazo[4,5-b]pyridinyl ring, a 3H-imidazo[4,5-c]pyridinyl ring, a [1,2,3]triazolo[1,5-a]pyridinyl ring, a 7H-pyrrolo[2,3- d]pyrimidinyl ring, a 1H-indazolyl ring, a 2H-indazolyl ring, a 1H-benzo[d]imidazolyl ring, a 1H-pyrazolo[3,4-d]pyrimidinyl ring, 1H-pyrazolo[3,4-d]pyrimidinyl ring, a [1,2,4]triazolo[4,3- b]pyridazinyl ring, 1H-pyrazolo[3,4-b]pyrazinyl ring, a 5H-pyrrolo[2,3-b]pyrazinyl ring, a 1H- pyrrolo[2,3-b]pyridinyl ring, or a [1,2,4]triazolo[1,5-a]pyrimidinyl ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is selected from a 1H-pyrazolo[3,4-b]pyridinyl ring, a 1H-pyrazolo[4,3-b]pyridinyl ring, 2H-pyrazolo[4,3-b]pyridinyl ring, a 3H-imidazo[4,5- b]pyridinyl ring, a 3H-imidazo[4,5-c]pyridinyl ring, a [1,2,3]triazolo[1,5-a]pyridinyl ring, a 7H- pyrrolo[2,3-d]pyrimidinyl ring, a 1H-indazolyl ring, a 2H-indazolyl ring, a 1H- benzo[d]imidazolyl ring, a 1H-pyrazolo[3,4-d]pyrimidinyl ring, 1H-pyrazolo[3,4-d]pyrimidinyl ring, a [1,2,4]triazolo[4,3-b]pyridazinyl ring, 1H-pyrazolo[3,4-b]pyrazinyl ring, a 5H- pyrrolo[2,3-b]pyrazinyl ring, a 1H-pyrrolo[2,3-b]pyridinyl ring, or a [1,2,4]triazolo[1,5- a]pyrimidinyl ring, or a [1,2,4]triazolo[4,3-a]pyridinyl ring. [00062] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 10- membered bicyclic heteroaryl ring having 1-4 nitrogen atoms. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 10-membered bicyclic heteroaryl ring having 1-2 nitrogen atoms. In some such embodiments, Ring A is a cinnolinyl ring, a 1,5-naphthyridinyl ring, a 1,6-naphthyridinyl ring, or a 2,6-naphthyridinyl ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 10-membered bicyclic heteroaryl ring having 1 nitrogen
atom. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a quinolinyl ring or an isoquinolinyl ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is a 5,6,7,8-tetrahydroquinolinyl ring. [00063] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is selected from:
[00064] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is selected from:
[00066] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some
embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is . In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. [00067] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is . In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. [00068] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some
embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. [00069] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some embodiments, of Formula I, I’, I”, II, II-a, III, or III-a Ring A is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ring A is
. In some embodiments, of Formula I, I’, I”, II, II-a, III, or III-a Ring A is
.In some embodiments, of Formula I, I’, I”, II, II-a, III, or III-a Ring A is
.
[00070] As defined generally above, each Rx is independently selected from oxo, halogen, - NO2, -(CH2)yR, –CN, –OR, -N(R)2, -SR, and -(CH2)zY. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Rx is oxo. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Rx is halogen. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Rx is –OR. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Rx is -N(R)2. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Rx is -(CH2)yR. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Rx is -(CH2)zY. It will be appreciated that a Rx group on Ring A can be independent of a Rx group on Ring C. In some embodiments, a Rx group on Ring A is different than a Rx group on Ring C. In some embodiments, the Rx group of Ring A is selected from any of the embodiments as herein described for Rx generally. In some embodiments, the Rx group of Ring C is selected from any of the embodiments as herein described for Rx generally. [00071] As defined generally above, each Y is selected from –OR, -C(O)R, -CO2R, -OC(O)R, -C(O)N(R)2, -N(R)C(O)R, -SO2R, -SO2N(R)2, and -N(R)SO2R; or two instances of Rx, together with the atoms to which they are attached, form a 5- to 6-membered saturated, partially unsaturated or aromatic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Y is –CO2R. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Y is –OR. [00072] As defined generally above for formula I”, Y is selected from -OR, -N(R)2, - O(CH2)2OR, -C(O)R, -CO2R, -OC(O)R, -C(O)N(R)2, -N(R)C(O)R, -SO2R, -SO2N(R)2, and - N(R)SO2R. In some embodiments of formula I”, Y is -OR. In some embodiments of formula I”, Y is -N(R)2. In some embodiments of formula I”, Y is -OCH2CH2OR. [00073] As defined generally above, L is a C1-4 aliphatic chain wherein one or more methylene units of the aliphatic chain are optionally and independently replaced by a group selected from – N(Ry)-, -C(=O)-, -O-, -S(O)2-, and -CF2-. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is a C1-3 aliphatic chain wherein one or more methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, -C(=O)-, -O-, -S(O)2-, and -CF2-. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is a C1-2 aliphatic chain wherein one or more methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, -C(=O)-, -O-, -S(O)2-, and -CF2-. [00074] In some embodiments of Formula I, I’, I’’, II, II-a, III, or III-a, L is a C1-4 aliphatic chain wherein one or more methylene units of the aliphatic chain are optionally and
independently replaced by a group selected from –N(Ry)-, -C(=O)-, -O-, and -CF2-. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is a C1-3 aliphatic chain wherein one or more methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, -C(=O)-, -O-, and -CF2-. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is a C1-2 aliphatic chain wherein one or more methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, -C(=O)-, -O-, and -CF2-. [00075] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is a C1-4 aliphatic chain wherein one or two methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, -C(=O)-, -O-, -S(O)2-, and -CF2-. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is a C1-3 aliphatic chain wherein one or two methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, -C(=O)-, -O-, -S(O)2-, and -CF2-. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is a C1-2 aliphatic chain wherein one or two methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, - C(=O)-, -O-, -S(O)2-, and -CF2-. [00076] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is a C1-4 aliphatic chain wherein one or two methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, -C(=O)-, -O-, and -CF2-. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is a C1-3 aliphatic chain wherein one or two methylene units of the aliphatic chain are optionally and independently replaced by a group selected from – N(Ry)-, -C(=O)-, -O-, and -CF2-. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is a C1-2 aliphatic chain wherein one or two methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, -C(=O)-, -O-, and - CF2-. [00077] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is a C1-4 aliphatic chain wherein one or two methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, -C(=O)-, and -O-. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is a C1-3 aliphatic chain wherein one or two methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, - C(=O)-, and -O-. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is a C1-2
aliphatic chain wherein one or two methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, -C(=O)-, and -O-. [00078] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is –C(=O)-. [00079] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is –N(Ry)-. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is –CH2N(Ry)- or –N(Ry)CH2-. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is –N(Ry)-. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is -OCH2- or -CH2O-. [00080] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is
.
In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is , wherein * denotes the attachment to Ring A. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-
a, L is , wherein * denotes the attachment to Ring A.
[00081] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is
.
In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is , wherein * denotes the attachment to Ring A. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-
a, L is , wherein * denotes the attachment to Ring A. [00082] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is
, wherein *
denotes the attachment to Ring A. In some embodiments of Formula I, I’, I”, II, II-a, III, or III- a, L is
, wherein * denotes the attachment to Ring A. [00083] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is
, wherein * denotes the attachment to Ring A. In some embodiments of Formula I, I’, I”, II, II-a, III, or III- a, L is
, wherein * denotes the attachment to Ring A.
[00084] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is . In some embodiments of Formula
, wherein * denotes the attachment to Ring A. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-
a, L is , wherein * denotes the attachment to Ring A.
[00085] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is . In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is
, wherein * denotes the attachment to Ring A. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is
, wherein * denotes the attachment to Ring A.
[00086] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is
. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is
, wherein * denotes the attachment to Ring A.
[00087] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is , wherein * denotes the attachment to Ring A. [00088] As defined generally above, each Ry is selected from hydrogen, C1-3 alkyl or lower haloalkyl. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ry is hydrogen. Accordingly, in some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is selected from – C(=O)-, –NH-, –CH2NH-, –NHCH2-,
wherein * denotes the attachment to Ring A.
[00089] in some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is selected from – C(=O)-, –NH-, -CH2O-, -OCH2-, –CH2NH-, –NHCH2-,
wherein * denotes the attachment to Ring A. [00090] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, L is
. In some embodiments of Formula I, I’, I’’, II, II-a, III, or III-a, L is
. [00091] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ry is C1-3 alkyl or lower haloalkyl. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ry is C1-3 alkyl. In some such embodiments, Ry is methyl or ethyl. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, Ry is lower haloalkyl. In some such embodiments, Ry is -CF3, -CH2F, or -CF2H. [00092] As defined generally above, each R is independently hydrogen or an optionally substituted group selected from C1-6 aliphatic, a 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, a 3- to 7-membered saturated or partially
unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5- to 6-membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is hydrogen. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted group selected from C1-6 aliphatic, a 3- to 7- membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, a 3- to 7- membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5- to 6-membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. [00093] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is optionally substituted C1-6 aliphatic. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is optionally substituted C1-4 aliphatic. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is optionally substituted C1-2 aliphatic. In some embodiments of Formula I, I’, I”, II, II- a, III, or III-a, R is C1-2 aliphatic optionally substituted with halogen. In some such embodiments, R is –CH3, -CH2CH3, -CH(CH3)2, or –CF3. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is –CH3, -CH2CH3, -CH(CH3)2, -CD(CH3)2, -CH(CH3)CH2CH3, or – CF3. [00094] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is C1-2 aliphatic optionally substituted with –(CH2)0-4ORº, wherein Rº is hydrogen or C1-6 aliphatic. In some such embodiments, Rº is substituted with –(CH2)0-2OR● or –(CH2)0-2OH, wherein R● is C1-4 aliphatic. [00095] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted 3- to 4-membered saturated monocyclic carbocyclic ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted 5- to 6-membered saturated monocyclic carbocyclic ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted cyclopropyl or cyclobutyl ring. [00096] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted phenyl.
[00097] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted 3- to 7-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted 3- to 4- membered saturated monocyclic heterocyclic ring having 1 heteroatom selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an oxetanyl or azetidinyl ring optionally substituted with –(CH2)0-4Rº, –(CH2)0-4ORº, or –(CH2)0- 4CO2Rº, wherein Rº is C1-6 aliphatic. In some such embodiments, Rº is substituted with –(CH2)0- 2OR● or –(CH2)0-2OH, wherein R● is C1-4 aliphatic. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted 5- to 6-membered saturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted piperidinyl ring or a tetrahydropyranyl ring. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is a piperidinyl ring optionally substituted with –(CH2)0-4Rº, –(CH2)0- 4ORº, or –(CH2)0-4CO2Rº, wherein Rº is C1-6 aliphatic. In some such embodiments, Rº is substituted with –(CH2)0-2OR● or –(CH2)0-2OH, wherein R● is C1-4 aliphatic. [00098] In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted 5- to 6-membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted 5-membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted 5-membered monocyclic heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted 5- membered monocyclic heteroaryl ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted 6-membered monocyclic heteroaryl ring having 1-2 nitrogen atoms. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, R is an optionally substituted 6- membered monocyclic heteroaryl ring having 1 nitrogen atom. [00099] As defined generally above, m is 0-3. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-am is 0. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, m is 1. In
some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, m is 2. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, m is 3. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, m is 2. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, m is 0-1. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, m is 2. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, m is 1-2. [000100] As defined generally above, y is 0-2. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, y is 0. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, y is 1. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, y is 2. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, y is 0-1. In some embodiments of Formula I, I’, I”, II, II- a, III, or III-a, y is 1-2. [000101] As defined generally above, z is 0-1. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, z is 0. In some embodiments of Formula I, I’, I”, II, II-a, III, or III-a, z is 1. [000102] In some embodiments, the compound is selected from a compound in Table 1. Table 1.
or a pharmaceutically acceptable salt thereof. [000103] In some embodiments, the compound is not a compound in Table 2. Table 2.
4. Pharmaceutical Compositions [000104] In some embodiments, the present disclosure provides a composition (e.g., a pharmaceutical composition) comprising a compound of this disclosure, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle. In certain embodiments, a composition of this disclosure is formulated for administration to a subject in need of such composition. In some embodiments, a composition of this disclosure is formulated for oral administration to a subject. [000105] The compounds and compositions, according to the method of the present disclosure, may be administered using any amount and any route of administration effective for treating or lessening the severity of a cancer, e.g., a cancer described herein.
[000106] Compositions of the present disclosure may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term "parenteral" as used herein includes subcutaneous, intravenous, intramuscular, intra- articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this disclosure may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non- toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer’s solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. [000107] For this purpose, any bland fixed oil may be employed including synthetic mono- or di-glycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used surfactants, such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation. [000108] Pharmaceutically acceptable compositions of this disclosure may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
[000109] Alternatively, pharmaceutically acceptable compositions of this disclosure may be administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols. [000110] Pharmaceutically acceptable compositions of this disclosure may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs. [000111] Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches may also be used. [000112] For topical applications, provided pharmaceutically acceptable compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers. Carriers for topical administration of compounds of this disclosure include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, provided pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water. [000113] For ophthalmic use, provided pharmaceutically acceptable compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride. Alternatively, for ophthalmic uses, the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum. [000114] Pharmaceutically acceptable compositions of this disclosure may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption
promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents. [000115] Most preferably, pharmaceutically acceptable compositions of this disclosure are formulated for oral administration. [000116] The amount of compounds of the present disclosure that may be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration. Preferably, provided compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a subject receiving these compositions. [000117] It should also be understood that a specific dosage and treatment regimen for any particular subject will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated. The amount of a compound of the present disclosure in the composition will also depend upon the particular compound in the composition. [000118] In some embodiments, the present disclosure provides a composition (e.g., a pharmaceutical composition) comprising a compound of this disclosure, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle. In certain embodiments, a composition of this disclosure is formulated for administration to a subject in need of such composition. In some embodiments, a composition of this disclosure is formulated for oral administration to a subject. [000119] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar--agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i)
lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents. [000120] Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like. [000121] The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. [000122] Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl
alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents. [000123] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer’s solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables. [000124] Injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use. [000125] Dosage forms for topical or transdermal administration of a compound of this disclosure include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this disclosure. Additionally, the present disclosure contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel. [000126] Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this disclosure with suitable non-irritating
excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound. [000127] In order to prolong the effect of a compound of the present disclosure, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide- polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues. [000128] Pharmaceutically acceptable compositions of this disclosure can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated. In certain embodiments, the compounds of the disclosure may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect. [000129] Provided compounds and compositions thereof are effective over a wide dose range. In some embodiments, a dose is from about 0.01 to about 1000 mg, from about 0.5 to about 100 mg, from about 1 to about 50 mg, or from about 5 to about 100 mg. Exact doses may depend upon routes of administration, forms in which compounds are administered, subjects (e.g., body weight, age, body surface area, etc.), conditions, disorders or diseases, and/or preferences and experiences of physicians. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the condition,
disorder, or disease, e.g., cancer, the particular agent, its mode of administration, and the like. In some embodiments, a fixed dose is administered. In some embodiments, two or more doses are about the same amount. In some embodiments, one or more doses are independently more than one or more other doses. For example, in some embodiments, one or more loading doses each independently of a higher amount are administered before one or more maintenance doses each independently of a lower amount. In some embodiments, two or more or all loading doses are about the same amount. In some embodiments, a loading dose is of a higher amount than another loading dose. In some embodiments, two or more or all maintenance doses are about the same amount. In some embodiments, a maintenance dose is of a higher amount than another maintenance dose. [000130] Compounds of the disclosure are preferably formulated in dosage unit form for ease of administration and uniformity of dosage. The expression “dosage unit form” as used herein refers to a physically discrete unit of agent appropriate for the subject to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present disclosure will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular subject or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts. [000131] Among other things, the present disclosure provides kits comprising any of the compounds and compositions described herein. In some embodiments, a kit comprises a solid composition. In some embodiments, a kit comprises a tablet, capsule, or pill. In some embodiments, a kit comprises a liquid composition. In some embodiments, a kit comprises instructions for performing any of the methods described herein. 5. Characterization and Assessment [000132] As appreciated by those skilled in the art, various technologies may be utilized to characterize and/or assess provided technologies in accordance with the present disclosure.
Certain useful technologies are described in the Examples. As demonstrated, among other things, the present disclosure describes various in vitro technologies suitable for assessing and characterizing provided technologies. In some embodiments, provided technologies are characterized and/or assessed, e.g., in in vitro systems, e.g., in cells. [000133] In some embodiments, provided technologies (e.g., compounds, compositions) are characterized and/or assessed using various in vitro assays known in the art. For example, in some embodiments, compounds are assessed using a HiBiT assay. The HiBiT assay reportedly can be used to provide quantitative measurement of proteins in a system, e.g., an in vitro system, e.g., a cell or population of cells. A HiBiT assay can be conducted in a high-throughput manner. By tagging a target protein with a Small BiT (HiBiT tag), the protein levels can reportedly be measured through the addition of the LgBiT component to form an active nanoluciferase enzyme. Luminescence generated by the nanoluciferase enzyme under assay conditions can then reportedly be quantitatively measured to provide quantitation of Small BiT-tagged protein levels. The HiBiT system is described in, e.g., Schwinn et al. ACS Chem Biol.2018 Feb 16;13(2):467- 474. In some embodiments, Small BiT-tagged CK1α is used in a HiBiT assay. In some embodiments, a HiBiT assay is used to assess level of targeted protein in a system, e.g., an in vitro system. In some embodiments, a HiBit assay is used to assess level of CK1α protein in a system, e.g., an in vitro system. In some embodiments, a HiBiT assay is used to assess level of targeted protein degradation in a system, e.g., an in vitro system, by a provided compound or composition. In some embodiments, a HiBit assay is used to assess level of CK1α protein degradation in a system, e.g., an in vitro system, by a provided compound or composition. In some embodiments, a HiBiT assay is conducted as described herein, e.g., in an example. [000134] In some embodiments, compounds are assessed using an assay to assess level of CK1α protein or level of CK1α activity in a system. In some embodiments, level of CK1α protein is assessed by an immunoassay. In some embodiments, level of CK1α protein is assessed by Western blot. In some embodiments, level of CK1α protein is assessed by capillary-based immunoassay. Various additional methods for assessing levels of CK1α protein or activity are described in the art, including those described in WO 2021/222542. [000135] In some embodiments, compounds are assessed using a TOPflash assay. The TOPflash assay reportedly utilizes a reporter plasmid comprising multiple (e.g., two sets of three) copies of a wild-type TCF binding site upstream of a thymidine kinase minimal promoter
and luciferase open reading frame (ORF). The TOPflash reporter plasmid is described in, e.g., Korinek et al. Science.1997 Mar 21;275(5307):1784-7. Using the TOPflash plasmid, an assay can reportedly be performed to provide quantitative measurement (through measurement of luciferase-driven luminescence) of β-catenin/TCF activity in a system (e.g., an in vitro system, e.g., a cell), which activates the expression from the luciferase ORF. This quantitative measurement may reportedly be used to assess Wnt signaling pathway activation in a system. In some embodiments, a TOPflash assay is used to assess level of Wnt signaling pathway activation in a system, e.g., an in vitro system, by a provided compound or composition. In some embodiments, a TOPflash assay is used to assess level of β-catenin activity in a system, e.g., an in vitro system, by a provided compound or composition. In some embodiments, a TOPflash assay is conducted as described herein, e.g., in an example. [000136] In some embodiments, compounds are assessed using an assay to assess level of p53 protein or level of p53 activity in a system. In some embodiments, level of p53 protein is assessed by an immunoassay. In some embodiments, level of p53 protein is assessed by Western blot. In some embodiments, level of p53 protein is assessed by capillary-based immunoassay. Various additional methods for assessing levels of p53 protein or activity are described in the art, including those described in WO 2021/222542. [000137] In some embodiments, a provided compound or composition is characterized or assessed in an in vivo system. In some embodiments, a provided compound or composition is characterized or assessed in an animal, e.g., a mouse, rat, pig, dog, non-human primate, e.g., a monkey. In some embodiments, a provided compound or composition is characterized or assessed in a human. In some embodiments, a provided compound or composition is characterized in xenograft model, e.g., a xenograft mouse model. Various xenograft models for assessment of anti-cancer and/or anti-tumor properties of compounds and compositions thereof are known in the art. [000138] Those skilled in the art reading the present disclosure will readily appreciate that other technologies, e.g., in vitro models (e.g., cell lines) for various conditions, disorders, or diseases, animals models for various conditions, disorders, or diseases, clinical trials, etc. may be designed and/or utilized to assess provided technologies (e.g., compounds, compositions, methods, etc.) in accordance with the present disclosure.
6. Uses and Applications [000139] As appreciated by those skilled in the art, compounds described herein are useful for many purposes. In some embodiments, provided technologies (e.g., compounds, compositions, methods, etc.) are useful for treating conditions, disorders, or diseases, e.g., various cancers as described herein. In some embodiments, provided technologies (e.g., compounds, compositions, methods, etc.) are useful for modulating cellular pathways, e.g., Wnt signaling pathway in a system. In some embodiments, provided technologies (e.g., compounds, compositions, methods, etc.) are useful for decreasing level of target proteins, e.g., CK1α and/or level of activity of target proteins, e.g., CK1α in a system. [000140] The Wnt signaling pathway, which may also be referred to the Wnt/β-catenin signaling pathway, reportedly involves a variety of proteins and modulates gene expression in cells (Komiya, Y. and Habas, R. Organogenesis.2008 Apr;4(2):68-75). Wnt proteins are reportedly secreted glycoproteins which bind to receptors belonging to the Frizzled (Fz) family of receptors on the outside of cells. Further, the interaction of a Wnt protein with a co-receptor may also be necessary; reported co-receptors include lipoprotein receptor-related protein (LRP) 5/6 and various receptor tyrosine kinases (RTKs). Wnt binding with these receptors has been reported as leading to modulation of the protein Axin, which may be translocated to membrane, bound to LRP 5/6, destabilized, and degraded. Additionally, upon Wnt binding to Fz, a cytoplasmically located Dishevelled (Dsh) protein may reportedly interact with Fz, become phosphorylated, and inhibit activity of glycogen synthase kinase 3 (GSK3). These actions have been reported to disrupt a destruction complex, which comprises Axin, GSK3, adenomatosis polyposis coli (APC), β-transducin repeat-containing E3 ubiquitin protein ligase (β-TrCP), and casein kinase 1α (CK1α). [000141] In the absence of Wnt signaling pathway activation, the destruction complex described above has been reported to target β-catenin for destruction. Such targeting reportedly involves phosphorylation of β-catenin by the proteins of the destruction complex, e.g., GSK3 and CK1α, and ensuing ubiquitination and proteasomal degradation. Accordingly, β-catenin levels are decreased and β-catenin is not translocated to the nucleus to effect gene transcription through interactions with TCF/LEF transcription factors, which reportedly repress target gene transcription in the absence of β-catenin. Whereas, with Wnt signaling pathway activation, the
destruction complex is reportedly disrupted, β-catenin degradation is reduced, and an increased level of β-catenin can be translocated to the nucleus to modulate transcription of target genes. [000142] Various genes have been reported as being regulated by Wnt signaling pathway activation, including genes related to cancer cell functioning, growth, and proliferation (Zhang, Y. and Wang, X. J Hematol Oncol.2020 Dec 4;13(1):165). In some embodiments, Wnt signaling pathway modulates expression of genes involved in any of the following non-limited functions: cellular proliferation, cellular differentiation, stem cell renewal, tissue homeostasis, apoptosis, migration, and/or invasion. In some embodiments, Wnt signaling pathway modulates expression of genes over-expressed or under-expressed in tumor cells. In some embodiments, Wnt signaling pathway modulates expression of genes over-expressed or under-expressed in cancer cells. [000143] Accordingly, without wishing to be bound by any particular theory, modulation of one or more proteins of a Wnt signaling pathway may be conducted to increase or decrease level of Wnt signaling pathway activation and/or downstream gene expression. In some embodiments, modulation of one or more proteins may modulate level of Wnt signaling pathway activation in a system, e.g., a cell. In some embodiments, modulation of a level of Wnt signaling pathway activation may involve increasing or decreasing Wnt signaling pathway activation. In some embodiments, modulation of a level of Wnt signaling pathway activation may involve increasing Wnt signaling pathway activation. In some embodiments, modulation of one or more proteins may modulate level of β-catenin in a system, e.g., a cell. In some embodiments, modulation of a level of β-catenin may involve increasing or decreasing the level of β-catenin. In some embodiments, modulation of one or more proteins may modulate level of expression of one or more genes in a system, e.g., a cell. In some embodiments, modulation of a level of expression of one or more genes may involve increasing or decreasing the level of expression of one or more genes. In some embodiments, modulation of one or more proteins may involve increasing or decreasing a level of one or more proteins. In some embodiments, modulation of one or more proteins may involve increasing or decreasing a level of activity of one or more proteins. In some embodiments, modulation of one or more proteins may involve degradation of the one or more proteins. In some embodiments, the present disclosure provides a method of modulating an oncogenic pathway, the method comprising contacting a biological sample with, or administering to a subject in need thereof, a compound of formula I, or a pharmaceutically salt
thereof. In some embodiments, a method of modulating an oncogenic pathway results in the activation of the pathway. In some embodiments, a method of modulating an oncogenic pathway results in the inhibition of the pathway. In some embodiments, the present disclosure provides a method of activating an oncogenic pathway, the method comprising contacting a biological sample with, or administering to a subject in need thereof, a compound of formula I, or a pharmaceutically salt thereof. In some such embodiments, the oncogenic pathway is a WNT- mediated pathway. [000144] The tumor suppressor gene, p53 reportedly regulates transcription of a variety of genes, including those involved in, e.g., DNA damage repair, cell cycle progression, and apoptosis. Further, p53 has been reported as the most frequently mutated gene in human cancers, and even in cancers without a p53 mutation, reduced levels of p53 activity have been reported. Accordingly, increasing and/or restoring levels of wild-type and/or functional p53 and p53 activity remains a potential route of action for cancer therapeutic agents. Levels of p53 protein and activity has been found to be regulated by a p53/MDM2/MDMX pathway. MDM2 is reportedly an E3 ubiquitin ligase that binds to and ubiquitinates p53 – leading to inhibition of p53 and degradation of p53 by the proteasome. MDMX reportedly acts a modulator of p53 activity by binding to p53 and inhibiting p53 transcriptional activity. Studies have shown that CK1α is able to interact with MDM2 and MDMX. CK1α has been reported to interact with MDM2 and thusly promote the binding of MDM2 to p53 and the subsequent ubiquitination, inhibition, and degradation of p53 (Huart, A. et al. J Biol Chem.2009 Nov 20;284(47):32384- 94). Additionally, CK1α has been reported to phosphorylate MDMX, which allows MDMX to interact with and inhibit p53 (Chen, L. et al. Mol Cell Biol.2005 Aug;25(15):6509-20). Thus, CK1α may act a positive regulator of MDM2 and/or MDMX activity, which in turn acts to decrease levels of p53 protein and activity. Potential CK1α involvement in p53/MDM2/MDMX pathways is discussed in, e.g., Jiang, S. et al. Cell Commun Signal.2018 May 24;16(1):23. Potential downregulation of p53 through the p53/MDM2/MDMX pathway has been reported in some cancers, including, e.g., acute myeloid leukemia (AML), wherein high levels of CK1α is associated with suppression of p53 and decreased overall survival (Xu, W. et al. Oncol Rep. 2020 Nov;44(5):1895-1904). [000145] Accordingly, without wishing to be bound by any particular theory, modulation of one or more proteins in a p53/MDM2/MDMX pathway may be conducted to increase or
decrease level of p53 protein and/or activity and/or downstream gene expression. In some embodiments, modulation of one or more proteins may modulate level of p53 protein in a system, e.g. a cell. In some embodiments, modulation of one or more proteins may modulate level of p53 activity in a system, e.g. a cell. In some embodiments, modulation of one or more proteins may modulate level of p53 binding activity in a system, e.g., a cell. In some embodiments, modulation of one or more proteins may modulate level of degradation of p53 protein in a system, e.g., a cell. In some embodiments, modulation of one or more proteins may modulate level of MDM2 activity in a system, e.g. a cell. In some embodiments, modulation of one or more proteins may modulate level of expression of one or more genes in a system, e.g., a cell. In some embodiments, modulation of one or more proteins may modulate level of expression of one or more p53-regulated genes in a system, e.g., a cell. In some embodiments, modulation of a level of expression of one or more genes may involve increasing or decreasing the level of expression of one or more genes. In some embodiments, modulation of one or more proteins may involve increasing or decreasing a level of one or more proteins. In some embodiments, modulation of one or more proteins may involve increasing or decreasing a level of activity of one or more proteins. In some embodiments, modulation of one or more proteins may involve degradation of the one or more proteins. In some embodiments, the present disclosure provides a method of inhibiting an oncogenic pathway, the method comprising contacting a biological sample with, or administering to a subject in need thereof, a compound of formula I, or a pharmaceutically salt thereof. In some such embodiments, the oncogenic pathway is a p53-mediated pathway. [000146] Casein kinase 1α (CK1α) refers to a protein encoded by, in humans, the CSNK1A1 gene. CK1α may also be known as casein kinase 1 isoform alpha, CKI-alpha, or CK1. Various CK1α sequences are readily available to those of skill in the art, including NCBI Reference Protein Sequences Accession Nos. NP_001020276.1, NP_001883.4, NP_001258670.1, and NP_001258671.1. Various technologies, e.g., assays, cells, etc., associated with CK1α have also been reported and can be utilized for characterization and/or assessment of provided technologies (e.g., compounds, compositions, methods, etc.) in accordance with the present disclosure. [000147] CK1α reportedly belongs to a family of proteins that act as serine/threonine kinases. As part of a destruction complex, CK1α phosphorylates β-catenin, which acts as a signal transduction protein in the Wnt signaling pathway. The phosphorylation of β-catenin by CK1α
has been reported to contribute to the subsequent ubiquitination and degradation of β-catenin. Other components which have been indicated to be part of the β-catenin destruction complex include, e.g., APC, GSK3, and Axin. CK1α reportedly also phosphorylates APC, which in turn assists in the association of APC with β-catenin (Ferrarese, A. et al. Biochemistry.2007 Oct 23;46(42):11902-10). In addition to functioning as a component of the β-catenin destruction complex, studies have suggested potential additional roles for CK1α in cellular functioning due to widespread localization in the cell and ubiquitous expression of the protein in different types of cells and tissues (Jiang, S. et al. Cell Commun Signal.2018 May 24;16(1):23). [000148] In some embodiments, a provided compound interacts with a CK1α polypeptide or protein. In some embodiments, a provided compound binds a CK1α polypeptide or protein. In some embodiments, a provided compound binds a CK1α polypeptide or protein and binds an additional polypeptide or protein. In some embodiments, a provided compound binds a CK1α polypeptide or protein and binds a CRBN polypeptide or protein. [000149] Cereblon (CRBN) refers to a protein encoded by, in humans, the CRBN gene. Various CRBN sequences are readily available to those of skill in the art, including NCBI Reference Protein Sequences Accession Nos. NP_057386.2 and NP_001166953.1. Various technologies, e.g., assays, cells, etc., associated with CRBN have also been reported and can be utilized for characterization and/or assessment of provided technologies (e.g., compounds, compositions, methods, etc.) in accordance with the present disclosure. [000150] CRBN reportedly acts a substrate receptor as part of the E3 ubiquitin ligase complex, which also includes CUL4, RBX1, and DDB1. As the substrate receptor for the complex, CRBN directly binds to proteins targeted for ubiquitin-mediated degradation. Through degradation of target proteins, CRBN may reportedly play a role in regulation of various cellular processes or pathways, including, e.g., the TLR4 signaling pathway and ATP metabolism. Studies have further shown that CRBN can be facilitated to bind to specific target proteins through the usage of immune modulatory drugs, therein leading to targeted degradation of the target proteins (Petzold et al. Nature.2016 Apr 7;532(7597):127-30). [000151] In some embodiments, a provided compound interacts with an E3 ubiquitin ligase protein. In some embodiments, a provided compound binds an E3 ubiquitin ligase protein. In some embodiments, a provided compound binds an E3 ubiquitin ligase protein and an additional polypeptide or protein. In some embodiments, a provided compound binds an E3 ubiquitin
ligase protein and a CK1α polypeptide or protein. In some embodiments, a provided compound interacts with a CRBN polypeptide or protein. In some embodiments, a provided compound binds a CRBN polypeptide or protein. In some embodiments, a provided compound binds a CRBN polypeptide or protein and an additional polypeptide or protein. In some embodiments, a provided compound binds a CRBN polypeptide or protein and a CK1α polypeptide or protein. [000152] In some embodiments, provided technologies (e.g., compounds, compositions) promote or facilitate binding of CRBN to target polypeptides or proteins. In some embodiments, provided technologies (e.g., compounds, compositions) promote or facilitate binding of CRBN to CK1α polypeptides or proteins. In some embodiments, provided technologies (e.g., compounds, compositions) promote or facilitate CRBN-mediated degradation of target polypeptides or proteins. In some embodiments, provided technologies (e.g., compounds, compositions) promote or facilitate CRBN-mediated degradation of CK1α polypeptides or proteins. In some embodiments, provided technologies (e.g., compounds, compositions) modulate level of CRBN- mediated degradation of CK1α polypeptides or proteins. In some embodiments, provided technologies (e.g., compounds, compositions) increase level of CRBN-mediated degradation of CK1α polypeptides or proteins. [000153] Studies have suggested that level of cellular pathway activity in cancers often follow a “just right” model, wherein inhibition of an oncogenic cellular pathway may prove lethal for cancer cells or wherein further activation (hyperactivation) may also prove lethal for cancer cells. Currently available cancer therapeutic agents are predominantly targeted to provide inhibition of oncogenic pathways to perturb cancer cell growth, proliferation, and division and to kill cancer cells. The Wnt signaling pathway has been previously implicated as an oncogenic in a variety of cancers, including cancers comprising a mutation of APC, e.g., APC mutant colorectal cancer. Mutations of APC in such cancers reportedly provide oncogenic activation of Wnt signaling pathway and oncogenic activation of target genes by increased β-catenin stability and/or activity. In some embodiments, Wnt signaling pathway is an oncogenic pathway. Recent research has indicated that further perturbation of proteins in the destruction complex (e.g., Axin, CK1α) in the Wnt signaling pathway may be used to further activate the Wnt signaling pathway and genes regulated thereby. The further activation of the Wnt signaling pathway reportedly results in lethal hyperactivation and death of cancer cells (Chang et al. Nat Genet.2023 Sep 25). Without wishing to be bound by any particular theory, the present disclosure provides the unique insight
that activation of oncogenic pathways, e.g., Wnt signaling pathway, by modulating CRBN- mediated degradation of CK1α using compounds described herein can be used to treat cancers, e.g., APC mutant cancers, e.g., APC mutant colorectal cancer. [000154] Insights contained herein recognize that hyperactivation of cellular pathways provides additional routes for cancer therapeutics to target various cancers. For example, such hyperactivation may be particularly useful in treatment of cancers comprising certain mutations (e.g., APC-mutant colorectal cancers), which may not be effectively targeted by available cancer therapeutics, or in treatment of subjects comprising cancers which have developed resistance to currently available inhibitory cancer therapeutics. Without wishing to be bound by any particular theory, exemplary compounds, compositions, and methods described herein can, among other things, provide anti-cancer properties and treatment of various cancers through the hyperactivation of cellular pathways, e.g., Wnt signaling pathway. See Chang et al. Nat Genet. 2023 Sep 25. [000155] In some embodiments, the present disclosure provides methods of hyperactivating an cellular pathway in a system, comprising administering or delivering an effective amount of a provided compound or composition thereof to a system. In some embodiments, the present disclosure provides methods of hyperactivating an oncogenic pathway in a system, comprising administering or delivering an effective amount of a provided compound or composition thereof to a system. In some embodiments, the present disclosure provides methods of hyperactivating a Wnt signaling pathway in a system, comprising administering or delivering an effective amount of a provided compound or composition thereof to a system. In some embodiments, the present disclosure provides methods of causing death of a cancer cell, comprising hyperactivating a cellular pathway in the cancer cell. In some embodiments, the present disclosure provides methods of causing death of a cancer cell, comprising hyperactivating an oncogenic pathway in the cancer cell. In some embodiments, the present disclosure provides methods of causing death of a cancer cell, comprising hyperactivating a Wnt signaling pathway in the cancer cell. [000156] In some embodiments, the present disclosure provides the insight that compounds which are able to bind at the interface of an E3 ubiquitin ligase protein, e.g., CRBN, and another protein such as, e.g., CK1α, can activate an oncogenic pathway to the point of lethality. Without wishing to be bound by any particular theory, it is believed that compounds of formula I interact
with residues of both CK1α and CRBN. In some such embodiments, the
moiety of formula I comprises one or more atoms or groups that interact with the side chains of CK1α Lys18, CK1α Arg21, CRBN Glu377, and/or CRBN His353. In certain embodiments, Ring A or a substituent on Ring A (i.e., an Rx group) comprises at least one atom that interacts with CK1α Arg21. In some embodiments, L comprises at least one atom that interacts with CK1α Lys18, CRBN Glu377, and/or CRBN His353. [000157] In some embodiments, the present disclosure provides a method of activating a cellular pathway, the method comprising contacting a biological sample with, or administering to a subject in need thereof, a compound of formula I, or a pharmaceutically salt thereof. In some embodiments, a cellular pathway is Wnt signaling pathway. In some embodiments, the present disclosure provides a method of activating an oncogenic pathway, the method comprising contacting a biological sample with, or administering to a subject in need thereof, a compound of formula I, or a pharmaceutically salt thereof. In some embodiments, the oncogenic pathway is a Wnt-mediated pathway. In some embodiments, a method described herein comprises administering or delivering to a subject (or contacting a biological sample with) a compound selected from those in Table 1. In some embodiments, a method described herein comprises administering or delivering to a subject (or contacting a biological sample with) a compound selected from those in Tables 1 and 2. [000158] In some embodiments, the present disclosure provides a method of modulating association of CK1α polypeptides and CRBN polypeptides in a system, comprising administering or delivering to the system an effective amount of a provided compound or composition thereof to the system. In some embodiments, the present disclosure provides a method of increasing association of CK1α polypeptides and CRBN polypeptides in a system, comprising administering or delivering to the system an effective amount of a provided compound or composition thereof to the system. [000159] Without wishing to be bound by any theory, the stabilization of interactions between CK1α polypeptides and CRBN polypeptides may increase such interactions and the degradation of CK1α polypeptides. In some embodiments, the present disclosure provides a method of modulating level of CK1α polypeptide in a system, comprising administering or delivering an effective amount of a provided compound or composition thereof to the system. In some
embodiments, the present disclosure provides a method of decreasing level of CK1α polypeptide in a system, comprising administering or delivering an effective amount of a provided compound or composition thereof to the system. In some embodiments, the present disclosure provides a method of modulating level of CK1α activity in a system, comprising administering or delivering to the system an effective amount of a provided compound or composition thereof to the system. In some embodiments, the present disclosure provides a method of decreasing level of CK1α activity in a system, comprising administering or delivering to the system an effective amount of a provided compound or composition thereof to the system. [000160] Without wishing to be bound by any particular theory, decreasing level of CK1α polypeptides and/or activity in a system may decrease active destruction complexes, which may increase level of Wnt pathway activation by decreasing β-catenin degradation. In some embodiments, the present disclosure provides a method of modulating level of Wnt pathway activation in a system, comprising administering or delivering an effective amount of a provided compound or composition thereof to the system. In some embodiments, the present disclosure provides a method of increasing Wnt pathway activation in a system, comprising administering or delivering an effective amount of a provided compound or composition thereof to the system. In some embodiments, the present disclosure provides a method of modulating level of β-catenin degradation in a system, comprising administering or delivering an effective amount of a provided compound or composition thereof to the system. In some embodiments, the present disclosure provides a method of decreasing level of β-catenin degradation in a system, comprising administering or delivering an effective amount of a provided compound or composition thereof to the system. In some embodiments, the present disclosure provides a method of modulating level of β-catenin in a system, comprising administering or delivering an effective amount of a provided compound or composition thereof to the system. In some embodiments, the present disclosure provides a method of increasing level of β-catenin in a system, comprising administering or delivering an effective amount of a provided compound or composition thereof to the system. In some embodiments, the present disclosure provides a method of modulating expression of Wnt pathway target genes in a system, comprising administering or delivering an effective amount of a provided compound or composition thereof to the system. In some embodiments, the present disclosure provides a method of increasing
expression of Wnt pathway target genes in a system, comprising administering or delivering an effective amount of a provided compound or composition thereof to the system. [000161] In some embodiments, the present disclosure provides a method of causing death of a cancer cell. In some embodiments, the present disclosure provides a method of causing death of a cancer cell, comprising contacting the cancer cell with an effective amount of a provided compound. In some embodiments, the present disclosure provides a method of causing death of a cancer cell, comprising contacting the cancer cell with an effective amount of a compound of Formula I. In some embodiments, the present disclosure provides a method of causing death of a cancer cell, comprising contacting the cancer cell with an effective amount of a compound of Table 1. In some embodiments, the present disclosure provides a method of causing death of a cancer cell, comprising contacting the cancer cell with an effective amount of a compound of Table 2. In some embodiments, a cancer cell is a cancer cell in a system. In some embodiments, a cancer cell is a cancer cell in a subject. [000162] In some embodiments, a system comprises CK1α polypeptides. In some embodiments, a system comprises CRBN polypeptides. In some embodiments, a system comprises CK1α polypeptides and CRBN polypeptides. In some embodiments, a system expresses CK1α polypeptides. In some embodiments, a system expresses CRBN polypeptides. In some embodiments, a system expresses CK1α polypeptides and CRBN polypeptides. [000163] In some embodiments, a system comprises an in vitro system. In some embodiments, a system is an in vitro system. In some embodiments, a system comprises an in vivo system. In some embodiments, a system is an in vivo system. [000164] In some embodiments, a system comprises a cell. In some embodiments, a system is a cell. In some embodiments, a cell is a tumor cell. In some embodiments, a cell is a cancer cell. In some embodiments, a system comprises a tissue. In some embodiments, a system is a tissue. In some embodiments, a system comprises a tumor. In some embodiments, a system is a tumor. In some embodiments, a system comprises an organ. In some embodiments, a system is an organ. In some embodiments, a system comprises an organism. In some embodiments, a system is an organism. In some embodiments, a system is a subject. In some embodiments, a system is an animal. In some embodiments, a system is a mammal, e.g., a mouse, a rat, a rabbit, a pig, a dog, a non-human primate, e.g., a monkey. In some embodiments, a system is a human.
[000165] In some embodiments, a level is reduced by at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% compared to absence of a provided compound or composition and/or presence of a reference compound or composition. In some embodiments, a level is increased by at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% compared to absence of a provided compound or composition and/or presence of a reference compound or composition. In some embodiments, such a reduction is achieved in a system, e.g., using various suitable assays (e.g., in vitro assays (e.g., in vitro cell-based assays), assays described in, e.g., the Examples). In some embodiments, a reference compound associates with CK1α and/or CRBN. In some embodiments, a reference compound binds CK1α and/or CRBN. In some embodiments, a reference compound is a compound disclosed in a Table herein. In some embodiments, a reference compound is a compound disclosed in Table 1. In some embodiments, a reference compound is a compound disclosed in Table 2. [000166] In some embodiments, the present disclosure provides methods using provided technologies (e.g., compounds, compositions). In some embodiments, a method comprises administering or delivering a compound or composition thereof described herein. In some embodiments, a method comprises administering or delivering a therapeutically effective amount of a compound or composition thereof described herein. In some embodiments, a method comprises administering or delivering a compound or composition thereof described herein and one or more cancer therapeutics, e.g., one or more cancer therapeutics described herein. In some embodiments, a method comprises administering or delivering a therapeutically effective amount of a compound or composition thereof described herein and a therapeutically effective amount of one or more cancer therapeutics, e.g., one or more cancer therapeutics described herein. [000167] In some embodiments, the present disclosure provides methods of treating, e.g., a condition, disorder, or disease using provided technologies (e.g., compounds, compositions). In some embodiments, a method of treating a condition, disorder, or disease comprises administering or delivering a compound or composition thereof described herein. In some embodiments, a method of treating a condition, disorder, or disease comprises administering or delivering a therapeutically effective amount of a compound or composition thereof described herein. In some embodiments, a method of treating a condition, disorder, or disease comprises
administering or delivering a compound or composition thereof described herein and one or more cancer therapeutics, e.g., one or more cancer therapeutics described herein. In some embodiments, a method of treating a condition, disorder, or disease comprises administering or delivering a therapeutically effective amount of a compound or composition thereof described herein and a therapeutically effective amount of one or more cancer therapeutics, e.g., one or more cancer therapeutics described herein. In some embodiments, a method of treating a cancer comprises administering or delivering a compound or composition thereof described herein. In some embodiments, a method of treating a cancer comprises administering or delivering a therapeutically effective amount of a compound or composition thereof described herein. In some embodiments, a method of treating a cancer comprises administering or delivering a compound or composition thereof described herein and one or more cancer therapeutics, e.g., one or more cancer therapeutics described herein. In some embodiments, a method of treating a cancer comprises administering or delivering a therapeutically effective amount of a compound or composition thereof described herein and a therapeutically effective amount of one or more cancer therapeutics, e.g., one or more cancer therapeutics described herein. [000168] In some embodiments, a condition, disorder, or disease comprises or is a neoplastic disorder. In some embodiments, a condition, disorder, or disease comprises or is a tumor. In some embodiments, a condition, disorder, or disease comprises or is a cancer, e.g., a cancer described herein. [000169] Provided technologies (e.g., compounds, compositions, methods) may be used in treatment of any cancer or tumor described herein. Cancer is a disease marked by the unregulated cellular growth and division. Such uncontrolled growth may result in a tumor, which may then become cancerous through spreading to other tissues and/or locations in the body. Cancers may be described in a variety of terms, including type of cell that formed the cancer, tissue or organ from which the cancer originated, rate of progression, etc. [000170] In some embodiments, a cancer may specifically be of any of the following non- limiting types: acinar cell carcinoma, acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), adenoid cystic carcinoma, adrenocortical carcinoma, agnogenic myeloid leukemia, anal cancer, ameoblastoma, appendix cancer, astroblastoma, astrocytoma, basal cell carcinoma, bile duct cancer, bladder cancer, blood cancer, bone cancer, bowel cancer, brain cancer, breast cancer, bronchioloalveolar carcinoma, bronchogenic carcinoma, Burkitt
lymphoma, cancer of unknown primary, cervical cancer, cholangiocarcinoma, chondroblastoma, chondrosarcoma, chordoma, chromophobe carcinoma, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), colorectal cancer, connective tissue cancer, craniopharyngioma, cutaneous lymphoma, cutaneous T-cell lymphoma, cystadenocarcinoma, ductal carcinoma, endocrine cancer, endometrial cancer, ependymoma, erythroleukemia, esophageal cancer, esophageal squamous cell carcinoma, essential thrombocythemia (ET), esthesioneuroblastoma, Ewing sarcoma, eye cancer, fallopian tube cancer, follicular adenocarcinoma, gallbladder cancer, gastric cancer, germ cell cancer, giant and spindle cell carcinoma, glomangiosarcoma, granular cell carcinoma, hairy cell leukemia, head and neck cancer, head and neck squamous cell carcinoma, hemangiopericytoma, hepatocellular carcinoma (HCC), Hodgkin lymphoma, hypopharyngeal cancer, infiltrating duct carcinoma, intraocular melanoma, juxtacortical osteosarcoma, Kaposi sarcoma, kidney cancer, large B-cell lymphoma, laryngeal cancer, leukemia, lip and oral cavity cancer, lipid cell tumor, liver cancer, lung cancer, lung squamous cell carcinoma, lymphoepithelial carcinoma, lymphoma, lymphosarcoma cell leukemia, medulloblastoma, melanoma, Merkel cell carcinoma (MCC), mesothelioma, midline tract carcinoma, mouth cancer, multiple myeloma, myelodysplastic syndrome (MDS), myeloma, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, nasopharyngeal carcinoma, neuroblastoma, nonencapsulating sclerosing carcinoma, non-Hodgkin lymphoma, non-small cell lung cancer, ocular cancer, odontosarcoma, oral cancer, osteosarcoma, ovarian cancer, oxyphilic adenocarcinoma, pancreatic cancer, papillary adenocarcinoma, papillary carcinoma, paraganglioma, paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, peritoneal cancer, pharyngeal cancer, pheochromocytoma, pilomatrix carcinoma, plasmacytoma, primary bone cancer, primary central nervous system lymphoma, primary effusion lymphoma (PEL), primary liver cancer, primary peritoneal cancer, prostate cancer, rectal cancer, renal cancer, renal cell carcinoma (RCC), retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sarcoma, secondary bone cancer, secondary liver cancer, skin cancer, small cell lung cancer, small intestine cancer, soft tissue sarcoma, squamous cell carcinoma, squamous cell skin cancer, stomach cancer, testicular cancer, throat cancer, thymic carcinoma, thymoma, thyroid cancer, trabecular adenocarcinoma, transitional cell cancer, transitional cell carcinoma, transitional cell cancer of the renal pelvis and ureter, urethral cancer, uterine cancer, uterine sarcoma, urothelial cancer, vaginal cancer, or vulvar cancer.
[000171] In some embodiments, a cancer comprises one or more particular mutations. In some embodiments, a cancer comprises one or more mutations of a component (e.g., a gene or protein encoded thereby) of a Wnt signaling pathway. In some embodiments, a cancer comprises one or more activating mutations of a component (e.g., a gene or protein encoded thereby) of a Wnt signaling pathway. In some embodiments, an activating mutation increases Wnt signaling pathway activation. In some embodiments, an activating mutation decreases degradation of β- catenin. In some embodiments, an activating mutation is a mutation in β-catenin or a mutation of a deconstruction complex gene or protein encoded thereby, e.g., APC, GSK3, Axin1, Axin2, CK1α, ZNRF3, or RNF43. In some embodiments, a cancer comprises one or more mutations of APC. In some embodiments, a cancer comprises one or more mutations of a p53/MDM2/MDMX pathway. In some embodiments, a cancer comprises one or more mutations of p53. [000172] In some embodiments, a cancer is a Wnt-mediated cancer. In some embodiments, a cancer is an APC-mediated cancer. In some embodiments, a cancer is selected from colorectal cancer (CRC) (e.g., APC colorectal cancer), small intestine (small bowel) cancer, thyroid cancer, brain cancer, pancreatic cancer, bile duct cancer, and hepatoblastoma. [000173] In some embodiments, a cancer is a p53-mediated cancer. In some embodiments, a cancer is a leukemia or a lymphoma. In some embodiments, a cancer is selected from primary effusion lymphoma (PEL), myelodysplastic syndrome (MDS), or acute myeloid lymphoma (AML). [000174] In some embodiments, the present disclosure also provides a method of treating a cancer, the method comprising administering a compound of formula I, or a pharmaceutically acceptable salt thereof, to a subject in need thereof. [000175] In some embodiments, a subject is an animal. In some embodiments, a subject is a mammal. In some embodiments, a subject is a mouse. In some embodiments, a subject is a rat. In some embodiments, a subject is a pig. In some embodiments, a subject is a dog. In some embodiments, a subject is a non-human primate, e.g., a monkey. In some embodiments, a subject is a human. In some embodiments, a subject is an adult, e.g., an adult human. In some embodiments, a subject is a child, e.g., a human child. In some embodiments, a subject is susceptible to or suffering from a condition, disorder, or disease described herein. In some
embodiments, a subject is susceptible to or suffering from a cancer, e.g., a cancer described herein. [000176] In some embodiments, a provided compound is a compound of Formula I. In some embodiments, a provided compound is a compound of Table 1 or Table 2. In some embodiments, a provided compound is a compound of Table 1. In some embodiments, a compound described herein is a compound of Table 2. In some embodiments, a compound described herein is a compound of Formula I. In some embodiments, a compound described herein is a compound of Table 1 or Table 2. In some embodiments, a compound described herein is a compound of Table 1. In some embodiments, a compound described herein is a compound of Table 2. [000177] Compounds and compositions described herein may be administered or delivered to a subject by any suitable route known in the art. For example, in some embodiments, a compound or composition described herein is administered by an auricular, buccal, conjunctival, cutaneous, endosinusial, endotracheal, enteral, epidural, inhalational, intraabdominal, intraarterial, intraarticular, intrabiliary, intrabronchial, intrabursal, intracapsular, intracardiac, intracaudal, intracerebral, intracervical, intracisternal, intracorneal, intracoronary, intradermal, intradiscal, intraductal, intraduodenal, intradural, intraepidermal, intraesophageal, intragastric, intragingival, intraileal, intraluminal, intralymphatic, intramedullary, intrameningeal, intramuscular, intraocular, intraorbital, intraosseous, intraovarian, intraparenchymal, intrapericardial, intraperitoneal, intrapleural, intraspinal, intrasternal, intratesticular, intrathecal, intrathoracic, intratubular, intratumor, intratympanic, intrauterine, intravascular, intravenous, intraventricular, intravesical, intravitreal, laryngeal, nasal, ophthalmic, oral, oropharyngeal, parenteral, percutaneous, periarticular, peridural, peripheral, rectal, subarachnoid, subcapsular, subconjunctival, subcutaneous, subcuticular, sublingual, submucosal, topical, transdermal, transmucosal, transnasal, transtracheal, transtympanic, urethral, or vaginal route, or any combination thereof. [000178] In some embodiments, provided compounds or compositions thereof (e.g., pharmaceutical compositions thereof) may also be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents, and catheters. Implantable devices coated with a compound of this disclosure are another embodiment of the present disclosure.
7. Combination Therapies [000179] In some embodiments, the present disclosure provides technologies (e.g., compositions, methods) for combination therapy, for example with other therapeutic agents and/or medical procedures. In some embodiments, a subject is administered a provided compound or composition and one or more additional treatments (including, e.g., a therapeutic agent or method). [000180] In some embodiments, an additional treatment comprises or is surgery. In some embodiments, an additional treatment comprises or is radiation. [000181] In some embodiments, an additional treatment comprises or is one or more additional therapeutic agents. In some embodiments, an additional therapeutic agent comprises or is one or more cancer therapeutics described herein. In some embodiments, an additional therapeutic agent comprises or is an antibody or antigen binding fragment thereof. In some embodiments, an additional therapeutic agent comprises or is a cell therapy, e.g., a genetically modified cell therapy or an engineered cell therapy. In some embodiments, an additional therapeutic agent comprises or is a chemotherapeutic. In some embodiments, an additional therapeutic agent comprises or is a hormone. In some embodiments, an additional therapeutic agent comprises or is radiation. In some embodiments, an additional therapeutic agent comprises or is a vaccine. In some embodiments, an additional therapeutic agent comprises or is a nucleic acid. In some embodiments, an additional therapeutic agent comprises or is an oligonucleotide. In some embodiments, an additional therapeutic agent comprises or is a RNAi molecule, e.g., a miRNA, a shRNA, a siRNA. In some embodiments, an additional therapeutic agent comprises or is a lipid. In some embodiments, an additional therapeutic agent comprises or is a polypeptide or a portion thereof. In some embodiments, an additional therapeutic agent comprises or is a protein or a portion thereof. In some embodiments, an additional therapeutic agent comprises or is a lipid. [000182] In some embodiments, an additional treatment is capable of preventing, treating, ameliorating, or slowing the progression of a condition, disorder, or disease when administered alone. In some embodiments, an additional treatment is capable of preventing, treating, ameliorating, or slowing the progression of a cancer when administered alone. In some embodiments, an additional treatment is capable of preventing, treating, ameliorating, or slowing the progression of a condition, disorder, or disease when administered in combination with a
provided compound or composition. In some embodiments, an additional treatment is capable of preventing, treating, ameliorating, or slowing the progression of a cancer when administered in combination with a provided compound or composition. [000183] Various cancer therapeutics are known in the art and can be used with provided technologies (e.g., compounds, compositions, methods) described herein. In some embodiments, a cancer therapeutic comprises or is an antibody or antigen-binding fragment thereof. In some embodiments, cancer therapeutic comprises or is a chemotherapeutic agent. In some embodiments, a cancer therapeutic comprises or is a hormone. In some embodiments, a cancer therapeutic comprises or is radiation. In some embodiments, a cancer therapeutic comprises or is a vaccine. In some embodiments, a cancer therapeutic is a biologic agent. In some embodiments, a cancer therapeutic comprises or is a cellular therapeutic, e.g., a stem cell, an engineered cell, a genetically modified cell. [000184] In some embodiments, one or more cancer therapeutics are used in combination with a compound or composition described herein. In some embodiments, one or more cancer therapeutics are administered or delivered to a subject in combination with a compound or composition described herein. In some embodiments, one or more cancer therapeutics are administered or delivered to a subject prior to administering or delivering a compound or composition described herein to the subject. In some embodiments, one or more cancer therapeutics are administered or delivered to a subject at the same time as administering or delivering a compound or composition described herein to the subject. In some embodiments, one or more cancer therapeutics are administered or delivered to a subject following administering or delivering a compound or composition described herein to the subject. [000185] Various antibodies known in the art can be used with provided technologies (e.g., compounds, methods) described herein. In some embodiments, a cancer therapeutic comprises or is an antibody or an antigen-binding fragment thereof. In some embodiments, an antibody comprises or is a monoclonal antibody. In some embodiments, an antibody comprises or is a bispecific antibody. In some embodiments, an antibody comprises or is a conjugated antibody. In some embodiments, an antibody comprises or is an antibody-drug conjugate (ADC). In some embodiments, an antibody comprises or is a labeled antibody. In some embodiments, a labeled antibody comprises or is a radiolabeled antibody.
[000186] Various targets associated with cancer are known in the art and can be targeted with an antibody or an antigen-binding fragment thereof. In some embodiments, an antibody comprises or is an anti-4-1BB antibody, an anti-5’-nucleotidase antibody, an anti-5T4 antibody, an anti-ALK1 antibody, an anti-AFP antibody, an anti-angiopoietin-2 antibody, an anti-AXL antibody, an anti-BAFF antibody, an anti-BCMA antibody, an anti-c-Met antibody, an anti- CA125 antibody, an anti-CCR4 antibody, an anti-CCR5 antibody, an anti-CD3 antibody, an anti- CD3E antibody, an anti-CD4 antibody, an anti-CD19 antibody, an anti-CD20 antibody, an anti- CD22 antibody, an anti-CD23 antibody, an anti-CD25 antibody, an anti-CD27 antibody, an anti- CD28 antibody, an anti-CD30 antibody, an anti-CD33 antibody, an anti-CD37 antibody, an anti- CD38 antibody, an anti-CD40 antibody, an anti-CD44 antibody, an anti-CD51 antibody, an anti- CD56 antibody, an anti-CD70 antibody, an anti-CD74 antibody, an anti-CD79B antibody, an anti-CD80 antibody, an anti-CD123 antibody, an anti-CD134 antibody, an anti-CD152 antibody, an anti-CD200 antibody, an anti-CD221 antibody, an anti-CD276 antibody, an anti-CD279 antibody, an anti-CD319 antibody, an anti-CEA antibody, an anti-CEACAM5 antibody, an anti- CLDN18 antibody, an anti-CSF1 antibody, an anti-CSF1R antibody, an anti-CSF2 antibody, an anti-CTGF antibody, an anti-CTLA4 antibody, an anti-CXCR4 antibody, an anti-DLL3 antibody, an anti-DLL4 antibody, an anti-DR5 antibody, an anti-EGFL7 antibody, an anti-EGFR antibody, an anti-ENG antibody, an anti-EpCAM antibody, an anti-FAP antibody, an anti-FGFR2 antibody, an anti-FOLR1 antibody, an anti-GCPII antibody, an anti-GD2 ganglioside antibody, an anti-GPC3 antibody, an anti-GPNMB antibody, an anti-GPRC5D antibody, an anti-GUCY2C antibody, an anti-HER1 antibody, an anti-HER2 antibody, an anti-HER3 antibody, an anti- HGFR antibody, an anti-IGF-2 antibody, an anti-IL-1α antibody, an anti-IL-2 antibody, an anti- IL-6 antibody, an anti-IL-13 antibody, an anti-MIF antibody, an anti-MSLN antibody, an anti- MUC1 antibody, an anti-MUC5AC antibody, an anti-Notch-1 antibody, an anti-PCDC1 antibody, an anti-PCDP1 antibody, an anti-PCSK9 antibody, an anti-PD-1 antibody, an anti-PD- L1 antibody, an anti-PDGFRA antibody, an anti-PTK7 antibody, an anti-ROR1 antibody, an anti-SDC1 antibody, an anti-SLAMF7 antibody, an anti-SLITRK6 antibody, an anti-STEAP1 antibody, an anti-TAG-72 antibody, an anti-TEM antibody, an anti-TGFβ antibody, an anti- TIGIT antibody, an anti-TN-C antibody, an anti-TRAIL-R1 antibody, an anti-TRAIL-R2 antibody, an anti-TWEAKR antibody, an anti-TYRP1 antibody, an anti-VEGF-A antibody, an anti-VEGFR-1 antibody, an anti-VEGFR-2 antibody, or an anti-vimentin antibody.
[000187] Various antibodies that may be used as a cancer therapeutic are described in the art. For example, in some embodiments, an antibody comprises or is abagovomab, abituzumab, adalimumab, adecatumumab, alemtuzumab, alirocumab, amatuximab, amivantamab, anatumomab mafentaox, andecaliximab, anetumab ravtansine, apolizumab, arcitumomab, ascrinvacumab, atezolizumab, avelumab, bavituximab, belimumab, bemarituzumab, benralizumab, bermekimab, bevacizumab, bezlotoxumab, bivatuzumab, blinatumomab, brentuximab vedotin, brodalumab, brolucizumab, brontictuzumab, burosumab, cabiralizumab, camidanlumab tesirine, camrelizumab, canakinumab, cantuzumab mertansine, cantuzumab ravtansine, capromab, carlumab, carotuximab, catumaxomab, cemiplimab, certolizumab pegol, cetrelimab, cetuximab, cibisatamab, cirmtuzumab, citatuzumab bogatox, cixutumumab, clivatuzumab tetraxetan, codrituzumab, cofetuzumab pelidotin, coltuximab ravtansine, conatumumab, cusatuzumab, dancetuzumab, dalotuzumab, daratumumab, demcizumab, denintuzumab mafodotin, denosumab, depatuxizumab mafodotin, derlotuximab biotin, detumomab, dinutuximab, dinutuximab beta, dostarlimab, drozitumab, duligotuzumab, dupilumab, durvalumab, dusigitumab, duvortuxizumab, ecromeximab, edrecolomab, elgemtumab, elotuzumab, emactuzumab, emapalumab, emibetuzumab, emicizumab, enapotamab vedotin, enavatuzumab, enfortumab vedotin, enoblituzumab, ensituximab, epcoritamab, epratuzumab, erenumab, ertumaxomab, etaracizumab, evolocumab, figitumumab, flanvotumab, flotetuzumab, fremanezumab, fresolimumab, futuximab, galcanezumab, galiximab, gancotamab, ganitumab, gatipotuzumab, gemtuzumab ozogamicin, girentuximab, glembatumumab vedotin, glofitamab, golimumab, guselkumab, ibalizumab, ibritumomab tiuxetan, icrucumab, idarucizumab, igovomab, iladatuzumab vedotin, imalumab, imaprelimab, imgatuzumab, indatuximab ravtansine, indium altumomab pentetate, indusatumab vedotin, inebilizumab, infliximab, inolimomab, inotuzumab ozogamicin, intetumumab, iodine tositumomab, ipilimumab, iratumumab, isatuximab, istiratumab, ixekizumab, labetuzumab, lacnotuzumab, ladiratuzumab vedotin, lanadelumab, lenzilumab, leronlimab, lexatumumab, lifastuzumab vedotin, loncastuximab tesirine, losatuxizumab vedotin, lilotomab satetraxetan, lintuzumab, lirilumab, lorvotuzumab mertansine, lucatumumb, lumretuzumab, mapatumumab, margetuximab, matuzumab, mepolizumab, metelimumab, milatuzumab, mirvetuximab soravtansine, mitumomab, modotuximab, mogamulizumab, monalizumab, mosunetuzumab, moxetumomab pasudodox, nacolomab tafenatox, naptumomab estafenatox, narnatumab,
natalizumab, navicixizumab, naxitamab, necitumumab, nesvacumab, nimotuzumab, nivolumab, nofetumomab merpentan, obiltoxaximab, obinutuzumab, ocaratuzumab, ocrelizumab, ofatumumab, olaratumab, oleclumab, omalizumab, omburtamab, onartuzumab, ontuxizumab, oportuzumab monatox, oregovomab, otlertuzumab, palivizumab, pamrevlumab, panitumumab, pankomab, parsatuzumab, pasotuxizumab, pateclizumab, patritumab, pembrolizumab, pemtumomab, pertuzumab, pidilizumab, pinatuzumab vedotin, pintumumab, polatuzumab vedotin, pritumumab, racotumomab, radretumab, ramucirumab, ranibizumab, ravulizumab, relatlimab, reslizumab, retifanlimab, rilotumumab, risankizumab, rituximab, robatumumab, romosozumab, rosmantuzumab, rovalpituzumab tesirine, sacituzumab govitecan, samalizumab, sarilumab, satumomab pendetide, secukinumab, seribantumab, sibrotuzumab, siltuximab, sintilimab, sitratumab vedotin, sofituzumab vedotin, solitomab, spartalizumab, tabalumab, tacatuzumab tetraxetan, tafasitamab, talacotuzumab, talquetamab, taplitumomab paptox, tarextumab, tavolimab, teclistamab, telisotuzumab, telisotuzumab vedotin, tenatumomab, tepoditamab, tetulomab, tigatuzumab, tildrakizumab, timigutuzumab, tiragotumab, tislelizumab, tisotumab vedotin, tocilizumab, tomuzotuximab, tositumomab, tovetumab, trastuzumab, trastuzumab deruxtecan, trastuzumab duocarmazine, trastuzumab emtansine, tremelimumab, tucotuzumab celmoleukin, ublituximab, ulocuplumab, urelumab, ustekinumab, utomilumab, vadastuximab talirine, vandortuzumab vedotin, vantictumab, vanucizumab, varisacumab, varlilumab, vedolizumab, veltuzumab, vesencumab, volociximab, vonlerolizumab, vorsetuzumab mafodotin, votumumab, zalutumumab, zanolimumab, zatuximab, zenocutuzumab, or zolbetuximab, or an antigen-binding fragment thereof, or any combination thereof. [000188] In some embodiments, a cancer therapeutic comprises or is a chemotherapeutic agent. Various chemotherapeutic agents are known in the art and can be used with provided technologies (e.g., compounds, compositions, methods) described herein. For example, in some embodiments, compounds of the present disclosure, or a pharmaceutically acceptable composition thereof, are administered in combination with chemotherapeutic agents to treat proliferative diseases and cancer. [000189] In some embodiments, a chemotherapeutic agent comprises or is an alkylating agent. In some embodiments, a chemotherapeutic agent comprises or is an antimetabolite. In some embodiments, a chemotherapeutic agent comprises or is a cytidine analog. In some embodiments, a chemotherapeutic agent comprises or is an antifolate. In some embodiments, a
chemotherapeutic agent comprises or is a nucleoside analogue. In some embodiments, a chemotherapeutic agent comprises or is a purine analog. In some embodiments, a chemotherapeutic agent comprises or is a pyrimidine agent. In some embodiments, a chemotherapeutic agent comprises or is an antimicrotubular agent. In some embodiments, a chemotherapeutic agent comprises or is topoisomerase inhibitor, e.g., a topoisomerase I inhibitor, a topoisomerase II inhibitor. In some embodiments, a chemotherapeutic agent comprises or is an anthracycline. In some embodiments, a chemotherapeutic agent comprises or is a taxane. In some embodiments, a chemotherapeutic agent comprises or is an antibiotic. In some embodiments, a chemotherapeutic agent comprises or is a proteasome inhibitor. In some embodiments, a chemotherapeutic agent comprises or is a platinum-containing compound. In some embodiments, a chemotherapeutic agent comprises or is a kinase inhibitor. In some embodiments, a chemotherapeutic agent comprises or is a tyrosine kinase inhibitor. In some embodiments, a Bcr-Abl kinase inhibitor. In some embodiments, a chemotherapeutic agent comprises or is an EGFR inhibitor. In some embodiments, a chemotherapeutic agent comprises or is a HER2 inhibitor. In some embodiments, a chemotherapeutic agent comprises or is a JAK inhibitor. In some embodiments, a chemotherapeutic agent comprises or is a mTOR inhibitor. In some embodiments, a chemotherapeutic agent comprises or is a PDGFR inhibitor. In some embodiments, a chemotherapeutic agent comprises or is a VEGFR inhibitor. In some embodiments, a chemotherapeutic agent comprises or is a retinoid. In some embodiments, a chemotherapeutic agent comprises or is a histone deacetylase inhibitor. In some embodiments, a chemotherapeutic agent comprises or is a DNA replication inhibitor. In some embodiments, a chemotherapeutic agent comprises or is a DNA synthesis inhibitor. In some embodiments, a chemotherapeutic agent comprises or is an RNA synthesis inhibitor. In some embodiments, a chemotherapeutic agent comprises or is a cytoskeletal disruptor. In some embodiments, a chemotherapeutic agent comprises or is a proteasome inhibitor. In some embodiments, a chemotherapeutic agent comprises or is a plant alkaloid. In some embodiments, a chemotherapeutic agent comprises or is a cell cycle disruptor. [000190] Various chemotherapeutic agents that may be used as a cancer therapeutic are described in the art. For example, in some embodiments, a chemotherapeutic agent is 5- fluorouracil, aclarubicin, actinomycin D, afatinib, aflibercept, alitretinoin, altretamine, anagrelide, arsenic trioxide, asparaginase, axitinib, azacitidine, belotecan, bendamustine,
bexarotene, bleomycin, bortezomib, bosutinib, busulfan, cabazitaxel, camptothecin, capecitabine, carboplatin, carmofur, carmustine, chlorambucil, chlormethine, cisplatin, cladribine, clofarabine, crizotinib, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, dasatinib, daunomycin, daunorubicin, decitabine, diaziquone, docetaxel, doxorubicin, entionstat, epirubicin, erlotinib, estramustine, etoposide, everolimus, exatecan, floxuridine, fludarabine, fluorouracil, folinic acid, fotemustine, gefitinib, gemcitabine, hexamethylmelamine, hydroxycarbamide, hydroxyurea, idarubicin, ifosfamide, imatinib, irinotecan, isotretinoin, ixabepilone, lapatinib, larotaxel, lenalidomide, lomustine, mechlorethamine, melphalan, merbarone, mercaptopurine, methotrexate, mitomycin, mitomycin C, mitoxantrone, mitozolomide, mustine, nedaplatin, nelarabine, nilotinib, N-nitroso-N-methylurea, novobiocin, oxalipatin, paclitaxel (e.g., solvent- based paclitaxel, protein-bound paclitaxel, nanoparticle albumin-bound paclitaxel), panobinostat, pazopanib, pemetrexed, pentostatin, pirarubicin, pomalidomide, ponatinib, prednisolone, prednisone, procarbazine, raltitrexed, regorafenib, romidepsin, ruxolitinib, semustine, sorafenib, streptozotocin, sunitinib, tamibarotene, tegafur, temozolomide, temsirolimus, teniposide, tesetaxel, thalidomide, thioguanine, thiotepa, topotecan, tretinoin, valproate, valrubicin, vandetanib, vemurafenib, vinblastine, vincristine, vindesine, vinflunine, vinorelbine, vismodegib, vorinostat, or zabadinostat, or a derivative thereof, or any combination thereof. [000191] Other compounds that may be effective in treating cancer, that are suitable for use with the provided technologies (e.g., compounds, compositions, methods) of the present disclosure, are known in the art and are described, for example, in Anand U., et al. Gene Dis. 2022 Mar 18;10(4):1367-1401; Huang C., et al. Biomedicine (Taipei).2017 Dec;7(4):23; Amjad M. T., et al., StatsPearls, 2023 Feb 27. [000192] In some embodiments, a provided compound and an additional therapeutic agent are administered or delivered in the same composition. In some embodiments, a provided compound and an additional therapeutic are administered or delivered in separate compositions. [000193] In some embodiments, an additional therapeutic agent may be part of a single dosage form, mixed together with a compound of this disclosure in a single composition. In some embodiments, an additional therapeutic agent may be administered separately from a provided compound or composition thereof, as part of a multiple dosage regimen. If administered as part of a multiple dosage regime, a provided compound or composition thereof and an additional therapeutic agent may be administered or delivered simultaneously or sequentially. For example,
in some embodiments, a provided compound may be administered simultaneously with the additional therapeutic agent. In some embodiments, a provided compound may be administered prior to an additional therapeutic agent (e.g., an inhibitor of a particular cancer pathway). In some embodiments, a provided compound is administered four, six, eight, or twelve hours prior to the additional therapeutic agent. In some embodiments, a provided compound is administered one, two, three, four, five, six, or seven days prior to the additional therapeutic agent. In some embodiments, a provided compound is administered one, two, three, or four weeks prior to the additional therapeutic agent. In some embodiments, a provided compound and an additional therapeutic agent are administered in a multi-dose regimen such as A→(B→A→)nB, wherein A is a provided compound, B is the additional therapeutic agent, n is 0-20 or more, and “→” is the time period between each dose of A and B. [000194] Among other things, compounds of formula I and compositions thereof may be administered or delivered to a subject who previously was administered or delivered any of the additional therapeutic agents described herein and whose cancer may exhibit one or more signs of resistance to the previously administered additional therapeutic agent. In some embodiments, a subject comprises a cancer resistant to one or more additional therapeutic agents. In some embodiments, a subject comprises a cancer resistant to one or more cancer therapeutic agents. In some embodiments, a subject comprises a cancer resistant to one or more additional therapeutic agents which were previously administered or delivered to the subject. [000195] The amount of both, a compound of formula I and an additional therapeutic agent (in those compositions which comprise an additional therapeutic agent as described above) that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. Preferably, compositions of this disclosure should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of a provided compound can be administered. [000196] In some embodiments, in a composition which comprises a provided compound and an additional therapeutic agent, a provided compound and an additional therapeutic agent may act synergistically. Therefore, for example, an amount of additional therapeutic agent in such compositions will be less than the amount required in a monotherapy utilizing only the additional therapeutic agent. In some embodiments, in such compositions a dosage of between 0.01 – 100 mg/kg body weight/day of an additional therapeutic agent may be administered.
[000197] An amount of additional therapeutic agent present in a composition of this disclosure will be no more than an amount that would normally be administered in a composition comprising that additional therapeutic agent as the only active agent. Preferably, an amount of an additional therapeutic agent in a presently disclosed composition will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent. EXAMPLES [000198] Example 1. Synthesis of Exemplary Compounds [000199] General synthetic schemes for Compounds of Formula I:
[000200] Example 1.1. Synthesis of N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5- yl]-6-methoxynaphthalene-2-carboxamide (I-15)
[000201] Into a 25 mL round-bottom flask were added 3-(5-amino-1-oxo-3H-isoindol-2-yl) piperidine-2,6-dione (30 mg, 0.116 mmol, 1 equiv), DMF (3.00 mL), 6-methoxynaphthalene-2- carboxylic acid (25.74 mg, 0.128 mmol, 1.1 equiv), DIEA (29.91 mg, 0.232 mmol, 2 equiv) and HATU (131.99 mg, 0.348 mmol, 3 equiv) at room temperature. The resulting mixture was stirred
for overnight at 65°C under nitrogen atmosphere. Desired product could be detected by LCMS. The reaction was quenched with water at room temperature. The resulting mixture was extracted with EA (2 x 10 mL). The combined organic layers were washed with brine (2x10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in Water (0.1% FA), 0% to 100% gradient in 30 min; detector, UV 254 nm. This resulted in N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5- yl]-6-methoxynaphthalene-2-carboxamide (35 mg, 68.00%) as a white solid. [000202] LC-MS: (ES, m/z): [M+H]+ : 444.10. [000203] 1H NMR (300 MHz, DMSO-d6) δ 11.00 (s, 1H), 10.66 (s, 1H), 8.55 (d, J = 1.7 Hz, 1H), 8.19 (d, J = 1.7 Hz, 1H), 8.07 – 7.84 (m, 4H), 7.73 (d, J = 8.3 Hz, 1H), 7.44 (d, J = 2.5 Hz, 1H), 7.28 (dd, J = 9.0, 2.5 Hz, 1H), 5.12 (dd, J = 13.2, 5.0 Hz, 1H), 4.57 – 4.24 (m, 2H), 3.93 (s, 3H), 2.93 (ddd, J = 17.8, 13.4, 5.3 Hz, 1H), 2.77 – 2.57 (m, 1H), 2.38 (td, J = 13.2, 4.5 Hz, 1H), 2.08 – 1.95 (m, 1H). [000204] Example 1.2. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-3- hydroxy-1H-indazole-5-carboxamide (I-24)
[000205] Synthesis of 1-(tert-butoxycarbonyl)-3-hydroxyindazole-5-carboxylic acid
[000206] To a solution of 3-hydroxy-1H-indazole-5-carboxylic acid (200 mg, 1.123 mmol, 1 equiv) in DMF (10 mL) In a 25mL round-bottom flask was added sodium hydride (53.88 mg, 1.348 mmol, 1.2 equiv, 60% in oil) at 0°C. The mixture was stirred for 15 min. Boc2O (367.53 mg, 1.684 mmol, 1.5 equiv) was added and the mixture was allowed to warm to room temperature and stirred for 1 overnight. The reaction was quenched with ice water(50 mL) at 0°C. The resulting mixture was extracted with EA (3x20 mL). The water phase was
neutralized after pH 7 with 1 mol/L hydrochloric acid aqueous solution. The water phase was lyophilized. This resulted in 1-(tert-butoxycarbonyl)-3-hydroxyindazole-5-carboxylic acid (524 mg, crude) as a solid. [000207] LC-MS: (ES, m/z): [M-H]-: 277.0 [000208] The synthesis of Compound I-24 was completed following the procedure in Example 1.1 using 1-(tert-butoxycarbonyl)-3-hydroxyindazole-5-carboxylic acid and Boc removal. [000209] LC-MS: (ES, m/z): [M+H]+: 418.10 [000210] 1H NMR (300 MHz, DMSO-d6) δ 11.01 (s, 1H), 10.45 (s, 1H), 8.44 (d, J = 1.6 Hz, 1H), 8.29 (d, J = 2.0 Hz, 1H), 7.99 – 7.93 (m, 2H), 7.58 (d, J = 8.3 Hz, 1H), 7.39 (d, J = 8.8 Hz, 1H), 5.13 (dd, J = 13.2, 5.0 Hz, 1H), 4.44 (d, J = 17.1 Hz, 1H), 4.31 (d, J = 17.0 Hz, 1H), 3.00 – 2.83 (m, 1H), 2.61 (d, J = 16.8 Hz, 1H), 2.47 – 2.37 (m, 1H) 2.08 – 1.95 (m, 1H). [000211] Example 1.3. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-7- methoxy-1,5-naphthyridine-3-carboxamide (I-7)
[000212] Synthesis of diethyl 2-(((5-methoxypyridin-3-yl)amino)methylene)malonate
[000213] Into a 100 mL round-bottom flask were added 5-methoxypyridin-3-amine (2.0 g, 16.110 mmol, 1 equiv), 1,3-diethyl 2-(ethoxymethylidene) propanedioate (3.5 g, 16.110 mmol, 1.0 equiv) and EtOH (60 mL) at room temperature. The resulting mixture was stirred for overnight at 85°C under air atmosphere. The resulting mixture was concentrated under vacuum. This resulted in 4.5 g crude product as a brown oil. The crude product was used in the next step directly without further purification. [000214] LC-MS: (ES,m/z):[M+H]+: 295.10 [000215] Synthesis of ethyl 4-chloro-7-methoxy-1,5-naphthyridine-3-carboxylate)
[000216] Into a 20 mL pressure tank reactor were added 1,3-diethyl 2-{[(5-methoxypyridin-3- yl) amino] methylidene}propanedioate (1.5 g, 5.097 mmol, 1 equiv) and POCl3 (15.68 g, 101.940 mmol, 20 equiv) at room temperature. The final reaction mixture was irradiated with microwave radiation for 1.5 h at 160°C. The resulting mixture was concentrated under vacuum. The reaction was quenched with water at 0°C. The mixture was basified to pH 8 with saturated Na2CO3 (aq.). The resulting mixture was extracted with EA (3 x 40 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (4:1) to afford ethyl 4-chloro-7-methoxy-1,5-naphthyridine-3-carboxylate) (350 mg, 30.5%) as a light yellow solid. [000217] LC-MS: (ES,m/z): [M+H]+:267.05 [000218] Synthesis of ethyl 7-methoxy-1,5-naphthyridine-3-carboxylate
[000219] Into a 25 mL round-bottom flask were added ethyl 4-chloro-7-methoxy-1,5- naphthyridine-3-carboxylate (140 mg, 0.525 mmol, 1.0 equiv), ACN (5.0 mL), triethylsilane (122.09 mg, 1.050 mmol, 2.0 equiv) and bis(triphenylphosphine)palladium(II) chloride (73.70 mg, 0.105 mmol, 0.2 equiv) at rt. The resulting mixture was stirred for 3h at 70°C. The reaction was monitored by LCMS. The reaction was quenched with water at room temperature. The resulting mixture was extracted with chloromethane (3 x 10mL). The combined organic layers were washed with brine (2x10 mL), dried over anhydrous MgSO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (3:1) to afford ethyl 7-methoxy-1,5-naphthyridine-3- carboxylate (50 mg, 41.01%) as a white solid. [000220] LC-MS: (ES,m/z):[M+H]+:233.10
[000221] 1H NMR (400 MHz, Chloroform-d) δ 9.45 (d, J = 2.1 Hz, 1H), 8.97 (d, J = 2.0 Hz, 1H), 8.80 (d, J = 2.8 Hz, 1H), 7.68 (d, J = 2.9 Hz, 1H), 4.49 (q, J = 7.1 Hz, 2H), 4.02 (s, 3H), 1.46 (t, J = 7.1 Hz, 3H). [000222] Synthesis of 7-methoxy-1,5-naphthyridine-3-carboxylic acid
[000223] Into a 25 mL round-bottom flask were added ethyl 7-methoxy-1,5-naphthyridine-3- carboxylate (50 mg, 0.215 mmol, 1.0 equiv), THF (3 mL) and H2O (1 mL) at room temperature. Then LiOH (10.31 mg, 0.430 mmol, 2.0 equiv) was added to the above mixture. The resulting mixture was stirred for 2h at room temperature. The reaction was monitored by LCMS. The mixture was acidified to pH 5 with 4M. HCl aqueous. The resulting mixture was concentrated under reduced pressure. This resulted in 7-methoxy-1,5-naphthyridine-3-carboxylic acid (75 mg, crude) as a white solid. The crude product was used directly in the next step. [000224] LC-MS: (ES,m/z):[M+H]+:205.10 [000225] The synthesis of Compound I-7 was completed following the procedure in Example 1.1 using 7-methoxy-1,5-naphthyridine-3-carboxylic acid. [000226] LC-MS: (ES,m/z):[M+H]+: 446.10 [000227] 1H NMR (300 MHz, DMSO-d6) δ 11.00 (s, 1H), 10.93 (s, 1H), 9.41 (d, J = 2.2 Hz, 1H), 9.00 (d, J = 2.2 Hz, 1H), 8.89 (d, J = 2.8 Hz, 1H), 8.20 (d, J = 1.6 Hz, 1H), 7.90 (q, J = 3.2 Hz, 2H), 7.76 (d, J = 8.3 Hz, 1H), 5.12 (dd, J = 13.2, 5.0 Hz, 1H), 4.51 (d, J = 17.3 Hz, 1H), 4.36 (d, J = 17.3 Hz, 1H), 4.04 (s, 3H), 2.96 – 2.84 (m, 1H), 2.61 (d, J = 16.6 Hz, 1H), 2.47 – 2.32 (m, 1H), 2.08 – 1.97 (m, 1H). [000228] Example 1.4. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-7- methoxy-2,6-naphthyridine-3-carboxamide (I-8)
[000229] Synthesis of 5-bromo-4-(dimethoxymethyl)-2-methoxypyridine
[000230] Into a 50 mL round-bottom flask was charged with 5-bromo-2-methoxypyridine-4- carbaldehyde (1 g, 4.629 mmol, 1 equiv) dissolved in MeOH (20 mL) under air. H2SO4 (0.57 mL, 10.694 mmol, 2.31 equiv) was added dropwise to the reaction. The reaction was stirred at room temperature for 4 h. Then, the reaction was detected by LCMS and m/z: 261 was detected. The mixture was basified to pH 7 with saturated Na2CO3 aqueous. The resulting mixture was extracted with EA (3 x 50 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. 5- Bromo-4-(dimethoxymethyl)-2-methoxypyridine (930 mg, 76.65%) was not purified and used for the next step. [000231] LC-MS: (ES, m/z): [M+H]+ 262.05 [000232] Synthesis of 4-(dimethoxymethyl)-6-methoxynicotinaldehyde
[000233] Into a 100 mL three necked round-bottom flask was charged with 5-bromo-4- (dimethoxymethyl)-2-methoxypyridine (3.1 g, 11.827 mmol, 1 equiv) dissolved in THF (50 mL) under Ar. The reaction was cooled to -60°C. n-BuLi (2.5 M in hexane, 5.6 mL, 14.192 mmol, 1.2 equiv) was added dropwise to the reaction. The reaction was stirred at -60°C for 0.5 h. Then, dimethylformamide (2.59 g, 35.481 mmol, 3 equiv) was added dropwise to the reaction. The reaction was stirred at -60 °C for 15 min. Then, the reaction was stirred at -30 °C for 15 min. The reaction was detected by LCMS and desired product was obtained. The reaction was quenched by the addition of saturated ammonium chloride aqueous (50 mL) at -30 °C. The resulting mixture was extracted with EA (3 x 100 mL). The combined organic layers were washed with saturated NaCl aqueous (3 x 50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue (2.0 g, 60.85%) was used for the next step without further purification. [000234] LC-MS: (ES, m/z): [M+H]+ 212.15
[000235] Synthesis of methyl (2E)-3-[4-(dimethoxymethyl)-6-methoxypyridin-3-yl]-2- acetamidoprop-2-enoate
[000236] Into a 50 mL round bottom flask was charged with methyl 2-(dimethoxyphosphoryl)- 2-acetamidoacetate (1.36 g, 5.681 mmol, 1.2 equiv) dissolved in DCM (20 mL). DBU (1.06 mL, 7.095 mmol, 1.50 equiv) was added dropwise to the reaction. Then, 4-(dimethoxymethyl)-6- methoxypyridine-3-carbaldehyde (1 g, 4.734 mmol, 1 equiv) dissolved in DCM was added dropwise to the reaction. The reaction was stirred at room temperature for 3 h. The reaction was detected by LCMS and desired product was detected. The reaction was quenched by the addition of aqueous Na2CO3 (20 mL) at room temperature. The resulting mixture was extracted with DCM (3 x 20 mL). The combined organic layers were washed with brine (3 x 20mL), dried over anhydrous MgSO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (3/1) to afford methyl (2E)-3-[4-(dimethoxymethyl)-6-methoxypyridin-3-yl]-2-acetamidoprop-2-enoate (1.4 g, 91.17%) as a white solid. [000237] LC-MS: (ES, m/z): [M+H]+ 325.15 [000238] Synthesis of methyl 7-methoxy-2,6-naphthyridine-3-carboxylate
[000239] Into a 50 mL round-bottom flask was charged with methyl (2E)-3-[4- (dimethoxymethyl)-6-methoxypyridin-3-yl]-2-acetamidoprop-2-enoate (700 mg, 2.158 mmol, 1 equiv) dissolved in toluene (20 mL) under air. TsOH (297.32 mg, 1.726 mmol, 0.8 equiv) was added to the reaction. The reaction was stirred at 110°C for 12 h. The reaction was detected by LCMS and desired product was obtained. The solvent was removed under reduced pressure. The
residue was quenched by the addition of aqueous NaHCO3 (20 mL). The resulting mixture was extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (3x20mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. Methyl 7-methoxy-2,6-naphthyridine-3-carboxylate (350 mg, 74.32%) was not purified and used for the next step. [000240] LC-MS: (ES, m/z): [M+H]+ 219.10 [000241] Synthesis of 7-Methoxy-2,6-naphthyridine-3-carboxylic acid
[000242] Into a 50 mL round-bottom flask was charged with methyl 7-methoxy-2,6- naphthyridine-3-carboxylate (300 mg, 1.604 mmol, 1 equiv) dissolved in THF (30 mL) under air. NaOH (320.76 mg, 8.020 mmol, 5 equiv) dissolved in H2O (8 mL) was added dropwise to the reaction. The reaction was stirred at room temperature for 2 h. The reaction was detected by LCMS and major product was detected. The solvent was removed under reduced pressure and was added water (10mL) and was extracted with EA (2 x 20 mL). The aqueous layer was acidified by the addition of aqueous HCl (5 mL, 1M). The resulting mixture was extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure.7- Methoxy-2,6-naphthyridine-3-carboxylic acid (150 mg, 45.8%) was not purified and used for the next step. [000243] LC-MS: (ES, m/z): [M+H]+ 205.10 [000244] The synthesis of Compound I-8 was completed following the procedure in Example 1.1 using 7-methoxy-2,6-naphthyridine-3-carboxylic acid. [000245] LC-MS: (ES, m/z): [M+H]+ 446.15 [000246] 1H NMR (400 MHz, DMSO-d6) δ 11.01 (d, J = 12.4 Hz, 1H), 9.49 (s, 2H), 8.82 (s, 1H), 8.32 (s, 2H), 8.07-8.05 (m, 1H), 7.73 (d, J = 8.0 Hz, 1H), 7.58 (s, 1H), 5.12 (d, J = 9.2 Hz, 1H), 4.52-4.33 (m, 2H), 4.05 (s, 3H), 2.67-2.63 (m, 1H), 2.50-2.33 (m, 2H), 2.03-1.89 (m, 1H). [000247] Example 1.5. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- [1,2,3]triazolo[1,5-a]pyridine-4-carboxamide (I-12)
[000248] The synthesis of Compound I-12 was completed following the procedure in Example 1.1 using [1,2,3]triazolo[1,5-a]pyridine-4-carboxylic acid. [000249] LC-MS: (ES, m/z): [M+1]+: 405.05 [000250] 1H NMR (400 MHz, DMSO-d6) δ 11.00 (s, 1H), 10.91 (s, 1H), 9.32 (dt, J = 7.0, 0.9 Hz, 1H), 8.46 (d, J = 1.0 Hz, 1H), 8.20 – 8.11 (m, 2H), 7.85 (dd, J = 8.3, 1.8 Hz, 1H), 7.76 (d, J = 8.3 Hz, 1H), 7.38 (t, J = 7.0 Hz, 1H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.50 (d, J = 17.3 Hz, 1H), 4.36 (d, J = 17.3 Hz, 1H), 2.95 - 2.90 (m, 1H), 2.68 – 2.50 (m, 1H), 2.43 – 2.37 (m, 1H), 2.09 – 1.98 (m, 1H). [000251] Example 1.6. Synthesis of 6-((2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5- yl)carbamoyl)-2-naphthoic acid (I-28)
[000252] The synthesis of Compound I-28 was completed following the procedure in Example 1.1 using naphthalene-2,6-dicarboxylic acid. [000253] LC-MS: (ES, m/z): [M+H]+: 458.00 [000254] 1H NMR (400 MHz, DMSO-d6) δ 13.29 (s, 1H), 10.99 (s, 1H), 10.83 (s, 1H), 8.68 (d, J = 11.2 Hz, 2H), 8.33 – 8.05 (m, 5H), 7.89 (dd, J = 8.3, 1.9 Hz, 1H), 7.75 (d, J = 8.3 Hz, 1H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.49 (d, J = 17.2 Hz, 1H), 4.34 (d, J = 17.1 Hz, 1H), 2.95 - 2.90 (m, 1H), 2.68 – 2.50 (m, 1H), 2.43 – 2.37 (m, 1H), 2.09 – 1.98 (m, 1H). [000255] Example 1.7. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-6- fluoroquinoline-2-carboxamide (I-3)
[000256] The synthesis of Compound I-3was completed following the procedure in Example 1.1 using 6-fluoroquinoline-2-carboxylic acid. [000257] LC-MS: (ES, m/z): [M+H]+: 433.15 [000258] 1H NMR (400 MHz, DMSO-d6) δ 11.05 (s, 2H), 8.64 (d, J = 8.6 Hz, 1H), 8.39 – 8.25 (m, 3H), 8.05 (dd, J = 8.3, 1.9 Hz, 1H), 7.97 (dd, J = 9.4, 2.9 Hz, 1H), 7.87 (d, J = 8.9, 3.0 Hz, 1H), 7.77 (d, J = 8.3 Hz, 1H), 5.13 (dd, J = 13.3, 5.1 Hz, 1H), 4.51 (d, J = 17.2 Hz, 1H), 4.37 (d, J = 17.2 Hz, 1H), 2.95 - 2.90 (m, 1H), 2.68 – 2.50 (m, 1H), 2.43 – 2.37 (m, 1H), 2.09 – 1.98 (m, 1H). [000259] Example 1.8. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-6- hydroxyquinoline-2-carboxamide (I-10)
[000260] The synthesis of Compound I-10 was completed following the procedure in Example 1.1 using 6-hydroxyquinoline-2-carboxylic acid. [000261] LC-MS: (ES, m/z): [M+H]+ 431.05 [000262] 1H NMR (400 MHz, DMSO-d6) δ 11.00 (s, 1H), 10.94 (s, 1H), 10.49 (s, 1H), 8.60 - 8.40 (d, J = 8.6 Hz, 1H), 8.29 (d, J = 1.8 Hz, 1H), 8.14 - 8.13 (m, 2H), 8.11- 8.03(m, 1H), 7.75 (d, J = 8.3 Hz, 1H), 7.50 - 7.48 (m, 1H), 7.27 (d, J = 2.7 Hz, 1H), 5.20 - 5.10 (m, 1H), 4.50 (d, J = 17.2 Hz, 1H), 4.36 (d, J = 17.2 Hz, 1H), 2.94 - 2.92 (m, 1H), 2.62 - 2.59 (m, 1H), 2.42 - 2.39 (m, 1H), 2.04 - 2.02 (m, 1H). [000263] Example 1.9. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-4- hydroxy-7-methoxyquinoline-3-carboxamide (I-13)
[000264] The synthesis of Compound I-13 was completed following the procedure in Example 1.1 using 4-hydroxy-7-methoxyquinoline-3-carboxylic acid. [000265] LC-MS: (ES, m/z): [M+H]+: 461.10
[000266] 1H NMR (400 MHz, DMSO-d6) δ 12.96 (s, 1H), 12.95 - 12.60 (m, 1H), 10.98 (s, 1H), 8.84 (d, J = 3.2 Hz, 1H), 8.23 (d, J = 8.9 Hz, 1H), 8.08 (s, 1H), 7.77 (d, J = 8.1 Hz, 1H), 7.71 (d, J = 8.0 Hz, 1H), 7.14 (d, J = 7.8 Hz, 2H), 5.10 (d, J = 13.0 Hz, 1H), 4.47 (d, J = 16.8 Hz, 1H), 4.33 (d, J = 17.1 Hz, 1H), 3.91 (d, J = 3.1 Hz, 3H), 2.95 - 2.90 (m, 1H), 2.68 – 2.50 (m, 1H), 2.43 – 2.37 (m, 1H), 2.09 – 1.98 (m, 1H). [000267] Example 1.10. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-4)
[000268] The synthesis of Compound I-4 was completed following the procedure in Example 1.1 using 1-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid. [000269] LC-MS: (ES, m/z): [M+H]+: 418.00 [000270] 1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.62 (s, 1H), 8.89 (d, J = 2.2 Hz, 1H), 8.61 (d, J = 2.5 Hz, 1H), 8.17 (s, 1H), 7.86 (d, J = 8.5 Hz, 1H), 7.72 (d, J = 8.2 Hz, 1H), 7.67 (d, J = 3.5 Hz, 1H), 6.66 (d, J = 3.5 Hz, 1H), 5.11 (dd, J = 13.4, 5.0 Hz, 1H), 4.48 (d, J = 17.2 Hz, 1H), 4.34 (d, J = 17.1 Hz, 1H), 3.88 (d, J = 2.5 Hz, 3H), 2.95 - 2.90 (m, 1H), 2.68 – 2.50 (m, 1H), 2.43 – 2.37 (m, 1H), 2.09 – 1.98 (m, 1H). [000271] Example 1.11. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5- yl)thiazolo[5,4-b]pyridine-5-carboxamide (I-11)
[000272] The synthesis of Compound I-11 was completed following the procedure in Example 1.1 using thiazolo[5,4-b]pyridine-5-carboxylic acid. [000273] LC-MS: (ES, m/z): [M+H]+:421.95 [000274] 1H NMR (400 MHz, DMSO-d6) δ 11.13 (s, 1H), 11.01 (s, 1H), 9.78 (s, 1H), 8.73 (d, J = 8.5 Hz, 1H), 8.37 (d, J = 8.5 Hz, 1H), 8.29 (s, 1H), 8.04 (dd, J = 8.4, 1.8 Hz, 1H), 7.75 (d, J =
8.3 Hz, 1H), 5.11 (dd, J = 13.3, 5.2 Hz, 1H), 4.49 (d, J = 17.2 Hz, 1H), 4.34 (d, J = 17.1 Hz, 1H), 2.95 - 2.90 (m, 1H), 2.68 – 2.50 (m, 1H), 2.43 – 2.37 (m, 1H), 2.09 – 1.98 (m, 1H). [000275] Example 1.12. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1H-pyrazolo[3,4-b]pyridine-5-carboxamide (I-21)
[000276] The synthesis of Compound I-21 was completed following the procedure in Example 1.1 using 1H-pyrazolo[3,4-b]pyridine-5-carboxylic acid. [000277] LC-MS: (ES, m/z): [M+H]+: 405.05 [000278] 1H NMR (400 MHz, DMSO-d6) δ 13.98 (s, 1H), 11.00 (s, 1H), 10.75 (s, 1H), 9.10 (d, J = 2.1 Hz, 1H), 8.88 (d, J = 2.1 Hz, 1H), 8.37 (s, 1H), 8.17 (d, J = 1.9 Hz, 1H), 7.89 – 7.82 (m, 1H), 7.74 (d, J = 8.3 Hz, 1H), 5.17 – 5.08 (m, 1H), 4.49 (d, J = 17.3 Hz, 1H), 4.35
= 17.3 Hz, 1H), 2.99 – 2.86 (m, 1H), 2.66 – 2.57 (m, 1H), 2.48 – 2.33 (m, 1H), 2.06 – 1.98 (m, 1H). [000279] Example 1.13. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- [1,2,4]triazolo[4,3-b]pyridazine-6-carboxamide (I-30)
[000280] The synthesis of Compound I-30 was completed following the procedure in Example 1.1 using [1,2,4]triazolo[4,3-b]pyridazine-6-carboxylic acid. [000281] LC-MS: (ES, m/z): [M+H]+: 406.0 [000282] 1H NMR (400 MHz, DMSO-d6) δ 11.11 (d, J = 88.5 Hz, 2H), 9.82 (s, 1H), 8.57 (d, J = 9.7 Hz, 1H), 8.19 (d, J = 1.8 Hz, 1H), 7.94 (dd, J = 8.5, 1.9 Hz, 1H), 7.87 (d, J = 9.6 Hz, 1H), 7.76 (d, J = 8.3 Hz, 1H), 5.12 (dd, J = 13.2, 5.1 Hz, 1H), 4.50 (d, J = 17.3 Hz, 1H), 4.36 (d, J = 17.3 Hz, 1H), 2.99 - 2.80 (m, 1H), 2.66 – 2.57 (m, 1H), 2.50 – 2.30 (m, 1H), 2.10 – 1.98 (m, 1H). [000283] Example 1.14. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1,6-naphthyridine-2-carboxamide (I-14)
[000284] The synthesis of Compound I-14 was completed following the procedure in Example 1.1 using 1,6-naphthyridine-2-carboxylic acid. [000285] LC-MS: (ES, m/z): [M+H]+: 416.00 [000286] 1H NMR (400 MHz, DMSO-d6) δ 11.11 (s, 1H), 11.02 (s, 1H), 9.57 (s, 1H), 8.87 (dd, J = 14.8, 7.2 Hz, 2H), 8.46 – 8.36 (m, 2H), 8.18 – 8.09 (m, 2H), 7.64 (d, J = 8.2 Hz, 1H), 5.14 (dd, J = 13.2, 5.1 Hz, 1H), 4.47 (d, J = 17.2 Hz, 1H), 4.34 (d, J = 17.2 Hz, 1H), 3.00 – 2.96 (m, 1H), 2.66 – 2.57 (m, 1H), 2.48 – 2.35 (m, 1H), 2.09 – 1.99 (m, 1H). [000287] Example 1.15. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 2-methyl-3H-imidazo[4,5-b]pyridine-6-carboxamide (I-5)
[000288] The synthesis of Compound I-5 was completed following the procedure in Example 1.1 using 2-methyl-3H-imidazo[4,5-b]pyridine-6-carboxylic acid. [000289] LC-MS: (ES, m/z): [M+H]+: 419.15 [000290] 1H NMR (400 MHz, DMSO-d6) δ 13.02 (s, 1H), 10.99 (s, 1H), 10.65 (s, 1H), 8.86 (s, 1H), 8.46 (s, 1H), 8.17 (s, 1H), 7.85 (d, J = 2.0 Hz, 1H), 7.73 (d, J = 11.2 Hz, 1H), 5.14-5.08 (m, 1H), 4.49 (d, J = 17.2 Hz, 1H), 4.34 (d, J = 17.1 Hz, 1H), 2.95 - 2.90 (m, 1H), 2.68 – 2.50 (m, 1H), 2.43 – 2.37 (m, 1H), 2.09 – 1.98 (m, 1H). [000291] Example 1.16. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-methyl-1H-benzo[d]imidazole-5-carboxamide (I-29)
[000292] The synthesis of Compound I-29 was completed following the procedure in Example 1.1 using 1-methyl-1H-benzo[d]imidazole-5-carboxylic acid. [000293] LC-MS: (ES,m/z): [M+H]+: 418.00 [000294] 1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.54 (s, 1H), 8.42 (s, 1H), 8.35 (s, 1H), 8.20 (s, 1H), 7.95 (d, J = 8.6 Hz, 1H), 7.89 (d, J = 8.5 Hz, 1H), 7.72 (dd, J = 8.4, 4.5 Hz, 2H), 5.11 (dd, J = 13.3, 5.0 Hz, 1H), 4.48 (d, J = 17.3 Hz, 1H), 4.34 (d, J = 17.2 Hz, 1H), 3.90 (s, 3H), 2.95 - 2.90 (m, 1H), 2.68 – 2.50 (m, 1H), 2.43 – 2.37 (m, 1H), 2.09 – 1.98 (m, 1H). [000295] Example 1.17. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 3H-imidazo[4,5-c]pyridine-6-carboxamide (I-9)
[000296] The synthesis of Compound I-9 was completed following the procedure in Example 1.1 using 3H-imidazo[4,5-c]pyridine-6-carboxylic acid. [000297] LC-MS: (ES, m/z): [M+H]+: 405.05 [000298] 1H NMR (400 MHz, DMSO-d6) δ 11.00 (s, 2H), 9.08 (s, 1H), 8.60 (s, 1H), 8.41 (s, 1H), 8.31 (s, 1H), 8.03 (d, J = 8.4 Hz, 1H), 7.72 (d, J = 8.3 Hz, 1H), 5.12 (dd, J = 13.3, 5.0 Hz, 1H), 4.49 (d, J = 17.2 Hz, 1H), 4.34 (d, J = 17.1 Hz, 1H), 2.95 - 2.90 (m, 1H), 2.68 – 2.50 (m, 1H), 2.43 – 2.37 (m, 1H), 2.09 – 1.98 (m, 1H). [000299] Example 1.18. Synthesis of 1-cyclopropyl-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-1H-pyrazolo[3,4-d]pyrimidine-6-carboxamide (I-20)
[000300] The synthesis of Compound I-20 was completed following the procedure in Example 1.1 using 1-cyclopropyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxylic acid. [000301] LC-MS: (ES, m/z):[M+H]+ 446.05 [000302] 1H NMR (400 MHz, DMSO-d6) δ 11.15 (s, 1H), 11.00 (s, 1H), 9.52 (s, 1H), 8.49 (s, 1H), 8.27 (d, J = 1.7 Hz, 1H), 7.99 (dd, J = 8.4, 1.8 Hz, 1H), 7.76 (d, J = 8.3 Hz, 1H), 5.12 (dd, J
= 13.3, 5.1 Hz, 1H), 4.51 (d, J = 17.3 Hz, 1H), 4.36 (d, J = 17.2 Hz, 1H), 4.20 - 4.10 (m, 1H), 3.00 - 2.80 (m, 1H), 2.66 – 2.57 (m, 1H), 2.50 - 2.30 (m, 1H), 2.07 – 1.98 (m, 1H), 1.34 – 1.23 (m, 2H), 1.26 – 1.16 (m, 2H). [000303] Example 1.19. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-(2-hydroxyethyl)-2-methyl-1H-benzo[d]imidazole-5-carboxamide (I-32)
[000304] The synthesis of Compound I-32 was completed following the procedure in Example 1.1 using 1-(2-hydroxyethyl)-2-methyl-1H-benzo[d]imidazole-5-carboxylic acid. [000305] LC-MS: (ES, m/z): [M+H]+ 497.25 [000306] 1H NMR (300 MHz, DMSO-d6) δ 11.00 (s, 1H), 10.51 (s, 1H), 8.23 (d, J = 21.0 Hz, 2H), 7.91 – 7.85 (m, 2H), 7.72 – 7.62 (m, nH), 5.11 (dd, J = 13.7, 5.2 Hz, 1H), 4.56 – 4.16 (m, 4H), 3.75 –3.68 (m, 3H), 2.96 – 2.87 (m, 2H), 2.67 – 2.54 (m, 4H), 2.44 - 2.38 (m, 1H), 2.05 – 1.97 (m, 1H). [000307] Example 1.20. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 7-hydroxy-[1,2,4]triazolo[1,5-a]pyrimidine-6-carboxamide (I-19)
[000308] The synthesis of Compound I-19 was completed following the procedure in Example 1.1 using 7-hydroxy-[1,2,4]triazolo[1,5-a]pyrimidine-6-carboxylic acid. [000309] LC-MS: (ES, m/z):[M+H]+: 422.00 [000310] 1H NMR (400 MHz, DMSO-d6) δ 11.73 (s, 1H), 10.98 (s, 1H), 8.81 (s, 1H), 8.27 (s, 1H), 8.08 (s, 1H), 7.76 – 7.63 (m, 2H), 7.21 -6.95 (m, 1H), 5.10 (dd, J = 13.3, 5.1 Hz, 1H), 4.47 (d, J = 17.2 Hz, 1H), 4.33 (d, J = 17.2 Hz, 1H), 2.96 – 2.87 (m, 1H), 2.67 – 2.54 (m, 1H), 2.44 - 2.38 (m, 1H), 2.05 – 1.97 (m, 1H). [000311] Example 1.21. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1H-pyrazolo[4,3-b]pyridine-5-carboxamide (I-6)
[000312] The synthesis of Compound I-6 was completed following the procedure in Example 1.1 using 1H-pyrazolo[4,3-b]pyridine-5-carboxylic acid. [000313] LC-MS: (ES, m/z): [M-H]+: 403.00 [000314] 1H NMR (400 MHz, DMSO-d6) δ 13.73 (s, 1H), 11.01 (s, 2H), 8.53 (s, 1H), 8.29 – 8.20 (m, 3H), 8.04 (d, J = 8.5 Hz, 1H), 7.73 (d, J = 8.3 Hz, 1H), 5.14 – 5.10 (m, 1H), 4.45 – 4.32 (m, 2H), 2.96 – 2.90 (m, 1H), 2.63 – 2.50 (m, 1H), 2.43 – 2.35 (m, 1H), 2.07– 2.00 (m, 1H). [000315] Example 1.22. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5- yl)benzo[c][1,2,5]thiadiazole-5-carboxamide (I-31)
[000316] The synthesis of Compound I-31 was completed following the procedure in Example 1.1 using benzo[c][1,2,5]thiadiazole-5-carboxylic acid. [000317] LC-MS: (ES, m/z): [M+H]+: 421.95 [000318] 1H NMR (400 MHz, DMSO-d6) δ 10.95 (d, J = 31.6 Hz, 2H), 8.80 (s, 1H), 8.23 (dd, J = 18.8, 9.2 Hz, 3H), 7.89 (d, J = 8.4 Hz, 1H), 7.76 (d, J = 8.4 Hz, 1H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.62 – 4.10 (m, 2H), 2.95 – 2.83 (m, 1H), 2.70 – 2.55 (m, 1H), 2.50 – 2.40 (m, 1H), 2.10- 1.99 (m, 1H). [000319] Example 1.23. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 3-methylbenzamide (I-17)
[000320] The synthesis of Compound I-17 was completed following the procedure in Example 1.1 using 3-methylbenzoic acid.
[000321] LC-MS: (ES, m/z): [M+H]+= 378.1 [000322] 1H NMR (300 MHz, DMSO-d6) δ 11.00 (s, 1H), 10.54 (s, 1H), 8.15 (d, J = 1.7 Hz, 1H), 7.91 – 7.66 (m, 4H), 7.44 (dd, J = 4.1, 1.7 Hz, 2H), 5.11 (dd, J = 13.2, 5.1 Hz, 1H), 4.57 – 4.23 (m, 2H), 2.99 – 2.87 (m, 1H), 2.61 (ddd, J = 17.1, 4.4, 2.3 Hz, 1H), 2.41 (s, 3H), 2.38 – 2.32 (m, 1H), 2.04 – 1.98 (m, 1H). [000323] Example 1.24. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 8-methoxy-2H-chromene-3-carboxamide (I-16)
[000324] The synthesis of Compound 16 was completed following the procedure in Example 1.1 using 8-methoxy-2H-chromene-3-carboxylic acid. [000325] LC-MS: (ES, m/z): [M+H]+ 448.15 [000326] 1H NMR (300 MHz, DMSO-d6) δ 11.00 (s, 1H), 10.38 (s, 1H), 8.06 (d, J = 1.7 Hz, 1H), 7.85 – 7.65 (m, 2H), 7.53 (d, J = 1.4 Hz, 1H), 7.11 – 6.80 (m, 3H), 5.11(dd, J = 5.1 Hz, 17.6 Hz, 1H), 4.97 (d, 0.9 Hz, 1H), 4.55 – 4.23 (m, 2H), 3.79 (s, 3H), 3.02 – 2.83 (m, 1H), 2.62 – 2.50 (m, 1H), 2.45 – 2.37 (m, 1H), 2.08 – 1.90 (m, 1H). [000327] Example 1.25. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 6-methoxy-2-naphthamide (I-15)
[000328] The synthesis of Compound I-15 was completed following the procedure in Example 1.1 using 6-methoxy-2-naphthoic acid. [000329] LC-MS: (ES, m/z): [M+H]+: 444.10. [000330] 1H NMR (300 MHz, DMSO-d6) δ 11.00 (s, 1H), 10.66 (s, 1H), 8.55 (d, J = 1.7 Hz, 1H), 8.19 (d, J = 1.7 Hz, 1H), 8.07 – 7.84 (m, 4H), 7.73 (d, J = 8.3 Hz, 1H), 7.44 (d, J = 2.5 Hz,
1H), 7.28 (dd, J = 9.0, 2.5 Hz, 1H), 5.12 (dd, J = 13.2, 5.0 Hz, 1H), 4.57 – 4.24 (m, 2H), 3.93 (s, 3H), 2.93 (ddd, J = 17.8, 13.4, 5.3 Hz, 1H), 2.77 – 2.57 (m, 1H), 2.38 (td, J = 13.2, 4.5 Hz, 1H), 2.08 – 1.95 (m, 1H). [000331] Example 1.26. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 6-hydroxy-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-2)
[000332] Synthesis of N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-6-methoxy-1H- pyrrolo[2,3-b] pyridine-5-carboxamide
[000333] Into a 50 mL round-bottom flask were added 6-methoxy-1H-pyrrolo[2,3-b] pyridine- 5-carboxylic acid (250 mg, 1.301 mmol, 1.0 equiv), 3-(5-amino-1-oxo-3H-isoindol-2-yl) piperidine-2,6-dione (337.28 mg, 1.301 mmol, 1.0 equiv), DMF (5 mL), DIEA (672.55 mg, 5.204 mmol, 4 equiv) and HATU (989.30 mg, 2.602 mmol, 2.0 equiv) at room temperature. The resulting mixture was stirred for overnight at 60°C. The mixture was directly purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in Water, 0% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-6-methoxy-1H-pyrrolo[2,3-b] pyridine-5-carboxamide (150 mg, 26.60%) as a brown solid. [000334] LC-MS: (ES, m/z): [M+H]+: 434.05 [000335] 1H NMR (400 MHz, DMSO-d6) δ 11.82 (s, 1H), 11.01 (s, 1H), 10.33 (s, 1H), 8.42 (s, 1H), 8.28 (d, J = 2.0 Hz, 1H), 7.94 (d, J = 13.0 Hz, 1H), 7.59 (d, J = 8.2 Hz, 1H), 7.36 – 7.30 (m, 1H), 6.50 (dd, J = 3.6, 1.7 Hz, 1H), 5.13 (dd, J = 13.3, 5.1 Hz, 1H), 4.45 (d, J = 17.2 Hz, 1H),
4.32 (d, J = 17.1 Hz, 1H), 4.07 (s, 3H), 2.95 (dd, J = 13.3, 5.1 Hz, 1H), 2.61 (d, J = 17.3 Hz, 1H), 2.41 (tt, J = 13.3, 6.7 Hz, 1H), 2.05-1.99 (m, 1H). [000336] Synthesis of N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-6-hydroxy-1H- pyrrolo[2,3-b] pyridine-5-carboxamide
[000337] Into a 25 mL round-bottom flask were added N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo- 3H-isoindol-5-yl]-6-methoxy-1H-pyrrolo[2,3-b] pyridine-5-carboxamide (45 mg, 0.104 mmol, 1 equiv) and BBr3 (1mmol/ml in DCM, 2 mL) at room temperature. The resulting mixture was stirred for additional overnight at room temperature. The reaction was quenched with Water/Ice at room temperature. The resulting mixture was extracted with DCM/MeOH 5:1 (3 x 20mL). The combined organic layers were washed with brine (3x20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xselect CSH Prep C18 OBD, 30*150mm, 5um; Mobile Phase A: water (0.1% TFA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 20% B to 39% B in 8 min; Wave Length: 254nm/220nm nm; RT1(min): 7.43) to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-6-hydroxy- 1H-pyrrolo[2,3-b] pyridine-5-carboxamide (9.3 mg, 20.33%) as a white solid. [000338] LC-MS: (ES, m/z): [M+H]+: 420.0 [000339] 1H NMR (400 MHz, DMSO-d6) δ 12.76 (s, 1H), 11.45 (s, 1H), 11.00 (s, 1H), 8.72 (s, 1H), 8.29 (d, J = 1.9 Hz, 1H), 7.72 (d, J = 8.1 Hz, 1H), 7.58 (d, J = 8.0 Hz, 1H), 6.98 (s, 1H), 6.46 (d, J = 3.4 Hz, 1H), 5.13 (dd, J = 13.2, 5.1 Hz, 1H), 4.43 (d, J = 17.1 Hz, 1H), 4.30 (d, J = 17.1 Hz, 1H), 2.99 – 2.85 (m, 1H), 2.61 (d, J = 16.7 Hz, 1H), 2.02 (s, 1H), 1.23 (s, 1H). [000340] Example 1.27. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 7-methoxycinnoline-3-carboxamide (I-25)
[000341] Synthesis of ethyl (2E)-3-(piperidin-1-yl) prop-2-enoate
[000342] Into a 25 mL round-bottom flask was charged with ethyl propiolate (1.0 g, 10.194 mmol, 1.0 equiv) dissolved in ACN (10 mL). piperidine (0.87 g, 10.194 mmol, 1.0 equiv) was added dropwise to the reaction. The reaction was stirred at room temperature for 15 min. Then, the reaction was detected by TLC and one new spot was detected. The solvent was removed under reduced pressure. The product ethyl (2E)-3-(piperidin-1-yl) prop-2-enoate (1.5 g, crude) was obtained as a yellow oil. The crude product was not purified and directly used for the next step. [000343] LC-MS: (ES, m/z): [M+H]+ 184.15 [000344] Synthesis of 3-methoxybenzenediazonium tetrafluoroborate
[000345] Into a 25 mL round-bottom flask were added M-anisidine (2.0 g, 16.240 mmol, 1.0 equiv) at room temperature. Then, HCl aqueous (8 mL, 6M) was added to the reaction. The reaction was cooled to 0 °C and Sodium nitrite (1.34 g, 19.420 mmol, 1.2 equiv) dissolved in H2O (1 mL) was added dropwise at 0 °C. The resulting mixture was stirred for additional 30 min at 0 °C. To the above mixture was added saturated sodium fluoroborate. The precipitation solid was washed by Et2O for several times. The product (1.2 g, crude) was obtained as a brown solid. The crude product was not purified and directly used for the next step. [000346] LC-MS: (ES, m/z): [M+H]+ no exact MS signal [000347] Synthesis of ethyl 7-methoxycinnoline-3-carboxylate
[000348] Into a 25 mL round-bottom flask was charged with ethyl (2E)-3-(piperidin-1-yl) prop- 2-enoate (500 mg, 2.728 mmol, 1.0 equiv) dissolved in ACN (10 mL).3-
methoxybenzenediazonium (604 mg, 4.469 mmol, 1.64 equiv) was added in portions to the reaction. The reaction was stirred at 80 °C for 1 h. The reaction was deteceted by LCMS and desired product was detected. The solvent was removed under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford ethyl 7- methoxycinnoline-3-carboxylate (200 mg, 31.56%) as a red solid. [000349] LC-MS: (ES, m/z): [M+H]+ 233.10 [000350] Synthesis of 7-methoxycinnoline-3-carboxylic acid
[000351] Into a 25 mL round-bottom flask was charged with ethyl 7-methoxycinnoline-3- carboxylate (200 mg, 0.861 mmol, 1.0 equiv), THF (3 mL) and H2O (3 mL). The mixture was stirred for 2h. Then the mixture was adjusted to PH =2 with 1M HCl aqueous. Then the mixture was extracted by EA (3X15 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product 7-methoxycinnoline-3-carboxylic acid (170 mg, crude) was obtained and used for the next step without further purification. [000352] LC-MS: (ES, m/z): [M+H]+ 205.10 [000353] Synthesis of N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-7- methoxycinnoline-3-carboxamide
[000354] Into a 10 mL microwave vial was charged with 7-methoxycinnoline-3-carboxylic acid (150 mg, 0.735 mmol, 1 equiv) dissolved in ACN (2 mL). POCl3 (337.90 mg, 2.205 mmol, 3 equiv) was added dropwise to the reaction. The reaction was stirred at 80 °C for 3 h. The reaction was detected by LCMS and desired product was detected. The reaction was quenched by the addition of H2O (10 mL) at 0 °C. Then, the resulting mixture was extracted with EA (3 x 20 mL). The combined organic layers were washed with NaCl (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was
purified by Pre-HPLC (Column: Xselect CSH Prep C18 OBD, 30*150mm, 5um; Mobile Phase A: Water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 22% B to 27% B in 12 min; Wave Length: 254nm/220nm nm; RT1(min): 11.07). The desired product N- [2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-7-methoxycinnoline-3-carboxamide (9.1 mg, 2.67%) was obtained as an off-white solid. [000355] LC-MS: (ES, m/z): [M+H]+: 446.05 [000356] 1H NMR (400 MHz, DMSO-d6) δ 11.48 (s, 1H), 8.92 (s, 1H), 8.48-8.43 (m, 2H), 8.36-8.30 (m, 1H), 8.18 (dd, J = 8.3, 2.1 Hz, 1H), 7.94 (d, J = 2.4 Hz, 1H), 7.68 – 7.63 (m, 2H), 5.14 (dd, J = 13.1, 5.1 Hz, 1H), 4.62 – 4.24 (m, 2H), 4.08 (s, 3H), 3.00 – 2.79 (m, 1H), 2.73 – 2.58 (m, 1H), 2.55-2.50(m, 1H), 2.21 – 1.74 (m, 1H). [000357] Example 1.28. Synthesis of 3-(5-[(7-methoxyisoquinolin-3-yl) amino]-1-oxo-3H- isoindol-2-ylpiperidine-2,6-dione (I-59)
[000358] Into a 25mL round-bottom flask were added 3-(5-amino-1-oxo-3H-isoindol-2-yl) piperidine-2,6-dione (100 mg, 0.386 mmol, 1.0 equiv), (1,3-bis[2,6-bis(heptan-4-yl) phenyl]-4,5- dichloro-2,3-dihydro-1H-imidazol-2-yldichloro(3-chloro-1lambda4-pyridin-1-yl) palladium (37.56 mg, 0.039 mmol, 0.1 equiv), cesium carbonate (251.34 mg, 0.772 mmol, 2.0 equiv) and dioxane (2 mL) at room temperature. The resulting mixture was stirred for 16h at 110°C under nitrogen atmosphere. The reaction was quenched by the addition of sat. ammonium chloride (aq.) (10mL) at room temperature. The resulting mixture was extracted with ethyl acetate (3 x 10mL). The combined organic layers were dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with dichloromethane / methanol (5:1) to afford 3-(5-[(7- methoxyisoquinolin-3-yl) amino]-1-oxo-3H-isoindol-2-ylpiperidine-2,6-dione (28.2 mg, 17.08%) as a yellow solid. [000359] LC-MS: (ES,m/z): [M+1]+=417.10 [000360] 1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 9.18 (s, 1H), 9.00 (s, 1H), 8.04 (d, J = 2.1 Hz, 1H), 7.74 – 7.64 (m, 2H), 7.48 (d, J = 8.3 Hz, 1H), 7.36 (d, J = 2.5 Hz, 1H), 7.29 (dd, J
= 9.0, 2.6 Hz, 1H), 7.19 (s, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 4.39 (d, J = 16.7 Hz, 1H), 4.26 (d, J = 16.7 Hz, 1H), 3.87 (s, 3H), 2.92 (ddd, J = 17.8, 13.5, 5.3 Hz, 1H), 2.61 (d, J = 17.5 Hz, 1H), 2.40 (tt, J = 13.2, 6.6 Hz, 1H), 2.07 – 1.96 (m, 1H). [000361] Example 1.29. Synthesis of 3-(5-{[(6-methoxynaphthalen-2-yl)methyl]amino}-1- oxo-3H-isoindol-2-yl)piperidine-2,6-dione (I-64)
[000362] Into a 25 mL round-bottom flask were added 3-(5-amino-1-oxo-3H-isoindol-2-yl) piperidine-2,6-dione (50 mg, 0.193 mmol, 1.0 equiv), MeOH (4 mL), 6-methoxynaphthalene-2- carbaldehyde (39.50 mg, 0.212 mmol, 1.1 equiv) and AcOH (0.02 mL, 0.349 mmol, 1.81 equiv) at room temperature. The resulting mixture was stirred for 1 h at room temperature. To the above mixture was added NaBH3CN (24.24 mg, 0.386 mmol, 2.0 equiv) at room temperature. The resulting mixture was stirred for additional 4 h at room temperature. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The reaction was quenched by the addition of water (20mL) at room temperature. The resulting mixture was extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (2x10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in Water (0.1% FA), 0% to 100% gradient in 40 min; detector, UV 254 nm. This resulted in 3-(5-{[(6-methoxynaphthalen-2-yl)methyl]amino}-1-oxo-3H- isoindol-2-yl)piperidine-2,6-dione (50 mg, 60.03%) as a white solid. [000363] LC-MS: (ES, m/z): [M+H]+= 430.10. [000364] 1H NMR (300 MHz, DMSO-d6) δ 10.91 (s, 1H), 7.86 – 7.70 (m, 3H), 7.53 – 7.26 (m, 3H), 7.18 – 6.98 (m, 2H), 6.79 – 6.62 (m, 2H), 4.99 (dd, J = 13.3, 5.1 Hz, 1H), 4.48 (d, J = 5.8 Hz, 2H), 4.32 – 4.04 (m, 2H), 3.86 (s, 3H), 2.88 (ddd, J = 17.2, 13.6, 5.4 Hz, 1H), 2.63 – 2.54 (m, 1H), 2.30 (qd, J = 13.2, 4.4 Hz, 1H), 2.04 – 1.80 (m, 1H). [000365] Example 1.30. Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(6-methoxyquinolin-3- yl)-1-oxoisoindoline-5-carboxamide (I-1)
[000366] The synthesis of Compound I-1 was completed following the procedure in Example 1.1 using 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxylic acid and 6- methoxyquinolin-3-amine. [000367] LC-MS: (ES, m/z): [M+H]+ 445.00 [000368] 1H NMR (300 MHz, DMSO-d6) δ 11.03 (s, 1H), 10.85 (s, 1H), 8.89 (dd, J = 51.3, 2.4 Hz, 2H), 8.40 – 8.12 (m, 2H), 7.90 (dd, J = 14.4, 8.5 Hz, 2H), 7.46 – 7.16 (m, 2H), 5.18 (dd, J = 13.3, 5.1 Hz, 1H), 4.70 – 4.39 (m, 2H), 3.91 (s, 3H), 2.96 (d, J = 12.9 Hz, 1H), 2.51-2.50 (m, 1H),2.49-2.47(m, 1H), 2.14 – 1.95 (m, 1H). [000369] Example 1.31. Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-((6- methoxynaphthalen-2-yl)methyl)-1-oxoisoindoline-5-carboxamide (I-23)
[000370] The synthesis of Compound I-23 was completed following the procedure in Example 1.1 using 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxylic acid and (6- methoxynaphthalen-2-yl)methanamine. [000371] LC-MS: (ES,m/z): [M+H]+:458.05 [000372] 1H NMR (300 MHz, DMSO-d6) δ 11.03 (s, 1H), 9.32 (t, J = 5.9 Hz, 1H), 8.14 (s, 1H), 8.06 (dd, J = 8.0, 1.4 Hz, 1H), 7.85-7.75 (m, 4H), 7.47 (dd, J = 8.5, 1.8 Hz, 1H), 7.31 (d, J = 2.6 Hz, 1H), 7.15 (dd, J = 8.9, 2.6 Hz, 1H), 5.16 (dd, J = 13.3, 5.1 Hz, 1H), 4.64 (d, J = 5.8 Hz, 2H), 4.54 (d, J = 17.6 Hz, 1H), 4.40 (d, J = 17.6 Hz, 1H), 3.87 (s, 3H), 2.93 (ddd, J = 17.7, 13.4, 5.3 Hz, 1H), 2.61 (d, J = 17.2 Hz, 1H), 2.41 (td, J = 13.3, 4.5 Hz, 1H), 2.03 (d, J = 12.5 Hz, 1H). [000373] Example 1.32. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-160)
[000374] Into a 25 mL round-bottom flask were added 3-(5-amino-1-oxo-3H-isoindol-2- yl)piperidine-2,6-dione (200 mg, 0.771 mmol, 1 equiv), 1-methylpyrrolo[2,3-b]pyridine-5- carboxylic acid (135.90 mg, 0.771 mmol, 1 equiv), DIEA (299.11 mg, 2.313 mmol, 3 equiv), DMF (5 mL) and HATU (439.98 mg, 1.157 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for 16h at 60°C under nitrogen atmosphere. The mixture was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C18 OBD Column 30*150mm 5μm, n; Mobile Phase A: Water(0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 9% B to 29% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 10.43) to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1- methylpyrrolo[2,3-b]pyridine-5-carboxamide (51.4 mg, 15.21%) as an off-white solid. [000375] LC-MS: (ES, m/z): [M+H]+: 418.00 [000376] 1H-NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.62 (s, 1H), 8.89 (d, J = 2.2 Hz, 1H), 8.61 (d, J = 2.5 Hz, 1H), 8.17 (s, 1H), 7.86 (d, J = 8.5 Hz, 1H), 7.72 (d, J = 8.2 Hz, 1H), 7.67 (d, J = 3.5 Hz, 1H), 6.66 (d, J = 3.5 Hz, 1H), 5.11 (dd, J = 13.4, 5.0 Hz, 1H), 4.48 (d, J = 17.2 Hz, 1H), 4.34 (d, J = 17.1 Hz, 1H), 3.88 (d, J = 2.5 Hz, 3H), 2.99 – 2.87 (m, 1H), 2.61 (d, J = 17.3 Hz, 1H), 2.39 (dt, J = 14.5, 7.2 Hz, 1H), 2.02 (d, J = 11.1 Hz, 1H). [000377] Example 1.33. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-methyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxamide (I-158)
[000378] Synthesis of 6-chloro-1-methyl-1H-pyrazolo[3,4-d]pyrimidine
[000379] Into a 25mL round-bottom flask were added methylhydrazine sulfate (447.97 mg, 3.108 mmol, 1.1 equiv) and Et3N (314.48 mg, 3.108 mmol, 1.1 equiv) in MeOH at 0°C. The resulting mixture was stirred for 0.5h at 0°C under N2 atmosphere. To the above mixture was added 2,4-dichloropyrimidine-5-carbaldehyde (500 mg, 2.825 mmol, 1 equiv) in portions. The resulting mixture was stirred for additional 1 h at 0°C. Then to the above mixture was added Et3N (314.48 mg, 3.108 mmol, 1.1 equiv) dropwise at 0 °C. The resulting mixture was stirred for additional 2h at 0°C. Desired product could be detected by LCMS. The residue was concentrated under reduced pressure and purified he residue was purified by silica gel column chromatography, eluted with PE/EA(40%) to afford 6-chloro-1-methylpyrazolo[3,4-d]pyrimidine (305 mg, 64.04%) as a white solid. [000380] LCMS (ES, m/z): no mass signal [000381] 1H-NMR: (300 MHz, DMSO-d6) δ 9.27 (s, 1H), 8.44 (s, 1H), 4.02 (s, 3H). [000382] Synthesis of methyl 1-methyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxylate
[000383] To a solution of 6-chloro-1-methylpyrazolo[3,4-d]pyrimidine (200 mg, 1.186 mmol, 1 equiv) in MeOH (10 mL) was added Pd(dppf)Cl2 (17.36 mg, 0.024 mmol, 0.02 equiv) and Et3N (240.11 mg, 2.372 mmol, 2 equiv) in a pressure tank. The mixture was purged with nitrogen for 3 mins and then was pressurized to 5 atm with carbon monoxide at 100°C for 1 overnight. The reaction mixture was cooled to room temperature and filtered to remove insoluble solids. The residue was purified by silica gel column chromatography, eluted with EA/PE (100%) to afford as a methyl 1-methylpyrazolo[3,4-d]pyrimidine-6-carboxylate (182 mg, 79.83%) as a white solid. [000384] LC MS (ES, m/z): [M+H]+: 193.0 [000385] 1H-NMR: (300 MHz, DMSO-d6) δ 9.47 (s, 1H), 8.52 (s, 1H), 4.12 (s, 3H), 3.96 (s, 3H). [000386] Synthesis of 1-methyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxylic acid
[000387] To a stirred mixture of methyl 1-methyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxylate (120 mg, 0.624 mmol, 1 equiv) in MeOH (4mL) were added NaOH (24.97 mg, 0.624 mmol, 1 equiv) in H2O (4 mL) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for additional 2 h at room temperature. Desired product could be detected by LCMS. The mixture was neutralized to pH 7 with HCl(aq). The filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water , 5% to 100% gradient in 30 min; detector, UV 254 nm.to afford (1-methylpyrazolo[3,4-d]pyrimidine-6-carboxylic acid (90 mg, 81%) as a white solid. [000388] LC MS (ES, m/z): [M+H]+: 179.00 [000389] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1-methyl-1H- pyrazolo[3,4-d]pyrimidine-6-carboxamide
[000390] Into a 25mL round-bottom flask were added 3-(5-amino-1-oxo-3H-isoindol-2- yl)piperidine-2,6-dione (90.23 mg, 0.348 mmol, 1 equiv), 2,6-dicarboxynaphthalene (41.69 mg, 0.193 mmol, 1 equiv), DIEA (89.96 mg, 0.696 mmol, 2 equiv), DMF (2 mL) and HATU (198.49 mg, 0.522 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for 16h at 60°C under nitrogen atmosphere. The mixture was purified by Prep-HPLC with the following conditions (Column: XBridge Prep Phenyl OBD Column 19*250 mm, 5m; Mobile Phase A: Water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 11% B to 26 % B in 10 min; Wave Length: 254nm/220nm nm; RT1 (min): 16.917) to afford N-[2-(2,6- dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1-methylpyrazolo[3,4-d]pyrimidine-6- carboxamide (26.7 mg, 17.41%) as a yellow solid. [000391] LC MS (ES, m/z): [M+H]+: 420.00 [000392] 1H-NMR: (400 MHz, DMSO-d6) δ 11.16 (s, 1H), 11.00 (s, 1H), 9.54 (s, 1H), 8.54 (s, 1H), 8.27 (d, J = 1.8 Hz, 1H), 8.00 (dd, J = 8.4, 1.8 Hz, 1H), 7.76 (d, J = 8.3 Hz, 1H), 5.12 (dd, J
= 13.3, 5.1 Hz, 1H), 4.51 (d, J = 17.2 Hz, 1H), 4.36 (d, J = 17.2 Hz, 1H), 4.19 (s, 3H), 2.93 (ddd, J = 17.3, 13.7, 5.4 Hz, 1H), 2.66 – 2.57 (m, 1H), 2.41 (qd, J = 13.0, 4.3 Hz, 1H), 2.10 – 1.98 (m, 1H). [000393] Example 1.34. Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-1H-pyrazolo[3,4-d]pyrimidine-6-carboxamide (I-157)
[
[000395] Into a 40 mL vial were added cyclobutyl-hydrazine hydrochloride (228.60 mg, 1.865 mmol, 1.1 equiv) and MeOH (10 mL) at room temperature. To the above mixture was added Et3N (566.06 mg, 5.593 mmol, 3.3 equiv) dropwise at 0°C. The resulting mixture was stirred for additional 0.5 h at 0°C. Then, to the above mixture was added 2,4-dichloropyrimidine-5- carbaldehyde (300 mg, 1.695 mmol, 1 equiv) in MeOH (10 mL) and Et3N (566.06 mg, 5.593 mmol, 3.3 equiv) dropwise at 0°C. The resulting mixture was stirred for additional 2 h at 0°C. The reaction was monitored by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (2:1) to afford 6-chloro-1-cyclobutylpyrazolo[3,4-d]pyrimidine (250 mg, 70.68%) as a yellow solid. [000396] LC-MS (ES, m/z): [M+H]+: 209.05 [000397] 1H-NMR: (400 MHz, DMSO-d6) δ 9.26 (s, 1H), 8.48 (s, 1H), 5.42 – 5.29 (m, 1H), 2.73 – 2.57 (m, 2H), 2.48 – 2.39 (m, 2H), 1.96 – 1.83 (m, 2H). [000398] Synthesis of methyl 1-cyclobutyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxylate
[000399] Into a 50 mL pressure tank reactor were added 6-chloro-1-cyclobutylpyrazolo[3,4- d]pyrimidine (230 mg, 1.102 mmol, 1 equiv), Pd(dppf)Cl2 (16.13 mg, 0.022 mmol, 0.02 equiv), TEA (223.10 mg, 2.204 mmol, 2 equiv) and MeOH (25 mL) at room temperature. The resulting mixture was stirred for overnight at 100°C under carbon monoxide (5 atm) atmosphere. Desired product could be detected by LCMS. The reaction was quenched with Water/Ice at room temperature and concentrated under reduced pressure. The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with water (1 x 20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1:4) to afford methyl 1-cyclobutylpyrazolo[3,4-d]pyrimidine-6-carboxylate (230 mg, 89.84%) as a brown yellow oil. [000400] LC-MS (ES, m/z): [M+H]+: 233.10 [000401] 1H-NMR: (400 MHz, DMSO-d6) δ 9.47 (s, 1H), 8.57 (s, 1H), 5.57 – 5.44 (p, J = 8.1 Hz, 1H), 3.96 (s, 3H), 2.77 – 2.63 (m, 2H), 2.51 – 2.42 (m, 2H), 2.02 – 1.86 (m, 2H). [000402] Synthesis of 1-cyclobutyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxylic acid
[000403] Into a 25 mL round-bottom flask were added methyl 1-cyclobutylpyrazolo[3,4- d]pyrimidine -6-carboxylate (100 mg, 0.431 mmol, 1 equiv), THF (2 mL), H2O (2 mL) and LiOH.H2O (27.10 mg, 0.646 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under air atmosphere. Desired product could be detected by LCMS. The mixture was neutralized to pH = 7 with HCl (aq.). The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 0% to 15% gradient in 15 min; detector, UV 254 nm. This resulted in 1- cyclobutylpyrazolo[3,4-d]pyrimidine-6-carboxylic acid (90 mg, 95.79%) as a yellow solid.
[000404] LC-MS (ES, m/z): [M+H]+: 219.10 [000405] Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1H- pyrazolo[3,4-d]pyrimidine-6-carboxamide
[000406] Into a 40 mL vial were added 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6- dione (100 mg, 0.386 mmol, 1 equiv), 1-cyclobutylpyrazolo[3,4-d]pyrimidine-6-carboxylic acid (84.17 mg, 0.386 mmol, 1 equiv), DIEA (299.11 mg, 2.316 mmol, 6 equiv), DMF (4 mL) and HATU (219.99 mg, 0.579 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 40% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 1-cyclobutyl-N-[2-(2,6- dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]pyrazolo[3,4-d]pyrimidine-6-carboxamide (78.8 mg, 43.84%) as an off-white solid. [000407] LC-MS (ES, m/z): [M+H]+: 460.20 [000408] 1H-NMR: (400 MHz, DMSO-d6) δ 11.13 (s, 1H), 11.00 (s, 1H), 9.54 (s, 1H), 8.60 (s, 1H), 8.26 (s, 1H), 7.99 (dd, J = 8.3, 1.8 Hz, 1H), 7.76 (d, J = 8.3 Hz, 1H), 5.72 – 5.59 (m, 1H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.51 (d, J = 17.2 Hz, 1H), 4.37 (d, J = 17.3 Hz, 1H), 3.00 – 2.87 (m, 1H), 2.80 – 2.66 (m, 2H), 2.66 – 2.57 (m, 1H), 2.56 – 2.51 (m, 2H), 2.47 – 2.35 (m, 1H), 2.07 – 1.89 (m, 3H). [000409] Example 1.35. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-methyl-1H-pyrazolo[4,3-b]pyridine-5-carboxamide (I-155)
[000410] Synthesis of methyl 1-methyl-1H-pyrazolo[4,3-b]pyridine-5-carboxylate and methyl 2-methyl-2H-pyrazolo[4,3-b]pyridine-5-carboxylate
[000411] Into a 40 mL vial were added 1H-pyrazolo[4,3-b]pyridine-5-carboxylic acid (500 mg, 0.306 mmol, 1 equiv) and DMF (4 mL) at room temperature. To the above mixture was added K2CO3 (468.06 mg, 3.386 mmol, 2 equiv) in portions at 0°C. The resulting mixture was stirred for additional 0.5 h at 0°C. To the above mixture was added CH3I (480.71 mg, 3.386 mmol, 2 equiv) dropwise at 0°C. The resulting mixture was stirred for additional overnight at room temperature. The reaction was monitored by LCMS. The resulting mixture was filtered, the filter cake was washed with DMF (2x3 mL). The filtrate was purified by reversed-phase flash
chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 20% to 30% gradient in 10 min; detector, UV 254 nm. This resulted in methyl 1-methyl-1H-pyrazolo[4,3-b]pyridine-5-carboxylate and methyl 2-methyl-2H-pyrazolo[4,3- b]pyridine-5 -carboxylate (300 mg, 92.66%, mixture) as an off-white solid. [000412] LC-MS (ES, m/z): [M+H]+: 191.95 [000413] Synthesis of 1-methyl-1H-pyrazolo[4,3-b]pyridine-5-carboxylic acid and 2-methyl- 2H-pyrazolo[4,3-b]pyridine-5-carboxylic acid
[000414] Into a 40 mL vial were added methyl 1-methyl-1H-pyrazolo[4,3-b]pyridine-5- carboxylate and methyl 2-methyl-2H-pyrazolo[4,3-b]pyridine-5-carboxylate (300 mg, mixture), THF (3 mL), H2O (3 mL) and LiOH.H2O (65.84 mg, 1.569 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature. Desired product could be detected by LCMS. The mixture was acidified to pH = 7 with HCl (aq.). The resulting mixture was concentrated under reduced pressure and purified by Prep-HPLC with the following conditions (Column: Sunfire prep C18 column, 30*150 mm, 5m; Mobile Phase A: Water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 2% B to 14% B in 10 min; Wave Length: 254 nm/ 220 nm; RT1(min): 10.93) to afford 1-methylpyrazolo[4,3- b]pyridine-5-carboxylic acid (80 mg, 43.17%) as an off-white solid and 2-methyl-2H- pyrazolo[4,3-b]pyridine-5-carboxylic acid (30 mg, 16.19%) as an off-white solid. [000415] LC-MS (ES, m/z): [M+H]+: 178.15 [000416] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1-methyl-1H- pyrazolo[4,3-b]pyridine-5-carboxamide
[000417] Into a 40 mL vial were added 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6- dione (80 mg, 0.309 mmol, 1 equiv), 1-methylpyrazolo[4,3-b]pyridine-5-carboxylic acid (54.67 mg, 0.309 mmol, 1 equiv), DIEA (239.29 mg, 1.854 mmol, 6 equiv), DMF (3 mL) and HATU (175.99 mg, 0.464 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at 60°C under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 35% to 45% gradient in 15 min; detector, UV 254 nm. This resulted in N-[2- (2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1-methylpyrazolo[4,3-b]pyridine-5- carboxamide (68.5 mg, 50.46%) as a white solid. [000418] LC-MS: (ES, m/z): [M+H]+: 419.10 [000419] 1H-NMR: (400 MHz, DMSO-d6) δ 11.00 (d, J = 8.0 Hz, 2H), 8.50 (d, J = 0.9 Hz, 1H), 8.40 (dd, J = 8.9, 1.0 Hz, 1H), 8.29 (d, J = 1.8 Hz, 1H), 8.23 (d, J = 8.9 Hz, 1H), 8.04 (dd, J = 8.3, 1.9 Hz, 1H), 7.72 (d, J = 8.3 Hz, 1H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.49 (d, J = 17.2 Hz, 1H), 4.34 (d, J = 17.2 Hz, 1H), 4.17 (s, 3H), 3.00 – 2.86 (m, 1H), 2.66 – 2.56 (m, 1H), 2.48 – 2.34 (m, 1H), 2.06 – 1.97 (m, 1H). [000420] Example 1.36. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 2-methyl-2H-pyrazolo[4,3-b]pyridine-5-carboxamide (I-154)
[000421] Into a 10 mL vial were added 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6- dione (30 mg, 0.116 mmol, 1 equiv), 2-methylpyrazolo[4,3-b]pyridine-5-carboxylic acid (20.50 mg, 0.116 mmol, 1 equiv), DIEA (89.73 mg, 0.696 mmol, 6 equiv), DMF (2 mL) and HATU (66.00 mg, 0.174 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for
overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The residue was purified by reversed- phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 20% to 35% gradient in 20 min; detector, UV 254 nm. This resulted in N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-2-methylpyrazolo[4,3-b]pyridine-5- carboxamide (41.1 mg, 83.62%) as a white solid. [000422] LC-MS (ES, m/z): [M+H]+: 419.15 [000423] 1H-NMR: (400 MHz, DMSO-d6) δ 10.98 (s, 2H), 8.90 (s, 1H), 8.34 – 8.27 (m, 2H), 8.10 – 7.99 (m, 2H), 7.72 (d, J = 8.3 Hz, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 4.49 (d, J = 17.2 Hz, 1H), 4.34 (d, J = 17.2 Hz, 1H), 4.31 (s, 3H), 3.00 – 2.86 (m, 1H), 2.66 – 2.56 (m, 1H), 2.48 – 2.34 (m, 1H), 2.05 – 1.98 (m, 1H). [000424] Example 1.37. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-methyl-1H-pyrazolo[3,4-b]pyridine-5-carboxamide (I-156)
[000425] Into a 40 mL vial were added 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6- dione (50 mg, 0.193 mmol, 1 equiv), 1-methylpyrazolo[3,4-b]pyridine-5-carboxylic acid (34.17 mg, 0.193 mmol, 1 equiv), DMF (2 mL), DIEA (149.55 mg, 1.158 mmol, 6 equiv) and HATU (109.99 mg, 0.289 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight 60°C. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 35% to 45% gradient in 10 min; detector, UV 254 nm. This resulted in N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1- methylpyrazolo[3,4 -b]pyridine-5-carboxamide (26.0 mg, 31.35%) as a white solid. [000426] LC-MS (ES, m/z): [M+H]+:419.05 [000427] 1H-NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.76 (s, 1H), 9.13 (d, J = 2.1 Hz, 1H), 8.89 (d, J = 2.1 Hz, 1H), 8.37 (d, J = 0.8 Hz, 1H), 8.16 (d, J = 1.7 Hz, 1H), 7.85 (dd, J = 8.3, 1.8 Hz, 1H), 7.74 (d, J = 8.3 Hz, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 4.49 (d, J = 17.3 Hz,
1H), 4.35 (d, J = 17.2 Hz, 1H), 4.13 (s, 3H), 3.00 – 2.86 (m, 1H), 2.66 – 2.57 (m, 1H), 2.47 – 2.33 (m, 1H), 2.07 – 1.97 (m, 1H). [000428] Example 1.38. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-methyl-1H-indazole-5-carboxamide (I-153) and N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-2-methyl-2H-indazole-5-carboxamide (I-152)
[000429] Into a 40 mL vial were added 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6- dione (100 mg, 0.386 mmol, 1 equiv), the mixture of 1-methylindazole-5-carboxylic acid and 2- methyl-2H-indazole-5-carboxylic acid (67.95 mg, 0.386 mmol, 1 equiv), DIEA (299.11 mg, 2.316 mmol, 6 equiv), DMF (4 mL) and HATU (219.99 mg, 0.579 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed- phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 30% to 50% gradient in 20 min; detector, UV 254 nm. The mixture of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1-methyl-1H-indazole-5-carboxamide and N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-2-methyl-2H-indazole-5- carboxamide(45 mg, mixture) obtained. The mixture was separated by SFC to afford N-(2-(2,6- dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1-methyl-1H-indazole-5-carboxamide (16.6 mg, 10.14%) as a yellow solid & N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-2-methyl-2H- indazole-5-carboxamide (14.6 mg, 8.92%) as an off-white solid. [000430] LC-MS (ES, m/z) (I-153): [M-H]-: 416.15 [000431] 1H-NMR (I-153): (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.60 (s, 1H), 8.51 (s, 1H), 8.27 (s, 1H), 8.18 (s, 1H), 8.02 (dd, J = 8.9, 1.6 Hz, 1H), 7.87 (dd, J = 8.3, 1.8 Hz, 1H), 7.78 (d, J = 8.9 Hz, 1H), 7.72 (d, J = 8.3 Hz, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 4.48 (d, J = 17.2 Hz,
1H), 4.34 (d, J = 17.2 Hz, 1H), 4.11 (s, 3H), 3.00 – 2.86 (m, 1H), 2.66 – 2.57 (m, 1H), 2.47 – 2.33 (m, 1H), 2.06 – 1.97 (m, 1H). [000432] LC-MS (ES, m/z) (I-152): [M-H]-: 416.20 [000433] 1H-NMR (I-152): (400 MHz, DMSO-d6) δ 11.00 (s, 1H), 10.52 (s, 1H), 8.54 – 8.49 (m, 1H), 8.31 – 8.24 (m, 2H), 8.07 – 7.97 (m, 2H), 7.82 – 7.74 (m, 1H), 7.60 (d, J = 8.3 Hz, 1H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.45 (d, J = 17.1 Hz, 1H), 4.32 (d, J = 17.1 Hz, 1H), 4.11 (s, 3H), 2.99 – 2.85 (m, 1H), 2.66 – 2.57 (m, 1H), 2.48 – 2.35 (m, 1H), 2.09 – 1.98 (m, 1H). [000434] Example 1.39. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 3-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-151)
[000435] Into a 25mL round-bottom flask were added 3-(5-amino-1-oxo-3H-isoindol-2-yl) piperidine-2,6-dione (200 mg, 0.771 mmol, 1 equiv), 3-methyl-1H-pyrrolo[2,3-b] pyridine-5- carboxylic acid (135.90 mg, 0.771 mmol, 1 equiv), DIEA (299.11 mg, 2.313 mmol, 3 equiv), DMF (10 mL) and HATU (439.98 mg, 1.157 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for 16h at 60°C under nitrogen atmosphere. Desired product could be detected by LCMS. The mixture was purified directly by Prep-HPLC with the following conditions (Column: Xselect CSH F-Phenyl OBD column 30*250 mm, 5μm; Mobile Phase A: Water (0.05% TFA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 12% B to28% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 11.32/12.08) to afford N-[2- (2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-3-methyl-1H-pyrrolo[2,3-b]pyridine-5- carboxamide (6.7 mg, 1.95%) as a white solid. [000436] LC-MS (ES, m/z):[M+H]+: 418.05 [000437] 1H-NMR: (300 MHz, DMSO-d6) δ 11.70 (s, 1H), 11.00 (s, 1H), 10.58 (s, 1H), 8.83 (d, J = 2.1 Hz, 1H), 8.57 (d, J = 2.1 Hz, 1H), 8.18 (s, 1H), 7.87 (d, J = 9.8 Hz, 1H), 7.73 (d, J = 8.3 Hz, 1H), 7.38 (s, 1H), 5.11 (dd, J = 13.2, 4.9 Hz, 1H), 4.41 (dd, J=18 Hz, 2H), 3.01-2.85 (m, 1H), 2.64 (s, 1H), 2.41 (d, J = 12.8 Hz, 1H), 2.34 (d, J = 1.1 Hz, 3H), 2.02 (s, 1H).
[000438] Example 1.40. Synthesis of (R)-N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5- yl)-3-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-150)
[000439] Into a 25mL round-bottom flask were added 3-(5-amino-1-oxo-3H-isoindol-2-yl) piperidine-2,6-dione (200 mg, 0.771 mmol, 1 equiv), 3-methyl-1H-pyrrolo[2,3-b] pyridine-5- carboxylic acid (135.90 mg, 0.771 mmol, 1 equiv), DIEA (299.11 mg, 2.313 mmol, 3 equiv), DMF (10 mL) and HATU (439.98 mg, 1.157 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for 16h at 60°C under nitrogen atmosphere. Desired product could be detected by LCMS. The mixture was purified by Prep-HPLC with the following conditions (Column: Xselect CSH F-Phenyl OBD column 30*250 mm, 5μm; Mobile Phase A: water (0.05% TFA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 12% B to28% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 11.32/12.08) to afford N-[2-(2,6- dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-3-methyl-1H-pyrrolo[2,3-b] pyridine-5- carboxamide (40 mg, 1.95%) as a white solid. The product was was purified by PREP_CHIRAL_HPLC- with the following conditions (Column: CHIRAL ART Cellulose-SC, 3*25 cm, 5 μm; Mobile Phase A: MtBE: DCM=1: 2(0.1%TFA), Mobile Phase B: MeOH; Flow rate: 40 mL/min; Gradient: isocratic 50; Wave Length: 288/224 nm; RT1 (min): 5.4; RT2 (min): 6.5; Sample Solvent: MeOH: DCM=1: 1(0.1% TFA); Injection Volume: 0.6 mL; Number Of Runs: 5) to afford N-{2-[(3R)-2,6-dioxopiperidin-3-yl]-1-oxo-3H-isoindol-5-yl}-3-methyl-1H- pyrrolo[2,3-b]pyridine-5-carboxamide (9.4 mg, 5.68%) as a white solid. [000440] LC-MS (ES, m/z):[M+H]+: 418.00 [000441] 1H-NMR: (300 MHz, DMSO-d6) δ 11.70 (s, 1H), 11.00 (s, 1H), 10.58 (s, 1H), 8.84 (d, J = 2.1 Hz, 1H), 8.58 (d, J = 2.2 Hz, 1H), 8.18 (s, 1H), 7.87 (d, J = 7.8 Hz, 1H), 7.73 (d, J = 8.3 Hz, 1H), 7.38 (s, 1H), 5.18 – 5.07 (m, 1H), 4.49 (d, J=18Hz, 1H), 4.37 (d, J=18Hz, 1H), 2.93 (s, 1H), 2.64 (s, 1H), 2.39 (s, 1H), 2.34 (d, J = 1.1 Hz, 3H), 2.02 (s, 1H). [000442] Example 1.41. Synthesis of (S)-N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5- yl)-3-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-149)
[000443] Into a 25mL round-bottom flask were added 3-(5-amino-1-oxo-3H-isoindol-2-yl) piperidine-2,6-dione (200 mg, 0.771 mmol, 1 equiv), 3-methyl-1H-pyrrolo[2,3-b] pyridine-5- carboxylic acid (135.90 mg, 0.771 mmol, 1 equiv), DIEA (299.11 mg, 2.313 mmol, 3 equiv), DMF (10 mL) and HATU (439.98 mg, 1.157 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for 16h at 60°C under nitrogen atmosphere. Desired product could be detected by LCMS. The mixture was purified by Prep-HPLC with the following conditions (Column: Xselect CSH F-Phenyl OBD column 30*250 mm, 5μm; Mobile Phase A: water (0.05% TFA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 12% B to28% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 11.32/12.08) to afford N-[2-(2,6- dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-3-methyl-1H-pyrrolo[2,3-b] pyridine-5- carboxamide (40 mg, 1.95%) as a white solid. The product was purified by PREP_CHIRAL_HPLC- with the following conditions (Column: CHIRAL ART Cellulose-SC, 3*25 cm, 5 μm; Mobile Phase A: MtBE: DCM=1: 2(0.1%TFA), Mobile Phase B: MEOH; Flow rate: 40 mL/min; Gradient: isocratic 50; Wave Length: 288/224 nm; RT1(min): 5.4; RT2(min): 6.5; Sample Solvent: MeOH: DCM=1: 1(0.1% TFA); Injection Volume: 0.6 mL; Number Of Runs: 5) to afford N-{2-[(3S)-2,6-dioxopiperidin-3-yl]-1-oxo-3H-isoindol-5-yl}-3-methyl-1H- pyrrolo[2,3-b]pyridine-5-carboxamide (8.7 mg, 5.34%) as a white solid. [000444] LC-MS (ES, m/z):[M+H]+: 418.05 [000445] 1H-NMR: (300 MHz, DMSO-d6) δ 11.70 (s, 1H), 11.00 (s, 1H), 10.58 (s, 1H), 8.84 (d, J = 2.1 Hz, 1H), 8.58 (d, J = 2.2 Hz, 1H), 8.18 (s, 1H), 7.87 (d, J = 7.8 Hz, 1H), 7.73 (d, J = 8.3 Hz, 1H), 7.38 (s, 1H), 5.18 – 5.07 (m, 1H), 4.49 (d, J=18Hz, 1H), 4.37 (d, J=18Hz, 1H), 2.93 (s, 1H), 2.64 (s, 1H), 2.39 (s, 1H), 2.34 (d, J = 1.1 Hz, 3H), 2.02 (s, 1H). [000446] Example 1.42. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-ethyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-147)
[000447] Synthesis of methyl 1-ethyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate
[000448] In a 50mL round-bottom flask, to a solution of methyl 1H-pyrrolo[2,3-b] pyridine-5- carboxylate (200 mg, 1.135 mmol, 1 equiv) in DMF (15 mL) was added Cs2CO3 (1109.64 mg, 3.405 mmol, 3 equiv) at 0 °C. The mixture was stirred for 15 min. Then iodoethane (194.76 mg, 1.249 mmol, 1.1 equiv) was added and the mixture was allowed to warm to room temperature and stirred for 1 overnight. Desired product could be detected by LCMS. The reaction was quenched with ice water (50 mL) at 0 °C. The resulting mixture was extracted with EA (3x50 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, H2O in ACN, 36% to 40% gradient in 4 min; detector, UV 254 nm. This resulted in methyl 1- ethylpyrrolo[2,3-b] pyridine-5-carboxylate (162 mg, 69.87%) as a yellow oil. [000449] LC-MS (ES, m/z): [M+H]+: 205.05 [000450] Synthesis of 1-ethyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000451] Into a 25 mL round-bottom flask were added methyl 1-ethylpyrrolo[2,3-b] pyridine- 5-carboxylate (100 mg, 0.490 mmol, 1 equiv) in MeOH (1 mL) and NaOH (19.58 mg, 0.490 mmol, 1 equiv) in H2O (1 mL) dropwises at 0 °C under N2 atmosphere. The resulting mixture was stirred for additional 2 h at room temperature. Desired product could be detected by LCMS. The mixture was neutralized to pH 7 with 1M HCl(aq). The resulting mixture was concentrated under reduced pressure. The crude product 1-ethylpyrrolo[2,3-b] pyridine-5-carboxylic acid (150 mg, crude) was used in the next step directly without further purification. [000452] LC-MS (ES, m/z): [M+H]+: 191.05 [000453] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1-ethyl-1H- pyrrolo[2,3-b]pyridine-5-carboxamide
[000454] Into a 25mL round-bottom flask were added 1-ethylpyrrolo[2,3-b] pyridine-5- carboxylic acid (93 mg, 0.489 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2-yl) piperidine- 2,6-dione (126.77 mg, 0.489 mmol, 1 equiv), DIEA (189.59 mg, 1.467 mmol, 3 equiv), DMF (2 mL) and HATU (278.88 mg, 0.734 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for 16h at 60°C under nitrogen atmosphere. Desired product could be detected by LCMS. The mixture was purified directly by Prep-HPLC with the following conditions (Column: XBridge Prep Phenyl OBD Column 19*250 mm, 5m; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 11% B to 26 % B in 10 min; Wave Length: 254nm/220nm nm; RT (min): 16.917) to afford N-[2-(2,6-dioxopiperidin-3- yl)-1-oxo-3H-isoindol-5-yl]-1-ethylpyrrolo[2,3-b] pyridine-5-carboxamide (17.4 mg, 8.20%) as a white solid. [000455] LC-MS (ES, m/z):[M+H]+: 432.05 [000456] 1H-NMR: (400 MHz, DMSO-d6) δ 11.00 (s, 1H), 10.62 (s, 1H), 8.87 (d, J = 2.1 Hz, 1H), 8.60 (d, J = 2.2 Hz, 1H), 8.18 (d, J = 1.7 Hz, 1H), 7.86 (dd, J = 8.4, 1.8 Hz, 1H), 7.77 – 7.69 (m, 2H), 6.66 (d, J = 3.5 Hz, 1H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.48 (d, J = 17.3 Hz, 1H), 4.41
– 4.30 (m, 3H), 2.97-2.88 (m, 1H), 2.61 (d, J = 17.2 Hz, 1H), 2.49-2.34 (m, 1H), 2.06 – 1.98 (m, 1H), 1.41 (t, J = 7.2 Hz, 3H). [000457] Example 1.43. Synthesis of 1-cyclopropyl-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-146)
[000458] Synthesis of methyl 1-cyclopropyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate
[000459] Into a 50mL round-bottom flask were added methyl 1H-pyrrolo[2,3-b] pyridine-5- carboxylate (200 mg, 1.135 mmol, 1 equiv), cyclopropylboronic acid (195.03 mg, 2.270 mmol, 2 equiv), Na2CO3 (240.64 mg, 2.270 mmol, 2 equiv), Cu(OAc)2 (206.20 mg, 1.135 mmol, 1 equiv), 2,2’-Bipyridine (177.30 mg, 1.135 mmol, 1 equiv) and DCE(5 mL) at room 140emperature. The mixture was stirred for 1 overnight at 110°C. Desired product could be detected by LCMS. The residue was purified directly by silica gel column chromatography, eluted with PE/EA (2/3) to afford methyl 1-cyclopropylpyrrolo[2,3-b] pyridine-5-carboxylate (150 mg, 61.10%) as a yellow oil. [000460] LC-MS (ES, m/z) :[M+H]+: 217.00 [000461] Synthesis of 1-cyclopropyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000462] Into a 25mL round-bottom flask were added methyl 1-cyclopropylpyrrolo[2,3-b] pyridine-5-carboxylate (100 mg, 0.462 mmol, 1 equiv) in MeOH (3 mL) and NaOH (18.50 mg, 0.462 mmol, 1 equiv) in H2O (3 mL) at 0 °C under N2 atmosphere. The resulting mixture was stirred for additional 2 h at room temperature. Desired product could be detected by LCMS. The mixture was neutralized to pH 7 with HCl (aq). The resulting mixture was concentrated under reduced pressure. The crude product 1-cyclopropylpyrrolo[2,3-b]pyridine-5-carboxylic acid (151 mg, crude) was used in the next step directly without further purification. [000463] LC-MS (ES, m/z): [M+H]+: 203.00 [000464] Synthesis of 1-cyclopropyl-N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1H- pyrrolo[2,3-b]pyridine-5-carboxamide
[000465] Into a 25mL round-bottom flask were added 1-cyclopropylpyrrolo[2,3-b] pyridine-5- carboxylic acid (100 mg, 0.495 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine- 2, 6-dione (128.21 mg, 0.495 mmol, 1 equiv), DIEA (191.75 mg, 1.485 mmol, 3 equiv), DMF (5 mL) and HATU (282.06 mg, 0.742 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for 16h at 60°C under nitrogen atmosphere. The mixture was purified directly by Prep-HPLC with the following conditions (Column: Xbridge Prep Phenyl OBD Column 19*250 mm, 5m; Mobile Phase A: water (0.1% FA), Mobile Phase B: CAN; Flow rate: 60 mL/min mL/min; Gradient: 12% B to 27 % B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 16.779) to afford 1-cyclopropyl-N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5- yl]pyrrolo[2,3-b]pyridine-5-carboxamide (64.9 mg, 29.06%) as a white solid. [000466] LC-MS (ES, m/z):[M+H]+: 444.05 [000467] 1H-NMR: (400 MHz, DMSO-d6) δ 11.00 (s, 1H), 10.63 (s, 1H), 8.89 (d, J = 2.1 Hz, 1H), 8.59 (d, J = 2.2 Hz, 1H), 8.18 (d, J = 1.8 Hz, 1H), 7.86 (dd, J = 8.3, 1.8 Hz, 1H), 7.73 (d, J =
8.3 Hz, 1H), 7.62 (d, J = 3.6 Hz, 1H), 6.62 (d, J = 3.6 Hz, 1H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.49 (d, J = 17.2 Hz, 1H), 4.34 (d, J = 17.2 Hz, 1H), 3.68 (tt, J = 6.9, 4.1 Hz, 1H), 2.93 (ddd, J = 17.2, 13.6, 5.4 Hz, 1H), 2.61 (ddd, J = 17.3, 4.5, 2.3 Hz, 1H), 2.41 (qd, J = 13.2, 4.4 Hz, 1H), 2.10 – 1.97 (m, 1H), 1.16 – 1.00 (m, 4H). [000468] Example 1.44. Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-148)
[000469] Synthesis of methyl 1-cyclobutyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate
[000470] In a 100 mL round-bottom flask were added methyl 1H-pyrrolo[2,3-b] pyridine-5- carboxylate (300 mg, 1.703 mmol, 1 equiv) in DMF (25 mL) and Cs2CO3 (1664.46 mg, 5.109 mmol, 3 equiv) at 0 °C. The mixture was stirred for 15 min. Then iodocyclobutane (340.92 mg, 1.873 mmol, 1.1 equiv) was added and the mixture was allowed to warm to RT and stirred for 1 overnight. Desired product could be detected by LCMS. The mixture was filtrated. The filtration was purified directly by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, H2O in ACN, 63% to 69% gradient in 8 min; detector, UV 254 nm. This resulted in methyl 1-cyclobutylpyrrolo[2,3-b]pyridine-5-carboxylate (181 mg, 46.16%) as a solid. [000471] LC-MS (ES, m/z): [M+ H]+:231.00
[000472] 1H-NMR: (400 MHz, DMSO-d6) 8.82 (d, J = 2.0 Hz 1H), 8.54 (d, J = 2.0 Hz, 1H), 7.97 (d, J = 3.6 Hz, 1H), 6.69 (d, J = 3.6 Hz, 1H), 5.43-5.30 (m, 1H), 3.89 (s, 3H), 2.61-2.54 (m, 1H), 2.48-2.39 (m, 2H), 1.86 (td, J = 10.0, 5.4 Hz, 2H). [000473] Synthesis of 1-cyclobutyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000474] Into a 25mL round-bottom flask were added methyl 1-cyclobutylpyrrolo[2,3-b] pyridine-5-carboxylate (100 mg, 0.434 mmol, 1 equiv) in MeOH (1 mL) and NaOH (17.37 mg, 0.434 mmol, 1 equiv) in H2O (1 mL) at 0 °C under N2 atmosphere. The resulting mixture was stirred for additional 2 h at room temperature. Desired product could be detected by LCMS. The mixture was neutralized to pH 7 with HCl(aq). The resulting mixture was concentrated under reduced pressure. The crude product 1-cyclobutylpyrrolo[2,3-b] pyridine-5-carboxylic acid) was used in the next step directly without further purification. [000475] LC-MS (ES, m/z): [M+H]+:217.00 [000476] Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1H- pyrrolo[2,3-b]pyridine-5-carboxamide
[000477] Into a 25mL round-bottom flask were added 3-(5-amino-1-oxo-3H-isoindol-2-yl) piperidine-2,6-dione (50.75 mg, 0.196 mmol, 0.83 equiv), 1-cyclobutylpyrrolo[2,3-b] pyridine-5- carboxylic acid (51 mg, 0.236 mmol, 1.00 equiv), DIEA (91.45 mg, 0.708 mmol, 3 equiv), DMF (5 mL) and HATU (134.52 mg, 0.354 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for 16h at 60°C under nitrogen atmosphere. The mixture was purified directly by Prep-HPLC with the following conditions (Column: XBridge Prep Phenyl OBD Column 19*250 mm, 5m; Mobile Phase A: water (0.1% FA), Mobile Phase B: MEOH; Flow rate: 60 mL/min mL/min; Gradient: 35% B to 50% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 12.35) to afford 1-cyclobutyl-N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol- 5-yl] pyrrolo[2,3-b] pyridine-5-carboxamide (21.5 mg, 19.53%) as an off-white solid. [000478] LC-MS (ES, m/z): [M+H]+:458.05
[000479] 1H-NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.61 (s, 1H), 8.87 (d, J = 2.1 Hz, 1H), 8.60 (d, J = 2.1 Hz, 1H), 8.18 (d, J = 1.8 Hz, 1H), 7.96 (d, J = 3.6 Hz, 1H), 7.86 (dd, J = 8.3, 1.8 Hz, 1H), 7.72 (d, J = 8.3 Hz, 1H), 6.70 (d, J = 3.6 Hz, 1H), 5.44 – 5.30 (m, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 4.42 (dd, J=18 Hz, 2H), 2.93 (ddd, J = 17.2, 13.5, 5.4 Hz, 1H), 2.64-2.52 (m, 3H), 2.48 – 2.33 (m, 3H), 2.08-2.00 (m, 1H), 1.91-1.84 (m, 2H). [000480] Example 1.45. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 3-methyl-3H-imidazo[4,5-b]pyridine-6-carboxamide (I-145)
[000481] Into a 40 mL vial were added 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6- dione (100 mg, 0.386 mmol, 1 equiv), 3-methylimidazo[4,5-b]pyridine-6-carboxylic acid (68.33 mg, 0.386 mmol, 1 equiv), DIEA (299.11 mg, 2.316 mmol, 6 equiv), DMF (4 mL) and HATU (219.99 mg, 0.579 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at 60°C. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 25% to 30% gradient in 10 min; detector, UV 254 nm. This resulted in N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-3- methylimidazo[4,5 -b]pyridine-6-carboxamide (9.6 mg, 5.88%) as a white solid. [000482] LC-MS (ES, m/z): [M+H]+: 419.00 [000483] 1H-NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.69 (s, 1H), 9.00 (d, J = 2.0 Hz, 1H), 8.71 (d, J = 2.0 Hz, 1H), 8.59 (s, 1H), 8.18 (d, J = 1.7 Hz, 1H), 7.87 (dd, J = 8.4, 1.8 Hz, 1H), 7.73 (d, J = 8.3 Hz, 1H), 5.11 (dd, J = 13.3, 5.0 Hz, 1H), 4.49 (d, J = 17.3 Hz, 1H), 4.35 (d, J = 17.2 Hz, 1H), 3.91 (s, 3H), 3.00 – 2.86 (m, 1H), 2.65 – 2.57 (m, 1H), 2.47 – 2.34 (m, 1H), 2.06 – 1.98 (m, 1H). [000484] Example 1.46. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 7-methoxyquinoline-3-carboxamide (I-144)
[000485] Into a 25mL round-bottom flask were added 3-(5-amino-1-oxo-3H-isoindol-2- yl)piperidine-2,6-dione (100 mg, 0.386 mmol, 1.00 equiv), 7-methoxyquinoline-3-carboxylic acid (78.37 mg, 0.386 mmol, 1.00 equiv), DIEA (149.55 mg, 1.158 mmol, 3 equiv), DMF (5.00 mL, 64.659 mmol, 167.51 equiv) and HATU (219.99 mg, 0.579 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for 16h at 60°C under nitrogen atmosphere. The mixture was purified by Prep-HPLC with the following conditions (Column: Sunfire prep C18 column, 30*150 mm, 5m; Mobile Phase A: Water(0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 10% B to25% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 15.73) to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-7- methoxyquinoline-3-carboxamide (93.2 mg, 53.06%) as an off-white solid. [000486] LC-MS (ES,m/z):[M+H]+: 445.05 [000487] 1H-NMR: (400 MHz, DMSO-d6) δ 11.00 (s, 1H), 10.83 (s, 1H), 9.32 (d, J = 2.2 Hz, 1H), 8.91 (d, J = 2.3 Hz, 1H), 8.18 (d, J = 1.7 Hz, 1H), 8.08 (d, J = 9.1 Hz, 1H), 7.87 (dd, J = 8.3, 1.8 Hz, 1H), 7.75 (d, J = 8.3 Hz, 1H), 7.51 (d, J = 2.5 Hz, 1H), 7.38 (dd, J = 9.0, 2.5 Hz, 1H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.50 (d, J = 17.2 Hz, 1H), 4.36 (d, J = 17.3 Hz, 1H), 3.98 (s, 3H), 2.93 (ddd, J = 17.2, 13.5, 5.3 Hz, 1H), 2.66 – 2.57 (m, 1H), 2.41 (qd, J = 13.3, 4.4 Hz, 1H), 2.06 – 1.99 (m, 1H). [000488] Example 1.47. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 6-methylquinoline-3-carboxamide (I-141)
[000489] Into a 25 mL round-bottom flask were added 3-(5-amino-1-oxo-3H-isoindol-2- yl)piperidine-2,6-dione (50 mg, 0.193 mmol, 1 equiv) and 6-methylquinoline-3-carboxylic acid (54.15 mg, 0.289 mmol, 1.5 equiv) in DMF (3 mL, 38.765 mmol, 201.01 equiv) were added HATU (109.99 mg, 0.289 mmol, 1.5 equiv) and DIEA (49.85 mg, 0.386 mmol, 2 equiv) in
portions at room temperature under air atmosphere. The resulting mixture was stirred for overnight at 60°C under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 30% to 70% gradient in 20 min; detector, UV 254 nm. This resulted in N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5- yl]-6-methylquinoline-3-carboxamide (23.9 mg, 28.64%) as a white solid. [000490] LC-MS (ES, m/z): [M+H]+=429.15 [000491] 1H-NMR: (400 MHz, DMSO-d6) δ 11.00 (s, 1H), 10.89 (s, 1H), 9.30 (d, J = 2.2 Hz, 1H), 8.87 (d, J = 2.2 Hz, 1H), 8.19 (d, J = 1.7 Hz, 1H), 8.03 (d, J = 8.6 Hz, 1H), 7.92 (s, 1H), 7.88 (dd, J = 8.3, 1.8 Hz, 1H), 7.79 – 7.72 (m, 2H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.50 (d, J = 17.2 Hz, 1H), 4.36 (d, J = 17.2 Hz, 1H), 2.95-2.90 (m, 1H), 2.62 (d, J = 17.8 Hz, 1H), 2.56 (s, 3H), 2.43-2.39 (m, 1H), 2.10 – 1.98 (m, 1H). [000492] Example 1.48. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-(oxetan-3-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-140)
[000493] Synthesis of methyl 1-(oxetan-3-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxylate
[000494] Into a 25mL round-bottom flask were added methyl 3-methyl-1H-pyrrolo[2,3- b]pyridine-5-carboxylate (200 mg, 1.052 mmol, 1 equiv) in DMF (15 mL) and Cs2CO3 (1109.64 mg, 3.405 mmol, 3 equiv) at 0 degrees C. The mixture was stirred for 15 min. Then 3- iodooxetane (229.74 mg, 1.249 mmol, 1.1 equiv) was dropwise at 0 ºC. The resulting mixture
was stirred for 1 overnight at 40 ºC under nitrogen atmosphere. Desired product could be detected by LCMS. The reaction involves filtration, washing the filter cake with DMF (2 x 5mL). The filtrate was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in H2O, 75 to 80% gradient in 10 min; detector, UV 254 nm. This resulted in methyl 1-(oxetan-3-yl)pyrrolo[2,3-b]pyridine-5- carboxylate (225 mg, 85.34%) as a white solid. [000495] LC-MS (ES, m/z) :[M+H]+: 233.05 [000496] Synthesis of 1-(oxetan-3-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000497] Into a 25mL round-bottom flask were added methyl 1-(oxetan-3-yl)pyrrolo[2,3- b]pyridine-5-carboxylate (100 mg, 0.431 mmol, 1 equiv) in MeOH (2 mL) and NaOH (17.22 mg, 0.431 mmol, 1 equiv) in H2O (2 mL) was dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for additional 2 h at room temperature. Desired product could be detected by LCMS. The mixture was neutralized to pH 7 with HCl (aq). The resulting mixture was concentrated under reduced pressure and was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in H2O, 75 to 80% gradient in 10 min; detector, UV 254 nm. This resulted in 1-(oxetan-3- yl)pyrrolo[2,3-b]pyridine-5-carboxylic acid (62 mg, 66%) as a white solid. [000498] LC-MS (ES, m/z): [M+H]+: 219.00 [000499] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1-(oxetan-3-yl)- 1H-pyrrolo[2,3-b]pyridine-5-carboxamide
[000500] Into a 25mL round-bottom flask were added 1-(oxetan-3-yl)pyrrolo[2,3-b]pyridine-5- carboxylic acid (182 mg, 0.834 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine- 2,6-dione (216.24 mg, 0.834 mmol, 1 equiv), DIEA (323.40 mg, 2.502 mmol, 3 equiv), DMF (5 mL) ) and HATU (475.70 mg, 1.251 mmol, 1.5 equiv) at room temperature. The resulting
mixture was stirred for 16h at 60°C under nitrogen atmosphere. The mixture was purified by Prep-HPLC with the following conditions (Column: Sunfire prep C18 column 30*150 mm, 5m; Mobile Phase A: Water(0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 10% B to 30% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 10.6) to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1-(oxetan-3-yl)pyrrolo[2,3- b]pyridine-5-carboxamide (26.0 mg, 6.73%) as a white solid. [000501] LC-MS (ES,m/z):[M+H]+: 460.10 [000502] 1H-NMR: (400 MHz, DMSO-d6) δ 11.01 (s, 1H), 10.65 (s, 1H), 8.88 (d, J = 2.1 Hz, 1H), 8.64 (d, J = 2.1 Hz, 1H), 8.18 (d, J = 1.8 Hz, 1H), 8.11 (d, J = 3.6 Hz, 1H), 7.86 (dd, J = 8.3, 1.8 Hz, 1H), 7.73 (d, J = 8.3 Hz, 1H), 6.80 (d, J = 3.6 Hz, 1H), 6.05 (p, J = 7.2 Hz, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 5.05 (d, J = 7.2 Hz, 4H), 4.48 (d, J = 17.3 Hz, 1H), 4.34 (d, J = 17.2 Hz, 1H), 2.93 (ddd, J = 17.4, 13.6, 5.4 Hz, 1H), 2.61 (d, J = 17.3 Hz, 1H), 2.40 (qd, J = 13.4, 4.5 Hz, 1H), 2.08 – 1.97 (m, 1H). [000503] Example 1.49. Synthesis of tert-butyl 3-(5-((2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)carbamoyl)-1H-pyrrolo[2,3-b]pyridin-1-yl)azetidine-1-carboxylate (I- 1
[000504] Synthesis of methyl 1-(1-(tert-butoxycarbonyl)azetidin-3-yl)-1H-pyrrolo[2,3- b]pyridine-5-carboxylate
[000505] Into a 25mL round-bottom flask were added methyl 1H-pyrrolo[2,3-b]pyridine-5- carboxylate (200 mg, 1.135 mmol, 1 equiv) in DMF (5 mL) and Cs2CO3 (1109.64 mg, 3.405 mmol, 3 equiv) at 0 degrees C. The mixture was stirred for 15 min. Then tert-butyl 3- iodoazetidine-1-carboxylate (353.53 mg, 1.249 mmol, 1.1 equiv) was dropwise at 0 degrees C.
The resulting mixture was stirred for 1 overnight at rt under nitrogen atmosphere. Desired product could be detected by LCMS. The reaction involves filtration, washing the filter cake with DMF (2x3mL). The filtrate was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in H2O, 75 to 80% gradient in 10 min; detector, UV 254 nm. This resulted in tert-butyl 3-[5-(methoxycarbonyl)pyrrolo[2,3- b]pyridin-1-yl]azetidine-1-carboxylate (305 mg, 81.08%) as a white solid. [000506] LC-MS (ES, m/z) :[M+H]+: 332.00 [000507] Synthesis of 1-(1-(tert-butoxycarbonyl)azetidin-3-yl)-1H-pyrrolo[2,3-b]pyridine-5- carboxylic acid
[000508] Into a 25mL round-bottom flask were added tert-butyl 3-[5- (methoxycarbonyl)pyrrolo[2,3-b]pyridin-1-yl]azetidine-1-carboxylate (100 mg, 0.302 mmol, 1 equiv) in THF (2 mL) and NaOH (12.07 mg, 0.302 mmol, 1 equiv) in H2O (2 mL) at 0°C. The resulting mixture was stirred for additional 2 h at room temperature. Desired product could be detected by LCMS. The mixture was neutralized to pH 7 with HCl(aq). The resulting mixture was concentrated under reduced pressure and was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in H2O, 5 to 80% gradient in 30 min; detector, UV 254 nm. This resulted in 1-[1-(tert- butoxycarbonyl)azetidin-3-yl]pyrrolo[2,3-b]pyridine-5-carboxylic acid (68 mg, 71%) as a white solid. [000509] LC-MS (ES, m/z): [M+H]+: 318.00 [000510] Synthesis of tert-butyl 3-(5-((2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5- yl)carbamoyl)-1H-pyrrolo[2,3-b]pyridin-1-yl)azetidine-1-carboxylate
[000511] Into a 25mL round-bottom flask were added 1-(oxetan-3-yl)pyrrolo[2,3-b]pyridine-5- carboxylic acid (182 mg, 0.834 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-
2,6-dione (163.40 mg, 0.630 mmol, 1 equiv), DIEA (244.37 mg, 1.890 mmol, 3 equiv), DMF (10 mL) and HATU (359.45 mg, 0.945 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for 16h at 60 C under nitrogen atmosphere. The mixture was purified by Prep- HPLC with the following conditions (Column: XBridge Shield RP18 OBD Column 30*150 mm, 5m; Mobile Phase A: Water(10 mmol/L H2O), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 24% B to 42% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 11.07) to afford tert-butyl 3-(5-{[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5- yl]carbamoyl}pyrrolo[2,3-b]pyridin-1-yl)azetidine-1-carboxylate (5.6 mg, 1.59%) as a white solid. [000512] LC-MS (ES, m/z):[M+H]+: 459.05 [000513] 1H-NMR: (300 MHz, DMSO-d6) δ 10.98 (s, 1H), 10.65 (s, 1H), 8.88 (d, J = 2.0 Hz, 1H), 8.64 (s, 1H), 8.18 (s, 1H), 7.96 (d, J = 3.6 Hz, 1H), 7.87 (d, J = 8.4 Hz, 1H), 7.73 (d, J = 8.4 Hz, 1H), 6.75 (d, J = 3.6 Hz, 1H), 5.65 (s, -1H), 5.11 (d, J = 8.7 Hz, -1H), 4.66 – 4.20 (m, 6H), 2.91 (d, J = 12.3 Hz, -1H), 2.62 (d, J = 17.3 Hz, -1H), 2.42 (d, J = 12.5 Hz, -1H), 2.03 (s, -1H), 1.45 (s, 9H). [000514] Example 1.50. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 5-methyl-5H-pyrrolo[2,3-b]pyrazine-2-carboxamide (I-132)
[000515] Synthesis of methyl 5-methyl-5H-pyrrolo[2,3-b]pyrazine-2-carboxylate
[000516] Into a 30 mL pressure tank reactor were added 2-bromo-5-methylpyrrolo[2,3- b]pyrazine (900 mg, 4.244 mmol, 1 equiv) and Pd(dppf)Cl2 (1552.79 mg, 2.122 mmol, 0.5 equiv), TEA (1288.47 mg, 12.732 mmol, 3 equiv) in MeOH(10 mL). The resulting mixture was stirred for overnight at 120°C under carbon monoxide atmosphere 5 atm. The reaction was monitored by LCMS. The mixture were collected by filtration and washed with MeOH (10 mL). The resulting mixture was concentrated under vacuum and dissolved in DMF (2 mL). The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 10% to 85% gradient in 35 min; detector, UV 254 nm to afford methyl 5-methylpyrrolo[2,3-b]pyrazine-2-carboxylate (750 mg, 92.43%) as a white solid. [000517] LC-MS (ES, m/z): [M +H]+:192.00 [000518] 1H-NMR: (400 MHz, Chloroform-d) δ 9.04 (s, 1H), 7.53 (d, J = 3.7 Hz, 1H), 6.78 (d, J = 3.7 Hz, 1H), 3.99 (s, 3H), 3.88 (s, 3H). [000519] Example 1.51. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 7-methoxy-3,4-dihydroisoquinoline-2(1H)-carboxamide (I-139)
[000520] To a stirred solution of phenyl N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5- yl]carbamate (150 mg, 0.395 mmol, 1 equiv) and 7-methoxy-1,2,3,4-tetrahydroisoquinoline (96.80 mg, 0.593 mmol, 1.5 equiv) in DMSO (2 mL) was added DIEA (153.31 mg, 1.185 mmol, 3 equiv) dropwise at room temperature under argon atmosphere. The resulting mixture was stirred for overnight at room temperature. Desired product could be detected by LCMS. The crude product (150 mg) was purified by Prep-HPLC with the following conditions(Column: Xselect CSH C18 OBD Column 30*150mm 5μm, n; Mobile Phase A: Water(0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 17% B to 37% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 10.32) to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo- 3H-isoindol-5-yl]-7-methoxy-3,4-dihydro-1H-isoquinoline-2-carboxamide (78.3 mg, 43.54%) as a white solid. [000521] LC-MS (ES, m/z): [M+H]+: 449.17
[000522] 1H NMR: (400 MHz, DMSO-d6) δ 10.96 (s, 1H), 8.93 (s, 1H), 7.83 – 7.78 (m, 1H), 7.63 – 7.53 (m, 2H), 7.14 – 7.07 (m, 1H), 6.77 (d, J = 7.0 Hz, 2H), 5.07 (dd, J = 13.3, 5.1 Hz, 1H), 4.64 (s, 2H), 4.40 (d, J = 17.1 Hz, 1H), 4.25 (d, J = 17.1 Hz, 1H), 3.72 (d, J = 10.2 Hz, 5H), 2.91 (ddd, J = 17.2, 13.6, 5.4 Hz, 1H), 2.79 (t, J = 5.9 Hz, 2H), 2.64 – 2.54 (m, 1H), 2.45 – 2.30 (m, 1H), 2.03 – 1.94 (m, 1H). [000523] Example 1.52. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 6-methoxy-3,4-dihydroisoquinoline-2(1H)-carboxamide (I-138)
[000524] To a stirred solution of phenyl N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5- yl]carbamate (150 mg, 0.395 mmol, 1 equiv) and 6-methoxy-1,2,3,4-tetrahydroisoquinoline (96.80 mg, 0.593 mmol, 1.5 equiv) in DMSO (2 mL) was added DIEA (153.31 mg, 1.185 mmol, 3 equiv) dropwise at room temperature under argon atmosphere. The resulting mixture was stirred for overnight at room temperature. Desired product could be detected by LCMS. The crude product (250mg) was purified by Prep-HPLC with the following conditions(Column: Sunfire prep C18 column, 30*150 mm, 5m; Mobile Phase A: Water(0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 20% B to40% B in 10 min; Wave Length: 254nm/220nm nm; RT (min): 11.22) to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H- isoindol-5-yl]-6-methoxy-3,4-dihydro-1H-isoquinoline-2-carboxamide (63.6 mg, 34.93%) as an off-white solid. [000525] LC-MS (ES, /z): [M+H]+:449.2 [000526] 1H NMR: (400 MHz, DMSO-d6) δ 10.96 (s, 1H), 8.91 (s, 1H), 7.81 (d, J = 1.6 Hz, 1H), 7.63 – 7.53 (m, 2H), 7.13 – 7.07 (m, 1H), 6.82 – 6.75 (m, 2H), 5.07 (dd, J = 13.3, 5.1 Hz, 1H), 4.59 (s, 2H), 4.39 (d, J = 17.1 Hz, 1H), 4.25 (d, J = 17.0 Hz, 1H), 3.71 (d, J = 13.8 Hz, 5H), 2.97 – 2.89 (m, 1H), 2.84 (t, J = 5.9 Hz, 2H), 2.59 (ddd, J = 17.3, 4.5, 2.3 Hz, 1H), 2.37 (qd, J = 13.2, 4.4 Hz, 1H), 1.98 (dtd, J = 12.5, 5.1, 2.1 Hz, 1H). [000527] Example 1.53. Synthesis of 1-cyclopropyl-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-1H-pyrazolo[3,4-d]pyrimidine-6-carboxamide (I-163)
[000528] Into a 40 mL vial were added 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6- dione (150 mg, 0.58 mmol, 1 equiv) and 1-cyclopropylpyrazolo[3,4-d]pyrimidine-6-carboxylic acid (118.14 mg, 0.58 mmol, 1 equiv) in DMF (5 mL) were added DIEA (448.66 mg, 3.47 mmol, 6 equiv) and HATU (329.98 mg, 0.87 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at 60°C under nitrogen atmosphere. Desired product could be detected by LCMS. The reaction was quenched with Water/Ice at room temperature. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water , 5% to 100% gradient in 30 min; detector, UV 254 nm. The crude product (120mg) was purified by Prep-HPLC with the following conditions (Column: Xselect CSH F-Phenyl OBD column 30*250 mm, 5μm; Mobile Phase A: Water(0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 10% B to 26% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 12.35) to afford 1- cyclopropyl-N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]pyrazolo[3,4-d]pyrimidine- 6-carboxamide (81.8 mg, 31.74%) as a white solid. [000529] LC-MS (ES, m/z): [M+H]+: 446.05 [000530] 1H-NMR: (400 MHz, DMSO-d6) δ 11.14 (s, 1H), 11.00 (s, 1H), 9.52 (s, 1H), 8.49 (s, 1H), 8.26 (d, J = 1.8 Hz, 1H), 7.99 (dd, J = 8.4, 1.9 Hz, 1H), 7.76 (d, J = 8.3 Hz, 1H), 5.13 (dd, J = 13.3, 5.1 Hz, 1H), 4.51 (d, J = 17.2 Hz, 1H), 4.37 (d, J = 17.2 Hz, 1H), 4.19 – 4.11 (m, 1H), 3.01 – 2.88 (m, 1H), 2.67 – 2.57 (m, 1H), 2.49 – 2.35 (m, 1H), 2.08 – 1.98 (m, 1H), 1.34 – 1.16 (m, 4H). [000531] Example 1.54. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 7H-pyrrolo[2,3-d]pyrimidine-2-carboxamide (I-137)
[000532] Into a 8mL vial ; To a stirred mixture of 3-(5-amino-1-oxo-3H-isoindol-2- yl)piperidine-2,6-dione (100 mg, 0.37mmol, 1 equiv) and 7H-pyrrolo[2,3-d]pyrimidine-2-
carboxylic acid (62.92 mg, 0.37 mmol, 1 equiv) in DMF (3 mL) were added DIEA (99.70 mg, 0.77 mmol, 2 equiv) and HATU (219.99 mg, 0.58 mmol, 1.5 equiv) in portions at room temperature under argon atmosphere. The resulting mixture was stirred for additional overnight at 60°C. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 5% to 100% gradient in 30 min; detector, UV 254 nm.to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-7H-pyrrolo[2,3- d]pyrimidine-2-carboxamide (5.4 mg, 3.46%) as a white solid. [000533] LC-MS (ES, m/z): [M+H]+:405.1 [000534] 1H-NMR: (400 MHz, DMSO-d6) δ 12.57 (s, 1H), 11.01 (d, J = 14.9 Hz, 2H), 9.21 (s, 1H), 8.29 (s, 1H), 7.99 (d, J = 8.2 Hz, 1H), 7.83 (s, 1H), 7.73 (d, J = 8.2 Hz, 1H), 6.77 (d, J = 3.4 Hz, 1H), 5.16 – 5.08 (m, 1H), 4.50 (d, J = 17.2 Hz, 1H), 4.35 (d, J = 17.3 Hz, 1H), 2.93 (s, 1H), 2.78 (s, 1H) 2.61 (d, J = 16.7 Hz, 1H), 2.41 (d, J = 13.1 Hz, 1H). [000535] Example 1.55. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5- yl)thiazolo[5,4-b]pyridine-5-carboxamide (I-162)
[000536] Into a 10 mL vial were added 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6- dione (70 mg, 0.270 mmol, 1 equiv), [1,3]thiazolo[5,4-b]pyridine-5-carboxylic acid (48.65 mg, 0.270 mmol, 1 equiv) and DMF (2 mL, 25.843 mmol, 95.72 equiv) at room temperature. To the above mixture was added DIEA (209.38 mg, 1.620 mmol, 6 equiv) and HATU (153.99 mg, 0.405 mmol, 1.5 equiv) in portions over 1 min at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo- 3H-isoindol-5-yl]-[1,3]thiazolo[5,4-b]pyridine-5-carboxamide (11.5 mg, 9.25%) as a brown solid.
[000537] LC-MS (ES, m/z): [M+H]+:421.90 [000538] 1H-NMR: (400 MHz, DMSO-d6) δ 11.12 (s, 1H), 10.99 (s, 1H), 9.79 (s, 1H), 8.72 (d, J = 8.4 Hz, 1H), 8.37 (d, J = 8.5 Hz, 1H), 8.29 (d, J = 1.8 Hz, 1H), 8.04 (dd, J = 8.4, 1.8 Hz, 1H), 7.74 (d, J = 8.3 Hz, 1H), 5.12 (dd, J = 13.3, 5.2 Hz, 1H), 4.50 (d, J = 17.1 Hz, 1H), 4.35 (d, J = 17.2 Hz, 1H), 2.93 (ddd, J = 17.7, 13.6, 5.3 Hz, 1H), 2.61 (d, J = 17.4 Hz, 1H), 2.44-2.37 (m, 1H), 2.10-1.98 (m, 1H). [000539] Example 1.56. Synthesis of 2-amino-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-1H-benzo[d]imidazole-5-carboxamide (I-135)
[000540] Synthesis of 2-((tert-butoxycarbonyl)amino)-1H-benzo[d]imidazole-5-carboxylic acid
[000541] Into a 40 mL vial were added 2-amino-1H-1,3-benzodiazole-5-carboxylic acid (250 mg, 1.411 mmol, 1 equiv), Boc2O (615.96 mg, 2.822 mmol, 2 equiv), EtOH (4 mL) and Guanidine hydrochloride (26.96 mg, 0.282 mmol, 0.2 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The reaction was quenched by the addition of Water (10 mL) at room temperature. The resulting mixture was extracted with EtOAc (2 x 20 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10mmol/L NH4HCO3), 5% to 50% gradient in 20 min;
detector, UV 254 nm. This resulted in 2-[(tert-butoxycarbonyl)amino]-1H-1,3-benzodiazole-5- carboxylic acid (105 mg, 26.84%) as a white solid. [000542] Synthesis of 2-amino-N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1H- benzo[d]imidazole-5-carboxamide
[000543] Into a 50 mL round-bottom flask were added 2-[(tert-butoxycarbonyl)amino]-1H-1,3- benzodiazole -5-carboxylic acid (105 mg, 0.379 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol- 2-yl)piperidine-2,6-dione (98.18 mg, 0.379 mmol, 1 equiv), DIEA (293.66 mg, 2.274 mmol, 6 equiv), DMF (4 mL) and HATU (215.98 mg, 0.569 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at 60°C under nitrogen atmosphere. The reaction was monitored by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH4HCO3), 5% to 50% gradient in 30 min; detector, UV 254 nm. The resulting mixture was concentrated under vacuum. The crude product (20 mg) was purified by Prep-HPLC with the following conditions (Column: XBridge Prep Phenyl OBD Column 19*250 mm, 5 m; Mobile Phase A: Water (0.05% TFA), Mobile Phase B: MeOH; Flow rate: 60 mL/min; Gradient: 20% B to 35% B in 10 min; Wave Length: 254nm/220 nm; RT1(min): 9.465) to afford 2-amino-N-[2- (2,6-dioxopiperidin-3-yl) -1-oxo-3H-isoindol-5-yl]-1H-1,3-benzodiazole-5-carboxamide (5.2 mg, 2.86%) as an off-white solid. [000544] LC-MS (ES, m/z): [M+H]+: 419.00 [000545] 1H NMR: (400 MHz, DMSO-d6) δ 12.74 (s, 1H), 10.99 (s, 1H), 10.60 (s, 1H), 8.68 – 8.46 (m, 2H), 8.15 (s, 1H), 7.95 (s, 1H), 7.90 (dd, J = 8.2, 1.6 Hz, 1H), 7.84 (d, J = 8.3 Hz, 1H), 7.73 (d, J = 8.4 Hz, 1H), 7.47 (d, J = 8.1 Hz, 1H), 5.11 (dd, J = 13.2, 5.1 Hz, 1H), 4.48 (d, J = 17.3 Hz, 1H), 4.34 (d, J = 17.3 Hz, 1H), 2.95 – 2.86 (m, 1H), 2.66 – 2.53 (m, 1H), 2.44 – 2.33 (m, 1H), 2.05 – 1.98 (m, 1H). [000546] Example 1.57. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-isopropyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxamide (I-143)
[000547] Into a 40 mL vial were added 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6- dione (100 mg, 0.386 mmol, 1 equiv), 1-isopropylpyrazolo[3,4-d]pyrimidine-6-carboxylic acid (79.53 mg, 0.386 mmol, 1 equiv), DIEA (299.11 mg, 2.316 mmol, 6 equiv), DMF (4 mL) and HATU (219.99 mg, 0.579 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 35% to 45% gradient in 10 min; detector, UV 254 nm. This resulted in N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1 - isopropylpyrazolo[3,4-d]pyrimidine-6-carboxamide (119.0 mg, 66.61%) as a white solid. [000548] LC-MS (ES, m/z): [M+H]+:448.05 [000549] 1H-NMR: (400 MHz, DMSO-d6) δ 11.13 (s, 1H), 11.00 (s, 1H), 9.53 (s, 1H), 8.55 (s, 1H), 8.26 (d, J = 1.8 Hz, 1H), 8.00 (dd, J = 8.3, 1.8 Hz, 1H), 7.76 (d, J = 8.3 Hz, 1H), 5.43 – 5.30 (m, 1H), 5.12 (dd, J = 13.2, 5.1 Hz, 1H), 4.51 (d, J = 17.3 Hz, 1H), 4.36 (d, J = 17.3 Hz, 1H), 3.00 – 2.86 (m, 1H), 2.66 – 2.57 (m, 1H), 2.49 – 2.34 (m, 1H), 2.10 – 1.98 (m, 1H), 1.56 (d, J = 6.7 Hz, 6H). [000550] Example 1.58. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 2-hydroxybenzo[d]thiazole-6-carboxamide (I-159)
[000551] Synthesis of 3-(tert-butoxycarbonyl)-2-oxo-2,3-dihydrobenzo[d]thiazole-6-carboxylic acid
[000552] Into a 25mL round-bottom flask were added 2-hydroxy-1,3-benzothiazole-6- carboxylic acid (100 mg, 0.512 mmol, 1 equiv) in DMF (2 mL) and sodium hydride (60% in oil, 14.75 mg, 0.614 mmol, 1.2 equiv) at 0 ºC. The mixture was stirred for 2 h. Then Boc2O (167.72 mg, 0.768 mmol, 1.5 equiv) was added and the mixture was allowed to warm to RT and stirred for 16h. The reaction was quenched with water at 0 ºC. The mixture was neutralized to pH 7 with HCl. The resulting mixture was concentrated under reduced pressure and lyophilized. This resulted in 3-(tert-butoxycarbonyl)-2-oxo-1,3-benzothiazole-6-carboxylic acid (67 mg, crude) as a solid. [000553] LC-MS (ES, m/z): [M-Boc-H]-: 193.90 [000554] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-2- hydroxybenzo[d]thiazole-6-carboxamide
[000555] Into a 25mL round-bottom flask were added 3-(tert-butoxycarbonyl)-2-oxo-1,3- benzothiazole-6-carboxylic acid (67 mg, 0.227 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol- 2-yl)piperidine-2,6-dione (58.82 mg, 0.227 mmol, 1 equiv), DMF (3 mL), DIEA (87.97 mg, 0.681 mmol, 3 equiv) and HATU (129.40 mg, 0.341 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for 16h at 60°C under nitrogen atmosphere. The mixture was purified by Prep-HPLC with the following conditions (Column: Sunfire prep C18 column, 30*150 mm, 5m; Mobile Phase A: Water(0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 7% B to25% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 13.03) to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-2-hydroxy- 1,3-benzothiazole-6-carboxamide (18.9 mg, 17.42%) as an off-white solid. [000556] LC-MS (ES, m/z): [M-H]-: 434.95
[000557] 1H-NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.49 (s, 1H), 8.20 (d, J = 1.8 Hz, 1H), 8.14 (d, J = 1.8 Hz, 1H), 7.90 (dd, J = 8.3, 1.9 Hz, 1H), 7.82 (dd, J = 8.4, 1.8 Hz, 1H), 7.71 (d, J = 8.3 Hz, 1H), 7.23 (d, J = 8.4 Hz, 1H), 5.10 (dd, J = 13.3, 5.1 Hz, 1H), 4.47 (d, J = 17.2 Hz, 1H), 4.33 (d, J = 17.2 Hz, 1H), 2.92 (ddd, J = 16.9, 13.5, 5.3 Hz, 1H), 2.65 – 2.56 (m, 1H), 2.39 (qd, J = 13.5, 4.8 Hz, 1H), 2.05 – 1.97 (m, 1H). [000558] Example 1.59. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1,4-dimethyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-136)
[000559] Synthesis of methyl 1,4-dimethyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate
[000560] Into a 25 mL round-bottom flask were added methyl 4-methyl-1H-pyrrolo[2,3- b]pyridine-5-carboxylate (45 mg, 0.237 mmol, 1 equiv), DMF (16 mL) and K2CO3 (49.05 mg, 0.355 mmol, 1.5 equiv) at room temperature. To the above mixture was added MeI (33.58 mg, 0.237 mmol, 1 equiv) and DMF (16 mL) in portions at room temperature. The resulting mixture was stirred for additional overnight at room temperature. Desired product could be detected by LCMS. The residue was purified by reverse flash chromatography with the following conditions: column, silica gel; mobile phase, MeCN in water, 5% to 95% gradient in 30 min; detector, UV 254 nm. This resulted in methyl 1,4-dimethylpyrrolo[2,3-b]pyridine-5-carboxylate (270 mg) as a brown solid. [000561] Synthesis of 1,4-dimethyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000562] Into a 50 mL round-bottom flask were added methyl 1,4-dimethylpyrrolo[2,3- b]pyridine-5-carboxylate (140 mg, 0.686 mmol, 1 equiv) and THF (1.5 mL) at room temperature. To the above mixture was added LiOH (131.34 mg, 5.488 mmol, 8 equiv) and H2O (1.5 mL) at room temperature. The resulting mixture was stirred for overnight at room temperature. The reaction was monitored by LCMS. The mixture was acidified to pH 5 with HCl (aq.). The resulting mixture was concentrated under vacuum and lyophilized This resulted in 1,4- dimethylpyrrolo[2,3-b]pyridine-5-carboxylic acid (400 mg, containing LiCl) as a white solid. [000563] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1,4-dimethyl-1H- pyrrolo[2,3-b]pyridine-5-carboxamide
[000564] Into a 25 mL round-bottom flask were added 1,4-dimethylpyrrolo[2,3-b]pyridine-5- carboxylic acid (80 mg, 0.421 mmol, 1 equiv) , DMF (4 mL) , DIEA (0.44 mL, 2.526 mmol, 6 equiv) and HATU (239.89 mg, 0.631 mmol, 1.5 equiv) at 0°C. To the above mixture was added 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6-dione (218.10 mg, 0.842 mmol, 2 equiv) at 0°C. The resulting mixture was stirred for 3 h at 90°C. The reaction was monitored by LCMS. The resulting was purified by Prep-HPLC with the following conditions (Column: XSelect CSH Fluoro Phenyl 30*150 mm, 5m; Mobile Phase A: Water(0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 13% B to28% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 13.62) to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1,4- dimethylpyrrolo[2,3-b]pyridine-5-carboxamide (11.9 mg, 6.40%) as an off-white solid. [000565] LC-MS (ES, m/z): [M+H]+ 432 [000566] 1H NMR: (400 MHz, DMSO-d6) δ 10.98 (s, 1H), 10.67 (s, 1H), 8.44 (s, 1H), 8.16 (d, J = 1.7 Hz, 1H), 7.81 – 7.74 (m, 1H), 7.71 (d, J = 8.3 Hz, 1H), 7.59 (d, J = 3.5 Hz, 1H), 6.67 (d, J = 3.5 Hz, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 4.47 (d, J = 17.3 Hz, 1H), 4.33 (d, J = 17.3 Hz,
1H), 3.85 (s, 3H), 2.92 (ddd, J = 17.8, 13.4, 5.3 Hz, 1H), 2.64 (s, 3H), 2.59 (s, 0H), 2.44 – 2.33 (m, 1H), 2.01 (d, J = 12.8 Hz, 1H). [000567] Example 1.60. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 4-fluoro-1-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-131)
[000568] Synthesis of methyl 4-fluoro-1-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate
[000569] To a stirred mixture of methyl 4-fluoro-1H-pyrrolo[2,3-b]pyridine-5-carboxylate (150 mg, 0.773 mmol, 1 equiv) and K2CO3 (160.15 mg, 1.159 mmol, 1.5 equiv) in DMF (6 mL) was added MeI (109.65 mg, 0.773 mmol, 1 equiv) dropwise at room temperature under argon atmosphere. The resulting mixture was stirred for overnight at room temperature. Desired product could be detected by LCMS. The reaction was quenched with Water(50 ml) at room temperature. The resulting mixture was extracted with EtOAc (3 x50 mL). The combined organic layers were washed with brine (2x50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was dissolved in DMF (5 mL). The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water , 5% to 80% gradient in 30 min; detector, UV 254 nm. This resulted in methyl 4-fluoro-1-methylpyrrolo[2,3-b]pyridine-5-carboxylate (100 mg, 62.17%) as a yellow solid. [000570] LC-MS (ES, m/z): [M +H]+: 209.00
[000571] 1H-NMR: (400 MHz, DMSO-d6) δ 8.75 (d, J = 9.4 Hz, 1H), 7.68 (d, J = 3.5 Hz, 1H), 6.71 (d, J = 3.5 Hz, 1H), 3.88 (d, J = 5.9 Hz, 6H). [000572] Synthesis of 4-fluoro-1-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000573] A mixture of methyl 4-fluoro-1-methylpyrrolo[2,3-b]pyridine-5-carboxylate (160 mg, 0.769 mmol, 1 equiv) and NaOH (61.48 mg, 1.538 mmol, 2 equiv) in THF (8 mL) and H2O (8 mL) was stirred for overnight at room temperature under argon atmosphere. Desired product could be detected by LCMS. The mixture was acidified to pH 4 with 1N HCl. The resulting mixture was concentrated under reduced pressure and was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in H2O, 5 to 80% gradient in 30 min; detector, UV 254 nm. This resulted in 4-fluoro-1- methylpyrrolo[2,3-b]pyridine-5-carboxylic acid (160 mg, 57.19%) as a white solid. The crude product mixture was used in the next step directly without further purification. [000574] LC-MS (ES, m/z): [M +H]+: 195.00 [000575] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-4-fluoro-1-methyl- 1H-pyrrolo[2,3-b]pyridine-5-carboxamide
[000576] A mixture of 4-fluoro-1-methylpyrrolo[2,3-b]pyridine-5-carboxylic acid (100 mg, 0.515 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6-dione (267.06 mg, 1.030 mmol, 2 equiv) and DIEA (199.70 mg, 1.545 mmol, 3 equiv), HATU (380.235 mg, 0.773 mmol, 1.5 equiv) in DMF (7.5 mL) was stirred overnight at 40°C under argon atmosphere. The reaction was monitored by LCMS. The resulting mixture was dissolved in DMF (5 mL) and purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water , 5% to 70% gradient in 30 min; detector, UV 254 nm
to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-4-fluoro-1-methylpyrrolo[2,3- b]pyridine-5-carboxamide as a white solid. [000577] LC-MS (ES, m/z): [M +H]+:436.00 [000578] 1H-NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.77 (s, 1H), 8.57 (d, J = 9.3 Hz, 1H), 8.11 (d, J = 1.6 Hz, 1H), 7.80 – 7.67 (m, 3H), 6.72 (d, J = 3.5 Hz, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 4.48 (d, J = 17.3 Hz, 1H), 4.34 (d, J = 17.3 Hz, 1H), 3.89 (s, 3H), 2.89 (s, 1H), 2.67 – 2.58 (m, 1H), 2.44 – 2.32 (m, 1H), 2.06 – 1.96 (m, 1H). [000579] Example 1.61. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1,3-dimethyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-142)
[000580] To a stirred mixture of 1,3-dimethylpyrrolo[2,3-b]pyridine-5-carboxylic acid (80 mg, 0.421 mmol, 1 equiv) and HATU (319.86 mg, 0.842 mmol, 2 equiv) in DMF (4 mL) were added DIEA (326.17 mg, 2.526 mmol, 6 equiv). The resulting mixture was stirred for 20 min at room temperature under argon atmosphere. To the above mixture was added 3-(5- amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6-dione (109.05 mg, 0.421 mmol, 1 equiv) in portions at room temperature. The resulting mixture was stirred for overnight at room temperature. Desired product could be detected by LCMS. The residue was purified by reversed- phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water , 10% to 50% gradient in 20 min; detector, UV 254 nm.This resulted in N-[2- (2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1,3-dimethylpyrrolo[2,3-b]pyridine-5- carboxamide (32.2 mg, 16.91%) as an off-white solid. [000581] LC-MS (ES, m/z): [M+H]+: 432.16 [000582] 1H NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.59 (s, 1H), 8.87 (d, J = 2.1 Hz, 1H), 8.59 (d, J = 2.1 Hz, 1H), 8.17 (d, J = 1.8 Hz, 1H), 7.87 (dd, J = 8.3, 1.8 Hz, 1H), 7.73 (d, J = 8.3 Hz, 1H), 7.41 (d, J = 1.3 Hz, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 4.49 (d, J = 17.2 Hz, 1H), 4.34 (d, J = 17.2 Hz, 1H), 3.82 (s, 3H), 2.93 (ddd, J = 17.8, 13.5, 5.3 Hz, 1H), 2.66 – 2.57 (m, 1H), 2.41 (dd, J = 13.1, 4.5 Hz, 1H), 2.06 – 1.98 (m, 1H).
[000583] Example 1.62. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-methyl-3-(trifluoromethyl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-133)
[000584] Synthesis of 5-bromo-1-methyl-3-(trifluoromethyl)-1H-pyrrolo[2,3-b]pyridine
[000585] Into a 100 mL 3-necked round-bottom flask were added 5-bromo-3-(trifluoromethyl)- 1H-pyrrolo[2,3-b]pyridine (900 mg, 3.396 mmol, 1 equiv) and K2CO3 (938.63 mg, 6.792 mmol, 2 equiv), MeI (723.00 mg, 5.094 mmol, 1.50 equiv) in DMF(10 mL) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for overnight at 25°C under argon atmosphere. The reaction was monitored by LCMS. The reaction was quenched by the addition of Water (50 mL) at room temperature. The mixture was extracted with EtOAc (3x50mL) and the combined organic layers were dried with Na2SO4, filtered, and concentrated under vacuum. The resulting mixture was dissolved in DMF (5 mL). The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 10% to 95% gradient in 25 min; detector, UV 254 nm to afford 5-bromo-1-methyl-3- (trifluoromethyl)pyrrolo[2,3-b]pyridine (790 mg, 82.53%) as a white solid. [000586] LC-MS (ES, m/z): [M +H]+: 279.00 [000587] 1H-NMR: (400 MHz, Chloroform-d) δ 8.38 (d, J = 2.1 Hz, 1H), 8.09 (dd, J = 2.1, 1.0 Hz, 1H), 7.48 (d, J = 1.4 Hz, 1H), 3.85 (s, 3H).
[000588] Synthesis of methyl 1-methyl-3-(trifluoromethyl)-1H-pyrrolo[2,3-b]pyridine-5- carboxylate
[000589] Into a 30 mL pressure tank reactor were added 5-bromo-1-methyl-3- (trifluoromethyl)pyrrolo[2,3-b]pyridine (690 mg, 2.473 mmol, 1 equiv) and Pd(dppf)Cl2 (904.61 mg, 1.236 mmol, 0.50 equiv), TEA (750.63 mg, 7.419 mmol, 3 equiv) in MeOH(10 mL). The resulting mixture was stirred for 2 days at 120°C under carbon monoxide atmosphere 10 atm. The reaction was monitored by LCMS. The mixture were collected by filtration and washed with MeOH (10 mL)). The resulting mixture was concentrated under vacuum and dissolved in DMF(4 mL). The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water , 10% to 85% gradient in 35 min; detector, UV 254 nm to afford methyl 1-methyl-3-(trifluoromethyl)pyrrolo[2,3-b]pyridine- 5-carboxylate as a white solid. [000590] LC-MS (ES, m/z): [M +H]+: 259.00 [000591] Synthesis of 1-methyl-3-(trifluoromethyl)-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000592] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1-methyl-3- (trifluoromethyl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide
[000593] A mixture of 1-methyl-3-(trifluoromethyl)-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid (85 mg, crude), 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6-dione (74.33 mg, 0.287 mmol, 1 equiv) and DIEA (222.32 mg, 1.722 mmol, 6 equiv), HATU (120 mg, 0.316 mmol, 1.1 equiv) in DMF (4 mL) was stirred 2 hours at 50°C under argon atmosphere. The reaction was monitored by LCMS. The residue was dissolved in DMF (10 mL).The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water , 10% to 80% gradient in 25 min; detector, UV 254 nm, The crude product was purified by Prep-HPLC with the following conditions (Column: XSelect CSH Fluoro Phenyl 30*150 mm, 5m; Mobile Phase A: Water(0.1%FA), Mobile Phase B: MeOH-- HPLC; Flow rate: 60 mL/min mL/min; Gradient: 40% B to60% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 12.37) to afford to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo- 3H-isoindol-5-yl]-1-methyl-3-(trifluoromethyl)pyrrolo[2,3-b]pyridine-5-carboxamide (18.8 mg, 12.9%) as a white solid. [000594] LC-MS (ES, m/z): [M -H]-: 484.10 [000595] 1H-NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.77 (s, 1H), 9.05 (d, J = 2.1 Hz, 1H), 8.70 (d, J = 2.0 Hz, 1H), 8.43 – 8.39 (m, 1H), 8.15 (d, J = 1.7 Hz, 1H), 7.87 (dd, J = 8.4, 1.9 Hz, 1H), 7.74 (d, J = 8.3 Hz, 1H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.49 (d, J = 17.2 Hz, 1H), 4.35 (d, J = 17.3 Hz, 1H), 3.95 (s, 3H), 3.00 – 2.86 (m, 1H), 2.61 (ddd, J = 16.9, 4.3, 2.3 Hz, 1H), 2.40 (tt, J = 13.2, 6.5 Hz, 1H), 2.10 – 1.97 (m, 1H). [000596] Example 1.63. Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-1H-indazole-5-carboxamide (I-124)
[000597] Synthesis of methyl 1-cyclobutyl-1H-indazole-5-carboxylate
[000598] Into a 40 mL vial were added methyl 1H-indazole-5-carboxylate (500 mg, 2.838 mmol, 1 equiv), Cs2CO3 (1849.41 mg, 5.676 mmol, 2 equiv), DMF (5 mL) and iodocyclobutane (568.20 mg, 3.122 mmol, 1.1 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The reaction involves filtration, washing the filter cake with DMF (2x3mL). The filtrate was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH4HCO3), 75% to 85% gradient in 10 min; detector, UV 254 nm. This resulted in methyl 1-cyclobutylindazole-5-carboxylate (300 mg, 45.91%) as an off-white solid. [000599] LC-MS (ES, m/z): [M+H]+: 231.10 [000600] 1H-NMR: (400 MHz, DMSO-d6) δ 8.48 (s, 1H), 8.32 (s, 1H), 7.94 (dd, J = 8.9, 1.5 Hz, 1H), 7.77 (d, J = 8.9 Hz, 1H), 5.38 – 5.26 (m, 1H), 3.88 (s, 3H), 2.72 – 2.57 (m, 2H), 2.51 – 2.42 (m, 2H), 1.95 – 1.82 (m, 2H). [000601] Synthesis of 1-cyclobutyl-1H-indazole-5-carboxylic acid
[000602] Into a 50 mL round-bottom flask were added methyl 1-cyclobutylindazole-5- carboxylate (150 mg, 0.651 mmol, 1 equiv), THF (3 mL), H2O (3 mL) and LiOH.H2O (32.80
mg, 0.781 mmol, 1.2 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The mixture was neutralized to pH = 7 with Cationic resin. T The resulting mixture was concentrated under reduced pressure and was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in H2O, 5 to 80% gradient in 30 min; detector, UV 254 nm. This resulted in 1-cyclobutylindazole-5-carboxylic acid (120 mg, 85.19%) as an off-white solid. [000603] LC-MS (ES, m/z): [M+H]+: 217.10 [000604] 1H-NMR: (400 MHz, DMSO-d6) δ 8.39 (s, 1H), 8.22 (s, 1H), 7.96 (dd, J = 8.9, 1.5 Hz, 1H), 7.65 (d, J = 8.8 Hz, 1H), 5.35 – 5.22 (m, 1H), 2.72 – 2.56 (m, 2H), 2.50 – 2.40 (m, 2H), 1.96 – 1.80 (m, 2H). [000605] Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1H- indazole-5-carboxamide
[000606] Into a 50 mL round-bottom flask were added 1-cyclobutylindazole-5-carboxylic acid (140 mg, 0.647 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6-dione (167.86 mg, 0.647 mmol, 1 equiv), DIEA (502.07 mg, 3.882 mmol, 6 equiv), DMF (7 mL) and HATU (369.26 mg, 0.971 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at 60 C under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 45% to 55% gradient in 10 min; detector, UV 254 nm. This resulted in 1-cyclobutyl-N-[2-(2,6- dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]indazole-5-carbox amide (104.9 mg, 35.03%) as a white solid. [000607] LC-MS (ES, m/z): [M+H]+: 458.05 [000608] 1H-NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.58 (s, 1H), 8.51 (d, J = 1.6 Hz, 1H), 8.33 (s, 1H), 8.18 (d, J = 1.7 Hz, 1H), 8.00 (dd, J = 8.9, 1.7 Hz, 1H), 7.87 (dd, J = 8.4, 1.8
Hz, 1H), 7.82 (d, J = 8.9 Hz, 1H), 7.72 (d, J = 8.3 Hz, 1H), 5.41 – 5.28 (m, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 4.48 (d, J = 17.2 Hz, 1H), 4.34 (d, J = 17.2 Hz, 1H), 3.00 – 2.86 (m, 1H), 2.73 – 2.56 (m, 3H), 2.54 – 2.51 (m, 1H), 2.49 – 2.32 (m, 2H), 2.07 – 1.97 (m, 1H), 1.96 – 1.83 (m, 2H). [000609] Example 1.64. Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-3-methyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxamide (I-128)
[000610] Synthesis of 6-chloro-1-cyclobutyl-3-methyl-1H-pyrazolo[3,4-d]pyrimidine
[000611] To a stirred mixture of 6-chloro-3-methyl-1H-pyrazolo[3,4-d]pyrimidine (500 mg, 2.966 mmol, 1 equiv) and cyclobutanol (641.60 mg, 8.898 mmol, 3 equiv) and PPh3 (1166.92 mg, 4.449 mmol, 1.5 equiv) in THF (6 mL) was added DIAD (899.62 mg, 4.449 mmol, 1.5 equiv) dropwise at 0°C under argon atmosphere. The resulting mixture was stirred for overnight at 60 °C under argon atmosphere. The reaction was monitored by LCMS. The reaction was quenched with Water at room temperature. The resulting mixture was extracted with EtOAc (3 x 10mL). The combined organic layers were washed with brine (1x20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1:1) to afford 6-chloro-1- cyclobutyl-3-methylpyrazolo[3,4-d]pyrimidine as an off-white solid. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel;
mobile phase, MeCN in Water , 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 6-chloro-1-cyclobutyl-3-methylpyrazolo[3,4-d]pyrimidine (360 mg, 54.51%) as a yellow solid. [000612] LC-MS (ES, m/z): [M+H]+:223 [000613] 1H NMR: (400 MHz, Methanol-d4) δ 9.05 (s, 1H), 5.40 – 5.27 (m, 1H), 2.86 – 2.70 (m, 2H), 2.62 (s, 3H), 2.58 – 2.26 (m, 2H), 2.06 – 1.83 (m, 2H). [000614] Synthesis of methyl 1-cyclobutyl-3-methyl-1H-pyrazolo[3,4-d]pyrimidine-6- carboxylate
[000615] To a stirred mixture of 6-chloro-1-cyclobutyl-3-methylpyrazolo[3,4-d]pyrimidine (310 mg, 1.392 mmol, 1 equiv) and Pd(dppf)Cl2 (203.73 mg, 0.278 mmol, 0.2 equiv) in MeOH (14 mL) was added TEA (0.58 mL, 4.176 mmol, 3 equiv). The resulting mixture was stirred for overnight at 120°C under carbon monoxide atmosphere 5 atm. The reaction was monitored by LCMS. The resulting mixture was filtered, the filter cake was washed with MeOH (3x20 mL). The filtrate was concentrated under reduced pressure. The residue was purified by reversed- phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 10% to 95% gradient in 10 min; detector, UV 254 nm. This resulted in methyl 1-cyclobutyl-3-methylpyrazolo[3,4-d]pyrimidine-6-carboxylate (300 mg) as a yellow solid. [000616] LC-MS (ES, m/z): [M+H]+:247 [000617] 1H NMR: (300 MHz, DMSO-d6) δ 9.43 (s, 1H), 5.41 (q, J = 8.4 Hz, 1H), 3.94 (s, 3H), 2.75 – 2.65 (m, 2H), 2.65 (s, 3H), 2.48 – 2.38 (m, 2H), 1.89 (td, J = 9.9, 5.6 Hz, 2H). [000618] Synthesis of 1-cyclobutyl-3-methyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxylic acid
[000619] To a stirred solution of methyl 1-cyclobutyl-3-methylpyrazolo[3,4-d]pyrimidine-6- carboxylate (300 mg, 1.218 mmol, 1 equiv) in H2O (0.02 mL)and THF (0.10 mL) was added NaOH (97.45 mg, 2.436 mmol, 2.00 equiv) in portions at room temperature under argon atmosphere. The resulting mixture was stirred for overnight at room temperature under argon atmosphere. The reaction was monitored by LCMS. The resulting mixture was concentrated under reduced pressure and lyophilized. The crude resulting mixture was used in the next step directly without further purification. [000620] LC-MS (ES, m/z): [M+H]+:233 [000621] Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-3- methyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxamide
[000622] To a stirred solution of 1-cyclobutyl-3-methylpyrazolo[3,4-d]pyrimidine-6-carboxylic acid (47 mg, 0.202 mmol, 1 equiv) and 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6-dione (52.47 mg, 0.202 mmol, 1 equiv) in DMF (1.5 mL) were added HATU (115.42 mg, 0.303 mmol, 1.5 equiv) and DIEA (0.11 mL, 0.606 mmol, 3 equiv) in portions at room temperature under argon atmosphere. The resulting mixture was stirred for overnight at 30 °C under argon atmosphere. The reaction was monitored by LCMS. The residue was dissolved in DMSO (2mL). The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 20% to 70% gradient in 13 min; detector, UV 254 nm. This resulted in 1-cyclobutyl-N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5- yl]-3-methylpyrazolo[3,4-d]pyrimidine-6-carboxamide (34.8 mg, 32.79%) as a yellow solid. [000623] LC-MS (ES, m/z): [M+H]+:474 [000624] 1H NMR: (400 MHz, DMSO-d6) δ 11.11 (s, 1H), 10.99 (s, 1H), 9.50 (s, 1H), 8.25 (s, 1H), 8.00 (dd, J = 8.3, 1.8 Hz, 1H), 7.76 (d, J = 8.3 Hz, 1H), 5.57 (p, J = 8.4 Hz, 1H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.51 (d, J = 17.2 Hz, 1H), 4.36 (d, J = 17.2 Hz, 1H), 2.93 (ddd, J = 18.2, 9.6, 3.8 Hz, 1H), 2.82 – 2.56 (m, 6H), 2.56 – 2.36 (m, 3H), 2.08 – 1.82 (m, 3H).
[000625] Example 1.65. Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-3-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-129)
[000626] Synthesis of methyl 1-cyclobutyl-3-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate
[000627] Into a 40 mL vial were added 3-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid (200 mg, 1.135 mmol, 1 equiv), Cs2CO3 (685.21 mg, 2.104 mmol, 2 equiv), DMF (5 mL) and iodocyclobutane (229.66 mg, 1.262 mmol, 1.2 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10mmol/L NH4HCO3), 60% to 100% gradient in 10 min; detector, UV 254 nm. This resulted in 1-cyclobutyl-3-methylpyrrolo[2,3-b]pyridine -5- carboxylic acid (190 mg, 72.68%) as an off-white solid. [000628] LC-MS (ES, m/z): [M+H]+: 245.05 [000629] 1H-NMR: (300 MHz, DMSO-d6) δ 8.80 (d, J = 2.0 Hz, 1H), 8.46 (d, J = 2.0 Hz, 1H), 7.72 (d, J = 1.3 Hz, 1H), 5.40 – 5.22 (m, 1H), 3.89 (s, 3H), 2.60 – 2.52 (m, 1H), 2.49 – 2.34 (m, 3H), 2.31 (d, J = 1.1 Hz, 3H), 1.92 – 1.75 (m, 2H). [000630] Synthesis of 1-cyclobutyl-3-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000631] Into a 40 mL vial were added methyl 1-cyclobutyl-3-methylpyrrolo[2,3-b]pyridine-5- carboxylate (190 mg, 0.778 mmol, 1 equiv), THF (4 mL), H2O (4 mL) and LiOH (27.94 mg, 1.167 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The mixture was neutralized to pH = 7 with HCl (aq.). The resulting mixture was filtered, the filter cake was washed with EtOAc (2 x 10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (0:1) to afford 1-cyclobutyl-3-methylpyrrolo[2,3 - b]pyridine-5-carboxylic acid (120 mg, 67.01%) as an off-white solid. [000632] LC-MS (ES, m/z): [M+H]+: 231.10 [000633] 1H-NMR: (400 MHz, DMSO-d6) δ 12.83 (s, 1H), 8.78 (d, J = 2.0 Hz, 1H), 8.43 (d, J = 2.0 Hz, 1H), 7.69 (d, J = 1.3 Hz, 1H), 5.37 – 5.24 (m, 1H), 2.54 (dd, J = 9.4, 2.7 Hz, 1H), 2.49 – 2.45 (m, 1H), 2.44 – 2.35 (m, 2H), 2.30 (d, J = 1.1 Hz, 3H), 1.89 – 1.76 (m, 2H). [000634] Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-3- methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide
[000635] Into a 40 mL vial were added 1-cyclobutyl-3-methylpyrrolo[2,3-b]pyridine-5- carboxylic acid (80 mg, 0.347 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine- 2,6-dione (90.07 mg, 0.347 mmol, 1 equiv), DIEA (269.42 mg, 2.082 mmol, 6 equiv), DMF (5 mL) and HATU (198.15 mg, 0.520 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel;
mobile phase, MeCN in Water (0.1% FA), 50% to 70% gradient in 10 min; detector, UV 254 nm. This resulted in 1-cyclobutyl-N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-3- methylpyrrolo [2,3-b]pyridine-5-carboxamide (28.4 mg, 17.28%) as a white solid. [000636] LC-MS (ES, m/z): [M+H]+: 472.10 [000637] 1H-NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.58 (s, 1H), 8.85 (d, J = 2.1 Hz, 1H), 8.57 (d, J = 2.1 Hz, 1H), 8.17 (d, J = 1.8 Hz, 1H), 7.87 (dd, J = 8.3, 1.8 Hz, 1H), 7.76 – 7.69 (m, 2H), 5.39 – 5.26 (m, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 4.49 (d, J = 17.2 Hz, 1H), 4.34 (d, J = 17.3 Hz, 1H), 2.99 – 2.86 (m, 1H), 2.66 – 2.52 (m, 3H), 2.47 – 2.37 (m, 3H), 2.36 (d, J = 1.1 Hz, 3H), 2.07 – 1.96 (m, 1H), 1.91 – 1.79 (m, 2H). [000638] Example 1.66. Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-1H-pyrazolo[3,4-b]pyrazine-5-carboxamide (I-130)
[000639] Synthesis of methyl 1H-pyrazolo[3,4-b]pyrazine-5-carboxylate
[000640] Into a 30 mL pressure tank reactor were added 6-chloro-1-cyclobutylpyrazolo[3,4- d]pyrimidine (20 mg, 0.096 mmol, 1 equiv), Pd(dppf)Cl2 (14.71 mg, 0.020 mmol, 0.02 equiv), TEA (203.39 mg, 2.010 mmol, 2 equiv) and MeOH (10 mL) at room temperature. The resulting mixture was stirred for overnight at 100°C under carbon monoxide (10 atm) atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column
chromatography, eluted with PE / EA (0:1) to afford methyl 1H-pyrazolo[3,4-b]pyrazine-5- carboxylate (8.5 mg, 47.48%) as a pink solid. [000641] LC-MS (ES, m/z): [M+H]+: 179.00 [000642] 1H-NMR: (400 MHz, DMSO-d6) δ 14.47 (s, 1H), 9.18 (s, 1H), 8.66 (s, 1H), 3.96 (s, 3H). [000643] Synthesis of methyl 1-cyclobutyl-1H-pyrazolo[3,4-b]pyrazine-5-carboxylate
[000644] Into a 20 mL vial were added methyl 1H-pyrazolo[3,4-b]pyrazine-5-carboxylate (75 mg, 0.421 mmol, 1 equiv), Cs2CO3 (274.33 mg, 0.842 mmol, 2 equiv), DMF (3 mL) and iodocyclobutane (114.93 mg, 0.631 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH4HCO3), 35% to 55% gradient in 20 min; detector, UV 254 nm. This resulted in methyl 1-cyclobutylpyrazolo[3,4-b]pyrazine-5- carboxylate (60 mg, 61.37%) as an off-white solid. [000645] LC-MS (ES, m/z): [M+H]+: 233.00 [000646] Synthesis of 1-cyclobutyl-1H-pyrazolo[3,4-b]pyrazine-5-carboxylic acid
[000647] Into a 20 mL vial were added methyl 1-cyclobutylpyrazolo[3,4-b]pyrazine-5- carboxylate (60 mg, 0.258 mmol, 1 equiv), THF (1.5 mL), H2O (1.5 mL) and LiOH.H2O (16.26 mg, 0.387 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The mixture was neutralized to pH = 7 with cationic resin.
The resulting mixture was filtered, the filter cake was washed with EtOAc (2 x 5 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (0:1) to afford 1-cyclobutylpyrazolo[3,4-b]pyrazine-5 - carboxylic acid (45 mg, 79.82%) as a white solid. [000648] LC-MS (ES, m/z): [M+H]+: 219.10 [000649] 1H-NMR: (400 MHz, DMSO-d6) δ 9.17 (s, 1H), 8.65 (s, 1H), 5.53 – 5.44 (m, 1H), 2.79 – 2.69 (m, 2H), 2.50 – 2.42 (m, 2H), 1.98 – 1.89 (m, 2H). [000650] Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1H- pyrazolo[3,4-b]pyrazine-5-carboxamide
[000651] Into a 40 mL vial were added 1-cyclobutylpyrazolo[3,4-b]pyrazine-5-carboxylic acid (40 mg, 0.183 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6-dione (47.52 mg, 0.183 mmol, 1 equiv), DIEA (142.15 mg, 1.098 mmol, 6 equiv), DMF (3 mL) and HATU (104.55 mg, 0.274 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The residue was purified by reversed- phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 45% to 55% gradient in 10 min; detector, UV 254 nm. This resulted in 1-cyclobutyl-N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]pyrazolo[3,4-b]pyrazine- 5-carboxamide (29.3 mg, 34.34%) as a white solid. [000652] LC-MS (ES, m/z): [M+H]+: 460.00 [000653] 1H-NMR: (300 MHz, DMSO-d6) δ 11.14 (s, 1H), 10.98 (s, 1H), 9.31 (s, 1H), 8.73 (s, 1H), 8.28 (s, 1H), 8.03 (dd, J = 8.3, 1.6 Hz, 1H), 7.74 (d, J = 8.3 Hz, 1H), 5.63 – 5.45 (m, 1H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.50 (d, J = 17.2 Hz, 1H), 4.35 (d, J = 17.4 Hz, 1H), 3.02 – 2.86 (m, 1H), 2.85 – 2.61 (m, 3H), 2.61 – 2.52 (m, 2H), 2.46 – 2.33 (m, 1H), 2.05 – 1.87 (m, 3H). [000654] Example 1.67. Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-1H-pyrazolo[3,4-b]pyridine-5-carboxamide (I-115)
[000655] Synthesis of methyl 1-cyclobutyl-1H-pyrazolo[3,4-b]pyridine-5-carboxylate
[000656] Into a 40 mL vial were added methyl 1H-pyrazolo[3,4-b]pyridine-5-carboxylate (300 mg, 1.693 mmol, 1 equiv), Cs2CO3 (827.59 mg, 2.540 mmol, 1.5 equiv), DMF (7 mL) and bromocyclobutane (228.61 mg, 1.693 mmol, 1 equiv) at room temperature. The resulting mixture was stirred for overnight at 80°C under nitrogen atmosphere. Desired product could be detected by LCMS. The reaction involves filtration, washing the filter cake with DMF (2x3mL). The filtrate was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH4HCO3), 65% to 75% gradient in 10 min; detector, UV 254 nm. This resulted in methyl 1-cyclobutylpyrazolo[3,4- b]pyridine-5-carboxylate (230 mg, 58.73%) as an off-white solid. [000657] LC-MS (ES, m/z): [M+H]+: 232.10 [000658] 1H-NMR: (400 MHz, DMSO-d6) δ 9.04 (d, J = 2.0 Hz, 1H), 8.82 (d, J = 2.0 Hz, 1H), 8.38 (s, 1H), 5.58 – 5.45 (m, 1H), 3.93 (s, 3H), 2.78 – 2.63 (m, 2H), 2.52 – 2.40 (m, 2H), 1.98 – 1.86 (m, 2H). [000659] Synthesis of 1-cyclobutyl-1H-pyrazolo[3,4-b]pyridine-5-carboxylic acid
[000660] Into a 50 mL round-bottom flask were added methyl 1-cyclobutylpyrazolo[3,4- b]pyridine-5 -carboxylate (200 mg, 0.865 mmol, 1 equiv), THF (4 mL), H2O (4 mL) and
LiOH.H2O (54.43 mg, 1.297 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The mixture was neutralized to pH = 7 with cationic resin. The resulting mixture was concentrated under reduced pressure and was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in H2O, 5 to 80% gradient in 30 min; detector, UV 254 nm. This resulted in 1-cyclobutylpyrazolo[3,4-b]pyridine-5-carboxylic acid (175 mg, 93.15%) as an off-white solid. [000661] LC-MS (ES, m/z): [M+H]+: 218.20 [000662] 1H-NMR: (400 MHz, DMSO-d6) δ 9.06 (d, J = 1.9 Hz, 1H), 8.71 (d, J = 1.9 Hz, 1H), 8.28 (s, 1H), 5.57 – 5.44 (m, 1H), 2.77 – 2.62 (m, 2H), 2.48 – 2.37 (m, 2H), 1.95 – 1.83 (m, 2H). [000663] Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1H- pyrazolo[3,4-b]pyridine-5-carboxamide
[000664] Into a 40 mL vial were added 1-cyclobutylpyrazolo[3,4-b]pyridine-5-carboxylic acid (170 mg, 0.783 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6-dione (202.90 mg, 0.783 mmol, 1 equiv), DIEA (606.88 mg, 4.698 mmol, 6 equiv), DMF (7 mL) and HATU (446.35 mg, 1.175 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at 60°C under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 45% to 55% gradient in 10 min; detector, UV 254 nm. This resulted in 1-cyclobutyl-N-[2-(2,6- dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]pyrazolo[3,4 -b]pyridine-5-carboxamide (55.5 mg, 15.10%) as a white solid. [000665] LC-MS (ES, m/z): [M+H]+: 459.05 [000666] 1H-NMR: (400 MHz, DMSO-d6) δ 11.00 (s, 1H), 10.75 (s, 1H), 9.11 (d, J = 2.1 Hz, 1H), 8.88 (d, J = 2.1 Hz, 1H), 8.42 (s, 1H), 8.17 (d, J = 1.7 Hz, 1H), 7.85 (dd, J = 8.3, 1.8 Hz,
1H), 7.74 (d, J = 8.3 Hz, 1H), 5.61 – 5.48 (m, 1H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.49 (d, J = 17.2 Hz, 1H), 4.35 (d, J = 17.2 Hz, 1H), 3.00 – 2.86 (m, 1H), 2.80 – 2.66 (m, 2H), 2.66 – 2.57 (m, 1H), 2.50 – 2.33 (m, 3H), 2.07 – 1.86 (m, 3H). [000667] Example 1.68. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-isopropyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-127)
[000668] Synthesis of methyl 1-isopropyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate
[000669] Into a 40 mL vial were added methyl 1H-pyrrolo[2,3-b]pyridine-5-carboxylate (500 mg, 2.838 mmol, 1 equiv), NaH (102.16 mg, 4.257 mmol, 1.5 equiv), DMF (5 mL) and 2- iodopropane (530.70 mg, 3.122 mmol, 1.1 equiv) at room temperature. The resulting mixture was stirred for 4 h at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 60% to 75% gradient in 10 min; detector, UV 254 nm. This resulted in methyl 1-isopropylpyrrolo[2,3- b]pyridine-5-carboxylate (320 mg, 51.66%) as a gray solid. [000670] LC-MS (ES, m/z): [M+H]+: 219.00 [000671] 1H-NMR: (400 MHz, DMSO-d6) δ 8.84 (d, J = 2.1 Hz, 1H), 8.55 (d, J = 2.1 Hz, 1H), 7.83 (d, J = 3.6 Hz, 1H), 6.67 (d, J = 3.6 Hz, 1H), 5.23 – 5.08 (m, 1H), 3.90 (s, 3H), 1.49 (d, J = 6.8 Hz, 6H). [000672] Synthesis of 1-isopropyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000673] Into a 50 mL round-bottom flask were added methyl 1-isopropylpyrrolo[2,3- b]pyridine-5-carboxylate (270 mg, 1.237 mmol, 1 equiv), THF (3 mL), H2O (3 mL) and LiOH.H2O (77.86 mg, 1.856 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS. The mixture was neutralized to pH = 7 with cationic resin. The resulting mixture was filtered, the filter cake was washed with MeOH (2 x 10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH4HCO3), 40% to 70% gradient in 10 min; detector, UV 254 nm. This resulted in 1-isopropylpyrrolo[2,3-b]pyridine-5-carboxylic acid (220 mg, 87.08%) as a white solid. [000674] LC-MS (ES, m/z): [M+H]+: 205.05 [000675] 1H-NMR: (400 MHz, DMSO-d6) δ 8.85 (d, J = 1.9 Hz, 1H), 8.44 (d, J = 1.9 Hz, 1H), 7.61 (d, J = 3.6 Hz, 1H), 6.48 (d, J = 3.6 Hz, 1H), 5.18 – 5.03 (m, 1H), 1.46 (d, J = 6.8 Hz, 6H). [000676] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1-isopropyl-1H- pyrrolo[2,3-b]pyridine-5-carboxamide
[000677] Into a 40 mL vial were added 1-isopropylpyrrolo[2,3-b]pyridine-5-carboxylic acid (100 mg, 0.490 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6-dione (126.95 mg, 0.490 mmol, 1 equiv), DIEA (379.71 mg, 2.940 mmol, 6 equiv), DMF (5 mL) and HATU (279.27 mg, 0.735 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the
following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 40% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in N-[2-(2,6-dioxopiperidin-3-yl)- 1-oxo-3H-isoindol-5-yl]-1-isopropylpyrrolo[2,3-b]pyridine-5-carboxamide (31.0 mg, 13.79%) as a white solid. [000678] LC-MS (ES, m/z): [M+H]+: 446.15 [000679] 1H-NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.60 (s, 1H), 8.87 (d, J = 2.2 Hz, 1H), 8.60 (d, J = 2.2 Hz, 1H), 8.18 (d, J = 1.7 Hz, 1H), 7.90 – 7.80 (m, 2H), 7.72 (d, J = 8.3 Hz, 1H), 6.68 (d, J = 3.6 Hz, 1H), 5.21 – 5.07 (m, 2H), 4.48 (d, J = 17.2 Hz, 1H), 4.34 (d, J = 17.2 Hz, 1H), 3.00 – 2.86 (m, 1H), 2.65 – 2.57 (m, 1H), 2.48 – 2.33 (m, 1H), 2.10 – 1.97 (m, 1H), 1.51 (d, J = 6.7 Hz, 6H). [000680] Example 1.69. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-isopropyl-3-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-126)
[000681] Synthesis of methyl 1-isopropyl-3-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate
[000682] To a stirred mixture of methyl 3-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate (200 mg, 1.052 mmol, 1 equiv) and Cs2CO3 (513.90 mg, 1.578 mmol, 1.5 equiv) in DMF (5 mL) was added 2-iodopropane (196.62 mg, 1.157 mmol, 1.1 equiv) dropwise at room temperature under argon atmosphere. The resulting mixture was stirred for overnight at 80°C. Desired product could be detected by LCMS. The reaction was quenched by the addition of
Water (50 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (3:1) to afford methyl 1- isopropyl-3-methylpyrrolo[2,3-b]pyridine-5-carboxylate (170 mg, 69.60%) as a yellow solid. [000683] LC-MS (ES, m/z): [M+H]+: 233.10 [000684] 1H-NMR: (400 MHz, DMSO-d6) δ 8.80 (d, J = 2.1 Hz, 1H), 8.46 (d, J = 2.0 Hz, 1H), 7.58 (d, J = 1.3 Hz, 1H), 5.15 – 5.03 (m, 1H), 3.88 (s, 3H), 2.31 (d, J = 1.1 Hz, 3H), 1.45 (d, J = 6.8 Hz, 6H). [000685] Synthesis of 1-isopropyl-3-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000686] To a stirred solution of methyl 1-isopropyl-3-methylpyrrolo[2,3-b]pyridine-5- carboxylate (150 mg, 0.646 mmol, 1 equiv) in THF (1 mL) was added NaOH (51.66 mg, 1.292 mmol, 2 equiv) in portions at room temperature under argon atmosphere. The resulting mixture was stirred for overnight at room temperature. Desired product could be detected by LCMS. The mixture was acidified to pH = 4 with HCl (aq.). The resulting mixture was concentrated under vacuum. This resulted in 1-isopropyl-3-methylpyrrolo[2,3-b]pyridine-5-carboxylic acid (100 mg, 70.95%) as a white solid. The crude product was used in the next step directly without further purification. [000687] LC-MS (ES, m/z): [M+H]+: 219.10 [000688] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1-isopropyl-3- methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide
[000689] To a stirred mixture of 1-isopropyl-3-methylpyrrolo[2,3-b]pyridine-5-carboxylic acid (50 mg, 0.229 mmol, 1 equiv) and HATU (130.66 mg, 0.344 mmol, 1.5 equiv) in DMF (2 mL) was added DIEA (0.12 mL, 0.687 mmol, 3 equiv) dropwise at room temperature under argon atmosphere. The mixture was stirred for 10 min at room temperature under argon atmosphere. To the above mixture was added 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6-dione (59.39 mg, 0.229 mmol, 1 equiv) in portions at room temperature. The resulting mixture was stirred for additional overnight at 60°C. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 20% to 95% gradient in 30 min; detector, UV 254 nm. This resulted in N-[2-(2,6-dioxopiperidin-3-yl)-1- oxo-3H-isoindol-5-yl]-1-isopropyl-3-methylpyrrolo[2,3-b]pyridine-5-carboxamide (12.8 mg, 11.56%) as a white solid. [000690] LC-MS (ES, m/z): [M-H]+: 458.15 [000691] 1H-NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.57 (s, 1H), 8.85 (d, J = 2.1 Hz, 1H), 8.57 (d, J = 2.2 Hz, 1H), 8.17 (d, J = 1.8 Hz, 1H), 7.87 (dd, J = 8.3, 1.8 Hz, 1H), 7.73 (d, J = 8.4 Hz, 1H), 7.58 (d, J = 1.3 Hz, 1H), 5.16 – 5.04 (m, 2H), 4.49 (d, J = 17.3 Hz, 1H), 4.34 (d, J = 17.3 Hz, 1H), 2.99 – 2.86 (m, 1H), 2.65 – 2.57 (m, 1H), 2.45 – 2.38 (m, 1H), 2.35 (d, J = 1.1 Hz, 3H), 2.06 – 1.98 (m, 1H), 1.47 (d, J = 6.7 Hz, 6H). [000692] Example 1.70. Synthesis of 4-(cyclobutylamino)-N-(2-(2,6-dioxopiperidin-3-yl)- 1-oxoisoindolin-5-yl)pyrimidine-2-carboxamide (I-125)
[000693] Synthesis of methyl 4-(cyclobutylamino)pyrimidine-2-carboxylate
[000694] Into a 50 mL round-bottom flask were added methyl 4-chloropyrimidine-2- carboxylate (450 mg, 2.608 mmol, 1 equiv), Et3N (527.75 mg, 5.216 mmol, 2 equiv) and DMF (13 mL) at room temperature. Then cyclobutylamine (278.19 mg, 3.912 mmol, 1.5 equiv) was added under nitrogen atmosphere. The resulting mixture was stirred for 2 h at room temperature under argon atmosphere. The reaction was monitored by LCMS. The residue was dissolved in DMF (5 mL) and directly purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 10% to 50% gradient in 25 min to afford methyl 4-(cyclobutylamino)pyrimidine-2-carboxylate (350 mg, 61.53%) as a white solid. [000695] LC-MS: (ES, m/z): [M+H]+: 208.00 [000696] Synthesis of 4-(cyclobutylamino)pyrimidine-2-carboxylic acid
[000697] Into a 50 mL round-bottom flask were added methyl 4-(cyclobutylamino)pyrimidine- 2-carboxylate (200 mg, 0.965 mmol, 1 equiv) and THF (2 mL): H2O(2 mL) at 25°C. Then NaOH (115.80 mg, 2.895 mmol, 3 equiv) was added under nitrogen atmosphere. The resulting mixture was stirred for 2 hours at room temperature under argon atmosphere. The reaction was monitored by LCMS. The filtrate was concentrated under reduced pressure. This resulted in 4- (cyclobutylamino)pyrimidine-2-carboxylic acid as a white solid. The crude product mixture was used in the next step directly without further purification. [000698] LC-MS (ES, m/z): [M+H]+: 194.10 [000699] Synthesis of 4-(cyclobutylamino)-N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5- yl)pyrimidine-2-carboxamide
[000700] A mixture of 4-(cyclobutylamino)pyrimidine-2-carboxylic acid (185 mg, 0.958 mmol, 1 equiv), 2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5-carboxylic acid (88.83 mg, 0.308 mmol, 0.90 equiv), HATU (1092.26 mg, 2.874 mmol, 3 equiv) and DIEA (185.64 mg,
1.437 mmol, 1.5 equiv) in DMF (5 mL) was stirred ovnenight at 60°C under argon atmosphere. The reaction was monitored by LCMS. The resulting mixture was dissolved in DMF (5 mL). The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 10% to 80% gradient in 25 min; detector, UV 254 nm to afford to afford 4-(cyclobutylamino)-N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H- isoindol-5-yl]pyrimidine-2-carboxamide (47.8 mg, 10.97%) as a white solid. [000701] LC-MS (ES, m/z): [M+H]+: 435.15 [000702] 1H-NMR: (400 MHz, DMSO-d6) δ 10.98 (s, 1H), 10.82 – 10.64 (m, 1H), 8.35 – 7.96 (m, 3H), 7.89 (dd, J = 8.4, 1.8 Hz, 1H), 7.71 (d, J = 8.3 Hz, 1H), 6.56 (d, J = 6.0 Hz, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 4.48 (d, J = 17.3 Hz, 1H), 4.33 (d, J = 17.2 Hz, 1H), 3.42 – 3.34 (m, 1H), 2.99 – 2.85 (m, 1H), 2.65 – 2.56 (m, 1H), 2.48 – 2.37 (m, 1H), 2.37 – 2.27 (m, 2H), 2.10 – 1.84 (m, 3H), 1.78 – 1.65 (m, 2H). [000703] Example 1.71. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-isopropyl-3-methyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxamide (I-121)
[000704] Synthesis of 1-(2,4-dichloropyrimidin-5-yl)ethan-1-ol
[000705] Into a 100 mL 3-necked round-bottom flask were added 2,4-dichloropyrimidine-5- carbaldehyde (2 g, 11.301 mmol, 1 equiv) in THF (30 mL, 370.283 mmol, 32.77 equiv) was added MeMgBr (1.35 g, 11.301 mmol, 1 equiv) dropwise -78°C under nitrogen atmosphere. The
resulting mixture was stirred for 2 h at -78°C under nitrogen atmosphere. Desired product could be detected by LCMS. The reaction was quenched by the addition of 1N HCI (30 mL) at 0°C. The resulting mixture was extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with brine (1 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (2:1) to afford 1-(2,4-dichloropyrimidin-5-yl)ethanol (1.5 g, 68.76%) as a yellow oil. [000706] LC-MS (ES, m/z): [M+H]+: 193.1 [000707] 1H-NMR: (400 MHz, DMSO-d6) δ 8.84 (d, J = 0.7 Hz, 1H), 4.94 (q, J = 6.5 Hz, 1H), 1.39 (d, J = 6.5 Hz, 3H). [000708] Synthesis of 1-(2,4-dichloropyrimidin-5-yl)ethan-1-one
[000709] Into a 40 mL round-bottom flask were added 1-(2,4-dichloropyrimidin-5-yl)ethanol (1.5 g, 7.771 mmol, 1 equiv) in DCM (10 mL, 157.306 mmol, 20.24 equiv) was added Dess- Martin (6.59 g, 15.542 mmol, 2 equiv) in portions at room temperature under air atmosphere. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS. The reaction was quenched by the addition of Water (10 mL) at 0°C. The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1:1) to afford 1-(2,4- dichloropyrimidin-5-yl)ethanone (1.3 g, 87.58%) as a yellow oil. [000710] LC-MS (ES, m/z): no mass signal [000711] 1H-NMR: (400 MHz, DMSO-d6) δ 9.15 (d, J = 0.7 Hz, 1H) , 2.6 (d, J = 6.5 Hz, 3H). [000712] Synthesis of 6-chloro-1-isopropyl-3-methyl-1H-pyrazolo[3,4-d]pyrimidine
[000713] Into a 40 mL round-bottom flask were added 1-(2,4-dichloropyrimidin-5-yl)ethanone (1.3 g, 6.806 mmol, 1 equiv) and isopropylhydrazine (1.01 g, 13.612 mmol, 2 equiv) in MeCN (10 mL, 190.242 mmol, 27.95 equiv) was added DIEA (1.76 g, 13.612 mmol, 2 equiv) in portions at room temperature under air atmosphere. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 30% to 80% gradient in 10 min; detector, UV 254 nm. This resulted in 6-chloro-1-isopropyl-3-methylpyrazolo[3,4- d]pyrimidine (400 mg, 27.90%) as a white solid. [000714] LC-MS (ES, m/z): [M+H]+: 211.1 [000715] 1H-NMR: (400 MHz, DMSO-d6) δ 9.23 (s, 1H), 5.00 – 5.02 (m, 1H), 2.58 (s, 3H), 1.48 (d, J = 6.7 Hz, 6H). [000716] Synthesis of methyl 1-isopropyl-3-methyl-1H-pyrazolo[3,4-d]pyrimidine-6- carboxylate
[000717] Into a 30 mL pressure tank reactor were added 6-chloro-1-isopropyl-3- methylpyrazolo[3,4-d]pyrimidine (200 mg, 0.949 mmol, 1 equiv) and Pd(dppf)Cl2 (69.47 mg, 0.095 mmol, 0.1 equiv) in MeOH (100.00 mL, 2468.966 mmol, 2601.65 equiv) was added TEA (192.14 mg, 1.898 mmol, 2 equiv) in portions at room temperature under air atmosphere. The resulting mixture was stirred for overnight at 100°C under CO atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 30% to 80% gradient in 10 min; detector, UV 254 nm. This resulted in methyl 1-isopropyl-3- methylpyrazolo[3,4-d]pyrimidine-6-carboxylate (200 mg, 89.93%) as a brown solid. [000718] LC-MS (ES, m/z): [M+H]+: 235.0 [000719] 1H-NMR: (400 MHz, DMSO-d6) δ 9.43 (s, 1H), 5.14 – 5.16 (m, 1H), 3.94 (s, 3H), 2.63 (s, 3H), 1.49 (d, J = 6.7 Hz, 6H). [000720] Synthesis of 1-isopropyl-3-methyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxylic acid
[000721] Into a 8 mL vial were added methyl 1-isopropyl-3-methylpyrazolo[3,4-d]pyrimidine- 6-carboxylate (100 mg, 0.427 mmol, 1 equiv) in THF (5 mL, 61.714 mmol, 144.57 equiv) and H2O (5 mL, 277.546 mmol, 650.18 equiv) was added LiOH (51.12 mg, 2.135 mmol, 5 equiv) in portions at room temperature under air atmosphere. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS. The mixture was neutralized to pH = 7 with conc. HCl. The resulting mixture was filtered, the filter cake was washed with MeOH (3 x 10 mL). The filtrate was concentrated under reduced pressure. The crude product used in the next step directly without further purification. This resulted in 1-isopropyl-3-methylpyrazolo[3,4-d]pyrimidine-6-carboxylic acid (80 mg, 85.10%) as a brown solid. [000722] LC-MS (ES, m/z): [M+H]+: 220.9 [000723] 1H-NMR: (400 MHz, DMSO-d6) δ 9.23 (s, 1H), 5.12 – 5.04 (m, 1H), 2.58 (s, 3H), 1.46 (d, J = 6.7 Hz, 6H). [000724] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1-isopropyl-3- methyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxamide
[000725] Into a 8 mL vial were added 1-isopropyl-3-methylpyrazolo[3,4-d]pyrimidine-6- carboxylic acid (70 mg, 0.318 mmol, 1 equiv) and 3-(5-amino-1-oxo-3H-isoindol-2- yl)piperidine-2,6-dione (98.89 mg, 0.382 mmol, 1.2 equiv) in DMF (7.00 mL, 90.493 mmol, 284.57 equiv) were added HATU (181.28 mg, 0.477 mmol, 1.5 equiv) and DIEA (82.16 mg, 0.636 mmol, 2 equiv) in portions at room temperature under air atmosphere. The resulting mixture was stirred for overnight at 60°C under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 30% to
80% gradient in 20 min; detector, UV 254 nm. This resulted in N-[2-(2,6-dioxopiperidin-3-yl)-1- oxo-3H-isoindol-5-yl]-1-isopropyl-3-methylpyrazolo[3,4-d]pyrimidine-6-carboxamide (73.9 mg, 49.27%) as a white solid. [000726] LC-MS (ES, m/z): [M+H]+: 462.00 [000727] 1H-NMR: 1H NMR (400 MHz, DMSO-d6) δ 11.11 (s, 1H), 11.00 (s, 1H), 9.49 (s, 1H), 8.26 (d, J = 1.8 Hz, 1H), 8.00 (dd, J = 8.4, 1.8 Hz, 1H), 7.76 (d, J = 8.3 Hz, 1H), 5.37 – 5.22 (m, 1H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.51 (d, J = 17.3 Hz, 1H), 4.36 (d, J = 17.3 Hz, 1H), 3.00 – 2.86 (m, 1H), 2.66 (s, 3H), 2.65 – 2.57 (m, 1H), 2.49 – 2.34 (m, 1H), 2.10 – 1.98 (m, 1H), 1.53 (d, J = 6.7 Hz, 6H). [000728] Example 1.72. Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-3,4-dimethyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-114)
[000729] Synthesis of 5-bromo-3,4-dimethyl-2-(trimethylsilyl)-1H-pyrrolo[2,3-b]pyridine
[000730] Into a 25 mL round-bottom flask were added 5-bromo-3-iodo-4-methylpyridin-2- amine (1900 mg, 6.072 mmol, 1 equiv) and LiCl (308.85 mg, 7.286 mmol, 1.2 equiv) in DMF (70 mL), Pd(OAc)2 (136.31 mg, 0.607 mmol, 0.1 equiv) and trimethyl(prop-1-yn-1-yl)silane (3407.55 mg, 30.360 mmol, 5 equiv) dropwise at room temperature under argon atmosphere. The resulting mixture was stirred for overnight at 90°C. Desired product was detected by LCMS, The resulting mixture was filtered, the filter cake was washed with MeOH (3 x 50 mL). The
filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 5% to100% gradient in 30 min; detector, UV 254 nm.to afford 5-bromo-3,4- dimethyl-2-(trimethylsilyl)-1H-pyrrolo[2,3-b]pyridine (740 mg, 41.00%) as a white solid. [000731] LC-MS (ES, m/z): [M+H]+: 296.95 [000732] Synthesis of 5-bromo-3,4-dimethyl-1H-pyrrolo[2,3-b]pyridine
[000733] Into a 8mL vial were added 5-bromo-3,4-dimethyl-2-(trimethylsilyl)-1H-pyrrolo[2,3- b]pyridine (710 mg, 2.388 mmol, 1 equiv) and HCl (g) in THF (10 mL) . The resulting mixture was stirred for 2 h at 55°C under nitrogen atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. This resulted in5- bromo-3,4-dimethyl-1H-pyrrolo[2,3-b]pyridine (480 mg, crude) as a solid. [000734] LC-MS (ES, m/z): [M+H]+: 224.90 [000735] Synthesis of methyl 3,4-dimethyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate
[000736] To a solution of 5-bromo-3,4-dimethyl-1H-pyrrolo[2,3-b]pyridine (400 mg, 1.777 mmol, 1 equiv) in MeOH (10 mL) was added Pd(dppf)Cl2 (130.03 mg, 0.178 mmol, 0.1 equiv) and Et3N (539.48 mg, 5.331 mmol, 3 equiv) in a pressure tank. The mixture was purged with nitrogen for 3 mins and then was pressurized to 40 atm. with carbon monoxide at 120°C for overnight. The reaction mixture was cooled to room temperature and filtered to remove insoluble solids. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water , 50% to 70% gradient in 10 min; detector, UV 254 nm.to afford as a methyl 3,4-dimethyl-1H-pyrrolo[2,3-b]pyridine-5- carboxylate (171 mg, 47.12%) as a white solid. [000737] LC-MS (ES, m/z): [M+H]+: 205.05
[000738] Synthesis of methyl 1-cyclobutyl-3,4-dimethyl-1H-pyrrolo[2,3-b]pyridine-5- carboxylate
[000739] Into a 25 mL round-bottom flask were added N-[2-(2,6-dioxo-1-{[2- (trimethylsilyl)ethoxy]methyl}piperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1H-pyrrolo[2,3- b]pyridine-6-carboxamide (10 mg, 0.019 mmol, 1 equiv) in DMF (5 mL) and Cs2CO3 (574.33 mg, 1.763 mmol, 1.5 equiv) at 0°C. The mixture was stirred for 15 min. Then iodocyclobutane (213.88 mg, 1.175 mmol, 1 equiv) was dropwise at 0°C. The resulting mixture was stirred for 1 overnight at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in H2O, 10% to 50% gradient in 30 min; detector, UV 254 nm. This resulted in methyl 1-cyclobutyl-3,4-dimethylpyrrolo[2,3- b]pyridine-5-carboxylate (150 mg, 49.41%) as a white solid. [000740] LC-MS (ES, m/z): [M+H]+: 259.00 [000741] Synthesis of 1-cyclobutyl-3,4-dimethyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000742] Into a 25 mL round-bottom flask were added methyl 1-cyclobutyl-3,4- dimethylpyrrolo[2,3-b]pyridine-5-carboxylate (140 mg, 0.542 mmol, 1 equiv) in MeOH (1 mL) and NaOH (43.35 mg, 1.084 mmol, 2 equiv) in H2O (1 mL) dropwises at 0°C under nitrogen atmosphere. The resulting mixture was stirred for additional 2 h at room temperature. Desired product could be detected by LCMS. The mixture was neutralized to pH = 7 with HCl(aq). The resulting mixture was concentrated under reduced pressure. The crude product (160 mg, crude) was used in the next step directly without further purification. [000743] LC-MS (ES, m/z): [M+H]+: 245.00
[000744] Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-3,4- dimethyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide
[000745] Into a 8 mL vial were added 1-cyclobutyl-3,4-dimethylpyrrolo[2,3-b]pyridine-5- carboxylic acid (40 mg, 0.164 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine- 2,6-dione (42.45 mg, 0.164 mmol, 1 equiv) and Pyridine (2 mL) at room temperature. To the above mixture was added POCl3 (75.31 mg, 0.492 mmol, 3 equiv) dropwise at room temperature. The resulting mixture was stirred for additional 1 h at room temperature. Desired product could be detected by LCMS. The crude product was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C18 OBD Column 30*150mm 5μm, n; Mobile Phase A: Water(0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 7% B to 27% B in 10 min; Wave Length: 254 nm/220 nm; RT1(min): 10.01) to afford 1-cyclobutyl-N-[2- (2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-3,4-dimethylpyrrolo[2,3-b]pyridine-5- carboxamide (14.5 mg, 17.78%) as a white solid. [000746] LC-MS (ES, m/z): [M+H]+: 486.15 [000747] 1H-NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.66 (s, 1H), 8.32 (s, 1H), 8.16 (s, 1H), 7.76 (dd, J = 8.4, 1.7 Hz, 1H), 7.70 (d, J = 8.2 Hz, 1H), 7.60 (s, 1H), 5.27 (p, J = 8.6 Hz, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 4.47 (d, J = 17.3 Hz, 1H), 4.33 (d, J = 17.3 Hz, 1H), 2.99 – 2.86 (m, 1H), 2.73 (s, 3H), 2.65 – 2.56 (m, 1H), 2.49 – 2.44 (m, 5H), 2.44 – 2.34 (m, 3H), 2.07 – 1.96 (m, 1H), 1.90 – 1.78 (m, 2H). [000748] Example 1.73. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-(propan-2-yl-2-d)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-120)
[000749] Synthesis of methyl 1-(propan-2-yl-2-d)-1H-pyrrolo[2,3-b]pyridine-5-carboxylate
[000750] Into a 40 mL vial were added methyl 1H-pyrrolo[2,3-b]pyridine-5-carboxylate (300 mg, 1.703 mmol, 1 equiv), (2-2H)propan-2-ol (114.45 mg, 1.873 mmol, 1.1 equiv), PPh3 (669.97 mg, 2.554 mmol, 1.5 equiv) and THF (8 mL) at room temperature. To the above mixture was added DIAD (516.50 mg, 2.554 mmol, 1.5 equiv) dropwise at 0°C. The resulting mixture was stirred for additional 4 h at room temperature. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The reaction was quenched by the addition of Water (15 mL) at room temperature. The resulting mixture was extracted with EtOAc (2 x 20 mL). The combined organic layers were washed with brine (1 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (3:1) to afford methyl 1-[(2-2H)propan- 2-yl]pyrrolo[2,3-b]pyridine-5-carboxylate (250 mg, 66.96%) as a yellow solid. [000751] LC-MS (ES, m/z): [M+H]+: 220.20 [000752] 1H-NMR: (400 MHz, Chloroform-d) δ 8.98 (d, J = 1.9 Hz, 1H), 8.56 (d, J = 2.0 Hz, 1H), 7.38 (d, J = 3.6 Hz, 1H), 6.57 (d, J = 3.7 Hz, 1H), 3.95 (s, 3H), 1.53 (s, 6H). [000753] Synthesis of 1-(propan-2-yl-2-d)-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000754] Into a 50 mL round-bottom flask were added methyl 1-[(2-2H)propan-2- yl]pyrrolo[2,3-b]pyridine -5-carboxylate (150 mg, 0.684 mmol, 1 equiv), THF (3 mL),H2O (3 mL) and LiOH.H2O (43.06 mg, 1.026 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS. The mixture was neutralized to pH = 7 with Cationic resin. The resulting mixture was filtered, the filter cake was washed with MeOH (2 x 10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water , 5% to 100% gradient in 30 min; detector, UV 254 nm.to afford to afford 1-[(2 -2H)propan-2-yl]pyrrolo[2,3-b]pyridine-5-carboxylic acid (130 mg, 92.59%) as an off-white solid. [000755] LC-MS (ES, m/z): [M+H]+: 206.20 [000756] 1H-NMR: (400 MHz, DMSO-d6) δ 12.88 (s, 1H), 8.82 (d, J = 2.0 Hz, 1H), 8.51 (d, J = 2.0 Hz, 1H), 7.80 (d, J = 3.6 Hz, 1H), 6.65 (d, J = 3.6 Hz, 1H), 1.48 (s, 6H). [000757] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1-(propan-2-yl-2- d)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide
[000758] Into a 40 mL vial were added 1-[(2-2H)propan-2-yl]pyrrolo[2,3-b]pyridine-5- carboxylic acid (190 mg, 0.926 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine- 2,6-dione (240.02 mg, 0.926 mmol, 1 equiv), DIEA (717.92 mg, 5.556 mmol, 6 equiv), DMF (7 mL) and HATU (528.01 mg, 1.389 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at 60°C under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 40% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in -(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-1-(propan-2-yl-2-d)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (103.6 mg, 24.71%) as a white solid.
[000759] LC-MS (ES, m/z): [M+H]+: 447.15 [000760] 1H-NMR: (400 MHz, DMSO-d6) δ 11.00 (s, 1H), 10.61 (s, 1H), 8.87 (d, J = 2.2 Hz, 1H), 8.60 (d, J = 2.1 Hz, 1H), 8.18 (d, J = 1.7 Hz, 1H), 7.90 – 7.80 (m, 2H), 7.73 (d, J = 8.3 Hz, 1H), 6.68 (d, J = 3.6 Hz, 1H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.49 (d, J = 17.3 Hz, 1H), 4.34 (d, J = 17.3 Hz, 1H), 3.00 – 2.86 (m, 1H), 2.66 – 2.57 (m, 1H), 2.48 – 2.33 (m, 1H), 2.10 – 1.98 (m, 1H), 1.50 (s, 6H). [000761] Example 1.74. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 4-hydroxy-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxamide (I-111)
[000762] Synthesis of 4-fluoro-6-iodo-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidine
[000763] Into a 50mL 3-necked round-bottom flask were added 4-fluoro-1- isopropylpyrazolo[3,4-d]pyrimidin-6-amine (50 mg, 0.256 mmol, 1 equiv) ,I2 (65.01 mg, 0.256 mmol, 1 equiv) ,CH2I2 (686.05 mg, 2.560 mmol, 10 equiv) ,THF (2 mL) ,(3-methylbutyl) nitrite (90.02 mg, 0.768 mmol, 3 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for additional 2 h at 66 C.The reaction was quenched by the addition of Water (5mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x10 mL). The combined organic layers were washed with brine (3x10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (10:1) to afford 4-fluoro-6-iodo-1-isopropylpyrazolo[3,4-d]pyrimidine (60 mg, 76.53%) as a yellow solid.
[000764] 1H-NMR: (400 MHz, DMSO-d6) δ 8.49 (s, 1H), 5.11 (hept, J = 6.7 Hz, 1H), 1.49 (d, J = 6.6 Hz, 6H). [000765] Synthesis of 4-fluoro-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidine-6-carbonitrile
[000766] A solution/mixture of 4-chloro-6-iodo-1-isopropylpyrazolo[3,4-d]pyrimidine (50 mg, 0.155 mmol, 1 equiv) and CuCN (58.52 mg, 0.654 mmol, 2 equiv) in DMF (5 mL) was stirred for 2h at 100 C under argon atmosphere.The resulting mixture was extracted with EtOAc/ACN (3 x 5 mL). dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water , 10% to 90% gradient in 10 min; detector, UV 254 nm.This resulted in 4-hydroxy-1-isopropylpyrazolo[3,4- d]pyrimidine-6-carbonitrile (10 mg, 15.06%) as a colorless oil. [000767] Synthesis of 4-hydroxy-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxylic acid
[000768] A solution of 4-hydroxy-1-isopropylpyrazolo[3,4-d]pyrimidine-6-carbonitrile (100 mg, 0.492 mmol, 1 equiv) and NaOH (98.42 mg, 2.460 mmol, 5 equiv) in THF (1 mL) was stirred at 50°C for overnight under argon atmosphere. Desired product could be detected by LCMS.~50% product. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water , 10% to 90% gradient in 10 min; detector, UV 254 nm.This resulted in 4-hydroxy-1-isopropylpyrazolo[3,4- d]pyrimidine-6-carboxylic acid (20 mg, 18.29%) as a white solid. [000769] 1H-NMR: (400 MHz, DMSO-d6) δ 14.52 (s, 0H), 12.01 (s, 1H), 8.13 (s, 1H), 5.21 – 4.89 (m, 1H), 1.57 – 1.40 (m, 6H). [000770] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-4-hydroxy-1- isopropyl-1H-pyrazolo[3,4-d]pyrimidine-6-carboxamide
[000771] Into a 50 mL round-bottom flask were added 4-hydroxy-1-isopropylpyrazolo[3,4- d]pyrimidine-6-carboxylic acid (60 mg, 0.270 mmol, 1.00 equiv) and 3-(5-amino-1-oxo-3H- isoindol-2-yl)piperidine-2,6-dione (84.01 mg, 0.324 mmol, 1.2 equiv) in DMF (3 mL)at room temperature. The resulting mixture was stirred at room temperature for 10min under argon atmosphere. To a stirred mixture of DIEA (69.80 mg, 0.540 mmol, 2 equiv) in DMF (3 mL) in portions at room temperature under argon atmosphere. The resulting mixture was stirred at room temperature for 1 h under argon atmosphere. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 220 nm.To afford N-[2-(2,6- dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-4-hydroxy-1-isopropylpyrazolo[3,4-d]pyrimidine- 6-carboxamide (30.6 mg, 23.23%) as a white solid. [000772] LC-MS (ES,m/z): [M+H]+:464 [000773] 1H NMR: (400 MHz, DMSO-d6) δ 12.43 (s, 1H), 10.99 (d, J = 16.7 Hz, 2H), 8.18 (d, J = 14.2 Hz, 2H), 7.95 (d, J = 8.3 Hz, 1H), 7.79 (d, J = 8.3 Hz, 1H), 5.32 (h, J = 6.7 Hz, 1H), 5.13 (dd, J = 13.2, 5.2 Hz, 1H), 4.58 – 4.17 (m, 2H), 2.93 (ddd, J = 18.1, 13.3, 5.3 Hz, 1H), 2.62 (dd, J = 25.1, 9.6 Hz, 1H), 2.46 – 2.28 (m, 1H), 2.17 – 1.87 (m, 1H), 1.50 (d, J = 6.6 Hz, 6H). [000774] Example 1.75. Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-4-fluoro-3-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-112)
[000775] Into a 8mL vial were added 1-cyclobutyl-4-fluoro-3-methylpyrrolo[2,3-b]pyridine-5- carboxylic acid (50 mg, 0.201 mmol, 1 equiv) and 3-(5-amino-1-oxo-3H-isoindol-2-
yl)piperidine-2,6-dione (62.66 mg, 0.241 mmol, 1.2 equiv) in DMF (2 mL, 25.843 mmol, 128.31 equiv) were added HATU (114.87 mg, 0.301 mmol, 1.5 equiv) and DIEA (52.06 mg, 0.402 mmol, 2 equiv) in portions at room temperature under air atmosphere. The resulting mixture was stirred for overnight at 60°C under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 30% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in 1-cyclobutyl-N-[2-(2,6- dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-4-fluoro-3-methylpyrrolo[2,3-b]pyridine-5- carboxamide (12.1 mg, 11.77%) as a white solid. [000776] LC-MS (ES, m/z): [M+H]+=490.2 [000777] 1H NMR: (400 MHz, DMSO-d6) δ 10.98 (s, 1H), 10.70 (s, 1H), 8.50 (d, J = 9.3 Hz, 1H), 8.11 (d, J = 1.7 Hz, 1H), 7.76 (dd, J = 8.3, 1.7 Hz, 1H), 7.72 (d, J = 7.7 Hz, 2H), 5.28 (p, J = 8.6 Hz, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 4.48 (d, J = 17.3 Hz, 1H), 4.34 (d, J = 17.3 Hz, 1H), 2.91-2.94 (m, 1H), 2.66 – 2.51 (m, 2H), 2.47 – 2.32 (m, 5H), 2.06 – 1.98 (m, 1H), 1.86 (td, J = 10.0, 5.4 Hz, 2H). [000778] Example 1.76. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 3-iodo-1-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-113)
[000779] Synthesis of methyl 1-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate
[000780] Into a 40 mL vial were added methyl 1H-pyrrolo[2,3-b]pyridine-5-carboxylate (400 mg, 2.270 mmol, 1 equiv), K2CO3 (627.58 mg, 4.540 mmol, 2 equiv), DMF (5 mL) and CH3I (483.40 mg, 3.405 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for
overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The residue was purified by reversed- phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH4HCO3), 45% to 55% gradient in 10 min; detector, UV 254 nm. This resulted in methyl 1-methylpyrrolo[2,3-b]pyridine-5-carboxylate (370 mg, 85.68%) as a white solid. [000781] LC-MS (ES ,m/z): [M+1]+=191.15 [000782] 1H NMR: (400 MHz, DMSO-d6) δ 8.84 (d, J = 2.1 Hz, 1H), 8.54 (d, J = 2.0 Hz, 1H), 7.66 (d, J = 3.5 Hz, 1H), 6.64 (d, J = 3.5 Hz, 1H), 3.88 (d, J = 7.0 Hz, 6H). [000783] Synthesis of methyl 3-iodo-1-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate
[000784] Into a 40 mL vial were added methyl 1-methylpyrrolo[2,3-b]pyridine-5-carboxylate (320 mg, 1.682 mmol, 1 equiv), acetone (10 mL) and NIS (567.78 mg, 2.523 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for 3 h at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1:2) to afford methyl 3-iodo-1- methylpyrrolo[2,3-b]pyridine-5-carboxylate (510 mg, 95.90%) as a orange solid. [000785] LC-MS (ES, m/z): [M+1]+=316.90 [000786] 1H NMR: (400 MHz, DMSO-d6) δ 8.84 (d, J = 2.0 Hz, 1H), 8.15 (d, J = 2.0 Hz, 1H), 7.92 (s, 1H), 3.89 (d, J = 16.8 Hz, 6H). [000787] Synthesis of 3-iodo-1-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000788] Into a 40 mL vial were added methyl 3-iodo-1-methylpyrrolo[2,3-b]pyridine-5- carboxylate (300 mg, 0.949 mmol, 1 equiv), THF (4 mL), H2O (4 mL) and LiOH.H2O (59.73 mg, 1.423 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired
product could be detected by LCMS. The mixture was neutralized to pH = 7 with cationic resin. The resulting mixture was filtered, the filter cake was washed with MeOH (2 x 10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (0:1) to afford 3-iodo-1-methylpyrrolo[2,3- b]pyridine-5 -carboxylic acid (257 mg, 89.64%) as a yellow solid. [000789] LC-MS (ES, m/z): [M+1]+=302.95 [000790] 1H NMR: (400 MHz, DMSO-d6) δ 8.85 (d, J = 2.0 Hz, 1H), 8.16 (d, J = 2.0 Hz, 1H), 7.91 (s, 1H), 3.88 (s, 3H). [000791] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-3-iodo-1-methyl- 1H-pyrrolo[2,3-b]pyridine-5-carboxamide
[000792] Into a 40 mL vial were added 3-iodo-1-methylpyrrolo[2,3-b]pyridine-5-carboxylic acid (100 mg, 0.331 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6-dione (85.83 mg, 0.331 mmol, 1 equiv), DIEA (256.72 mg, 1.986 mmol, 6 equiv), DMF (5 mL) and HATU (188.81 mg, 0.497 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at 60°C under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 40% to 55% gradient in 15 min; detector, UV 254 nm. This resulted in N-[2- (2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-3-iodo-1-methylpyrrolo[2,3 -b]pyridine-5- carboxamide (10 mg, 5.32%) as a white solid. [000793] LC-MS (ES, m/z): [M+1]+=543.85 [000794] 1H NMR: (400 MHz, DMSO-d6) δ 11.00 (s, 1H), 10.73 (s, 1H), 8.92 (s, 1H), 8.37 (s, 1H), 8.17 (s, 1H), 7.93 (s, 1H), 7.88 (d, J = 8.2 Hz, 1H), 7.74 (d, J = 8.3 Hz, 1H), 5.12 (dd, J = 13.3, 5.1 Hz, 1H), 4.50 (d, J = 17.1 Hz, 1H), 4.36 (d, J = 17.1 Hz, 1H), 3.90 (s, 3H), 2.95 – 2.88 (m, 1H), 2.66 – 2.57 (m, 1H), 2.46 – 2.35 (m, 1H), 2.07 – 1.99 (m, 1H).
[000795] Example 1.77. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-(1-methylpiperidin-4-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-116)
[000796] Synthesis of methyl 1-(1-methylpiperidin-4-yl)-1H-pyrrolo[2,3-b]pyridine-5- carboxylate
[000797] Into a 40 mL vial were added methyl 1H-pyrrolo[2,3-b]pyridine-5-carboxylate (300 mg, 1.703 mmol, 1 equiv), 4-bromo-1-methylpiperidine (454.85 mg, 2.554 mmol, 1.5 equiv) and DMF (1 mL) at room temperature. To the above mixture was added Cs2CO3 (832.23 mg, 2.554 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for additional overnight at room temperature. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10mmol/L NH4HCO3), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in tert-butyl 4-[5- (methoxycarbonyl)pyrrolo[2,3- b]pyridin-1-yl]piperidine-1-carboxylate (140 mg, 13.04%) as a yellow oil. [000798] LC-MS (ES, m/z): [M+ H]+: 274.10 [000799] Synthesis of 1-(1-methylpiperidin-4-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000800] Into a 8 mL vial were added methyl 1-(1-methylpiperidin-4-yl)pyrrolo[2,3- b]pyridine-5-carboxylate (30 mg, 0.110 mmol, 1 equiv)and MeOH (1 mL) at room temperature. To the above mixture was added LiOH H2O (27.63 mg, 0.660 mmol, 6 equiv) in H2O (1 mL) dropwise at room temperature. The resulting mixture was stirred for additional overnight at room temperature. Desired product could be detected by LCMS. The mixture/residue was neutralized to pH 7 with conc. HCl. The resulting mixture was concentrated under reduced pressure & lyophilized. The crude product was used in the next step directly without further purification. [000801] LC-MS (ES, m/z): [M+H]+:260.00 [000802] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1-(1- methylpiperidin-4-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide
[000803] Into a 8 mL vial were added 1-(1-methylpiperidin-4-yl)pyrrolo[2,3-b]pyridine-5- carboxylic acid (5 mg, 0.019 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6- dione (5.00 mg, 0.019 mmol, 1 equiv) and Pyridine (1 mL) at room temperature. To the above mixture was added POCl3 (14.78 mg, 0.095 mmol, 5 equiv) dropwise at room temperature. The resulting mixture was stirred for additional 1 h at room temperature. Desired product could be detected by LCMS. The reaction was quenched with Water at 0 °C. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm. The crude product was purified by Prep-HPLC with the following conditions (column, XBridge Prep; mobile phase, MeCN in Water (0.1% TFA), 10% to 50% gradient in 10 min; detector, UV 254 nm) to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1-(1-methylpiperidin-4- yl)pyrrolo[2,3-b]pyridine-5-carboxamide (2.8 mg, 4.77%) as a white solid. [000804] LC-MS (ES, m/z): [M+H]+:501.10
[000805] 1H NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.65 (d, J = 2.5 Hz, 1H), 8.88 (d, J = 2.1 Hz, 1H), 8.65 (d, J = 2.1 Hz, 1H), 8.17 (d, J = 1.7 Hz, 1H), 7.86 (d, J = 8.3 Hz, 1H), 7.78 – 7.64 (m, 2H), 6.74 (d, J = 3.6 Hz, 1H), 5.18 – 4.94 (m, 2H), 4.48 (d, J = 17.2 Hz, 1H), 4.34 (d, J = 17.3 Hz, 1H), 3.62 (d, J = 12.3 Hz, 2H), 3.06-2.95(m, 1H), 2.89 (d, J = 27.9 Hz, 3H), 2.68- 2.57(m, 1H), 2.47 – 2.30 (m, 4H), 2.23 (d, J = 13.5 Hz, 2H), 2.11 – 1.90 (m, 2H). [000806] Example 1.78. Synthesis of tert-butyl 4-(5-((2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)carbamoyl)-1H-pyrrolo[2,3-b]pyridin-1-yl)piperidine-1-carboxylate (I- 117)
[000807] Synthesis of methyl 1-(1-(tert-butoxycarbonyl)piperidin-4-yl)-1H-pyrrolo[2,3- b]pyridine-5-carboxylate [000808] Into a 40 mL vial were added methyl 1H-pyrrolo[2,3-b]pyridine-5-carboxylate (500 mg, 2.838 mmol, 1 equiv), tert-butyl 4-bromopiperidine-1-carboxylate (824.69 mg, 3.122 mmol, 1.1 equiv) and DMF (5 mL, 12.922 mmol) at room temperature. To the above mixture was added Cs2CO3 (1387.05 mg, 4.257 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for additional overnight at room temperature. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10mmol/L NH4HCO3), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in tert-butyl 4-[5-
(methoxycarbonyl)pyrrolo[2,3-b]pyridin-1-yl]piperidine-1-carboxylate (140 mg, 13.04%) as a yellow solid. [000809] LC-MS (ES, m/z): [M+ H]+:360.20 [000810] 1H NMR: (400 MHz, DMSO-d6) δ 8.83 (d, J = 2.0 Hz, 1H), 8.55 (d, J = 2.1 Hz, 1H), 7.86 (d, J = 3.7 Hz, 1H), 6.67 (d, J = 3.6 Hz, 1H), 4.96 (dq, J = 11.4, 6.6, 5.7 Hz, 1H), 4.13 (d, J =13.0 Hz, 2H), 3.88 (s, 3H), 2.02-1.89 (m, 4H), 1.44 (s, 9H). [000811] Synthesis of 1-(1-(tert-butoxycarbonyl)piperidin-4-yl)-1H-pyrrolo[2,3-b]pyridine-5- carboxylic acid
[000812] Into a 8 mL vial were added tert-butyl 4-[5-(methoxycarbonyl)pyrrolo[2,3-b]pyridin- 1-yl]piperidine- 1-carboxylate (130 mg, 0.362 mmol, 1 equiv)and MeOH (1 mL) at room temperature. To the above mixture was added LiOH ● H2O (91.06 mg, 2.172 mmol, 6 equiv) in H2O (1 mL) dropwise at room temperature. The resulting mixture was stirred for additional overnight at room temperature. Desired product could be detected by LCMS. The mixture/residue was neutralized to pH 7 with conc. HCl. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 1-[1-(tert-butoxycarbonyl)piperidin-4- yl]pyrrolo[2,3-b]pyridine- 5-carboxylic acid (110 mg, 83.65%) as a yellow oil. [000813] LC-MS (ES, m/z): [M+H]+:460.10 [000814] 1H NMR: (400 MHz, DMSO-d6) 8.76 (d, J = 1.9 Hz, 1H), 8.33 (d, J = 1.9 Hz, 1H), 7.60 (d, J = 3.6 Hz, 1H), 6.45 (d, J = 3.6 Hz, 1H), 4.89 (p, J = 8.0 Hz, 1H), 4.14 (s, 1H), 3.17 (s, 1H), 2.93 (d, J = 30.1 Hz, 2H), 1.91 (h, J = 4.1, 3.6 Hz, 4H), 1.44 (s, 9H). [000815] Synthesis of tert-butyl 4-(5-((2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5- yl)carbamoyl)-1H-pyrrolo[2,3-b]pyridin-1-yl)piperidine-1-carboxylate
[000816] Into a 8 mL vial were added 1-[1-(tert-butoxycarbonyl)piperidin-4-yl]pyrrolo[2,3- b]pyridine-5-carboxylic acid (10 mg, 0.029 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2- yl)piperidine-2,6-dione (7.51 mg, 0.029 mmol, 1 equiv) and pyridine (1 mL) at room temperature. To the above mixture was added POCl3 (22.19 mg, 0.145 mmol, 5 equiv) dropwise at room temperature. The resulting mixture was stirred for additional 1 h at room temperature. Desired product could be detected by LCMS. The reaction was quenched with Water at 0 °C. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm. The crude product was purified by Prep-HPLC with the following conditions (Xselect CSH F-Phenyl OBD column; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm) to afford tert-butyl 4-(5-{[2- (2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]carbamoyl}pyrrolo[2,3-b]pyridin-1- yl)piperidine-1-carboxylate (9.5 mg, 54.42%) as a white solid. [000817] LC-MS (ES, m/z): [M-Boc+H]+:487.05 [000818] 1H NMR: (400 MHz, DMSO-d6) δ10.99 (s, 1H), 10.61 (s, 1H), 8.86 (d, J = 2.1 Hz, 1H), 8.61 (d, J = 2.1 Hz, 1H), 8.17 (d, J = 1.8 Hz, 1H), 7.85 (d, J = 6.5, 2.7 Hz, 2H), 7.72 (d, J = 8.4 Hz, 1H), 6.68 (d, J = 3.6 Hz, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 4.95 (td, J = 10.9, 10.58, 5.0 Hz, 1H), 4.48 (d, J = 17.3 Hz, 1H), 4.34 (d, J = 17.2 Hz, 1H), 4.14 (s, 2H), 2.98-2.87 (m, 2H), 2.61 (d, J = 16.9 Hz, 1H), 2.40 (dd, J = 13.0, 4.5 Hz, 1H), 2.08-1.86(m, 6H), 1.44 (s, 9H). [000819] Example 1.79. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-(piperidin-4-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-118)
[000820] Synthesis of methyl 1-(1-(tert-butoxycarbonyl)piperidin-4-yl)-1H-pyrrolo[2,3- b]pyridine-5-carboxylate
[000821] Into a 40 mL vial were added methyl 1H-pyrrolo[2,3-b]pyridine-5-carboxylate (500 mg, 2.838 mmol, 1 equiv), tert-butyl 4-bromopiperidine-1-carboxylate (824.69 mg, 3.122 mmol, 1.1 equiv) and DMF (5 mL, 12.922 mmol) at room temperature. To the above mixture was added Cs2CO3 (1387.05 mg, 4.257 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for additional overnight at 80°C. Desired product could be detected by LCMS. The resulting mixture was filtered, the filter cake was washed with DMF (3 x 3 mL). The filtrate was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10mmol/L NH4HCO3), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in tert-butyl 4-[5-(methoxycarbonyl)pyrrolo[2,3-b]pyridin-1- yl]piperidine-1-carboxylate (140 mg, 13.04%) as a yellow solid. [000822] LC-MS (ES, m/z): [M+ H]+: 360.20 [000823] 1H NMR: (400 MHz, DMSO-d6) δ 8.83 (d, J = 2.0 Hz, 1H), 8.55 (d, J = 2.1 Hz, 1H), 7.86 (d, J = 3.7 Hz, 1H), 6.67 (d, J = 3.6 Hz, 1H), 4.96 (dq, J = 11.4, 6.6, 5.7 Hz, 1H), 4.13 (d, J =13.0 Hz, 2H), 3.88 (s, 3H), 2.02-1.89 (m, 4H), 1.44 (s, 9H). [000824] Synthesis of 1-(1-(tert-butoxycarbonyl)piperidin-4-yl)-1H-pyrrolo[2,3-b]pyridine-5- carboxylic acid
[000825] Into a 8 mL vial were added tert-butyl 4-[5-(methoxycarbonyl)pyrrolo[2,3-b]pyridin- 1-yl]piperidine- 1-carboxylate (130 mg, 0.362 mmol, 1 equiv)and MeOH (1 mL) at room temperature. To the above mixture was added LiOH.H2O (91.06 mg, 2.172 mmol, 6 equiv) in H2O (1 mL) dropwise at room temperature. The resulting mixture was stirred for additional overnight at room temperature. Desired product could be detected by LCMS. The mixture/residue was neutralized to pH 7 with conc. HCl. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 1-[1-(tert-butoxycarbonyl)piperidin-4- yl]pyrrolo[2,3-b]pyridine- 5-carboxylic acid (110 mg, 83.65%) as a yellow oil. [000826] LC-MS (ES, m/z): [M+H]+:460.10 [000827] 1H NMR: (400 MHz, DMSO-d6) δ 8.76 (d, J = 1.9 Hz, 1H), 8.33 (d, J = 1.9 Hz, 1H), 7.60 (d, J = 3.6 Hz, 1H), 6.45 (d, J = 3.6 Hz, 1H), 4.89 (p, J = 8.0 Hz, 1H), 4.14 (s, 1H), 3.17 (s, 1H), 2.93 (d, J = 30.1 Hz, 2H), 1.91 (h, J = 4.1, 3.6 Hz, 4H), 1.44 (s, 9H). [000828] Synthesis of tert-butyl 4-(5-((2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5- yl)carbamoyl)-1H-pyrrolo[2,3-b]pyridin-1-yl)piperidine-1-carboxylate
[000829] Into a 8 mL vial were added 1-[1-(tert-butoxycarbonyl)piperidin-4-yl]pyrrolo[2,3- b]pyridine-5-carboxylic acid (10 mg, 0.029 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2- yl)piperidine-2,6-dione (7.51 mg, 0.029 mmol, 1 equiv) and Pyridine (1 mL) at room temperature. To the above mixture was added POCl3 (22.19 mg, 0.145 mmol, 5 equiv) dropwise at room temperature. The resulting mixture was stirred for additional 1 h at room temperature. Desired product could be detected by LCMS. The reaction was
quenched with Water at 0°C. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm. The crude product was purified by Prep-HPLC with the following conditions (Xselect CSH F-Phenyl OBD column; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm) to afford tert-butyl 4-(5-{[2- (2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]carbamoyl}pyrrolo[2,3-b]pyridin-1- yl)piperidine-1-carboxylate (9.5 mg, 54.42%) as a white solid. [000830] LC-MS (ES, m/z): [M-Boc+H]+:487.05 [000831] 1H NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.61 (s, 1H), 8.86 (d, J = 2.1 Hz, 1H), 8.61 (d, J = 2.1 Hz, 1H), 8.17 (d, J = 1.8 Hz, 1H), 7.85 (d, J = 6.5, 2.7 Hz, 2H), 7.72 (d, J = 8.4 Hz, 1H), 6.68 (d, J = 3.6 Hz, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 4.95 (td, J = 10.9, 10.58, 5.0 Hz, 1H), 4.48 (d, J = 17.3 Hz, 1H), 4.34 (d, J = 17.2 Hz, 1H), 4.14 (s, 2H), 2.98-2.87 (m, 2H), 2.61 (d, J = 16.9 Hz, 1H), 2.40 (dd, J = 13.0, 4.5 Hz, 1H), 2.08-1.86(m, 6H), 1.44 (s, 9H). [000832] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1-(piperidin-4-yl)- 1H-pyrrolo[2,3-b]pyridine-5-carboxamide
[000833] Into a 8 mL vial were added tert-butyl 4-(5-{[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H- isoindol-5-yl]carbamoyl}pyrrolo[2,3-b]pyridin-1-yl)piperidine-1-carboxylate (45 mg, 0.077 mmol, 1 equiv) and DCM (2 mL) at room temperature. To the above mixture was added TFA (0.4 mL) dropwise at room temperature. The resulting mixture was stirred for additional 1 h at room temperature. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm. The crude product was purified by Prep-HPLC with the following conditions (Xselect CSH F-Phenyl OBD column; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10
min; detector, UV 254 nm) to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1- (piperidin-4-yl)pyrrolo[2,3- b]pyridine-5-carboxamide (25.0 mg, 6.37%) as a white solid. [000834] LC-MS (ES, m/z): [M +H]+:487.05 [000835] 1H NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.61 (s, 1H), 8.86 (d, J = 2.0 Hz, 1H), 8.62 (d, J = 2.1 Hz, 1H), 8.26(s, 1H), 8.17 (s, 1H), 7.88-7.69 (m, 3H), 6.69 (d, J = 3.5 Hz, 1H), 5.11 (dd, J = 13.4, 5.0 Hz, 1H), 4.88 (s, 1H), 4.48 (d, J = 17.3 Hz, 1H), 4.34 (d, J = 17.2 Hz, 1H), 3.20 (s, 1H), 2.99-2.76 (m, 4H), 2.61 (d, J = 17.1 Hz, 1H), 2.45-2.37 (m, 1H), 2.12-1.91 (m, 5H). [000836] Example 1.80. Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)- 1-(tetrahydro-2H-pyran-4-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-119)
[000837] Synthesis of methyl 1-(tetrahydro-2H-pyran-4-yl)-1H-pyrrolo[2,3-b]pyridine-5- carboxylate
[000838] Into a 50 mL round-bottom flask were added methyl 1H-pyrrolo[2,3-b]pyridine-5- carboxylate (800 mg, 4.541 mmol, 1 equiv) and 4-iodooxane (1925.63 mg, 9.082 mmol) in DMF (15.00 mL) was added Cs2CO3 (2959.05 mg, 9.082 mmol, 2 equiv) in portions at room temperature under air atmosphere. The resulting mixture was stirred for overnight at 80°C under nitrogen atmosphere. Desired product could be detected by LCMS. The resulting mixture was filtered, the filter cake was washed with DMF (2 x 5 mL), The filtrate was purified by reversed-
phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 30% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in methyl 1-(oxan-4-yl)pyrrolo[2,3-b]pyridine-5-carboxylate (100 mg, 8.46%) as a white solid. [000839] LC-MS (ES, m/z): [M+H]+=261.2 [000840] Synthesis of 1-(tetrahydro-2H-pyran-4-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000841] Into a 50 mL round-bottom flask were added methyl 1-(oxan-4-yl)pyrrolo[2,3- b]pyridine-5-carboxylate (110 mg, 0.423 mmol, 1 equiv) in THF (3.00 mL), H2O (1.00 mL) was added LiOH (50.61 mg, 2.115 mmol, 5 equiv) at room temperature under air atmosphere. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was acidified to pH 7 with conc HCl. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 30% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in 1-(oxan-4-yl)pyrrolo[2,3-b]pyridine-5-carboxylic acid (80 mg, 76.87%) as a brown solid. The crude product used in the next step directly without further purification. [000842] LC-MS (ES, m/z): [M+H]+=247.2 [000843] 1H NMR: (400 MHz, DMSO-d6) δ 12.71 (s, 1H), 8.81 (s, 1H), 8.53 – 8.48 (m, 1H), 7.84 (d, J = 3.6 Hz, 1H), 6.65 (d, J = 3.6 Hz, 1H), 4.99-5.01 (m, 1H), 4.00-4.02 (m, 2H), 3.56 (td, J = 12.0, 1.9 Hz, 2H), 2.10-2.12 (m, 2H), 1.87-1.89 (m, 2H). [000844] Synthesis of N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1-(tetrahydro-2H- pyran-4-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide
[000845] Into a 25 mL round-bottom flask were added 1-(oxan-4-yl)pyrrolo[2,3-b]pyridine-5- carboxylic acid (70 mg, 0.284 mmol, 1 equiv) and 3-(5-amino-1-oxo-3H-isoindol-2- yl)piperidine-2,6-dione (81.06 mg, 0.312 mmol, 1.1 equiv) in DMF (7.00 mL, 90.372 mmol, 318.21 equiv) were added HATU (162.12 mg, 0.426 mmol, 1.5 equiv) and DIEA (73.48 mg, 0.568 mmol, 2 equiv) at room temperature. The resulting mixture was stirred for overnight at 60°C under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 30% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1-(oxan-4- yl)pyrrolo[2,3-b]pyridine-5-carboxamide (37.9 mg, 26.17%) as a white solid. [000846] LC-MS (ES, m/z): [M-H]- : 488.2 [000847] 1H NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.61 (s, 1H), 8.87 (d, J = 2.1 Hz, 1H), 8.62 (d, J = 2.1 Hz, 1H), 8.17 (d, J = 1.8 Hz, 1H), 7.86 (dd, J = 7.6, 2.6 Hz, 2H), 7.73 (d, J = 8.3 Hz, 1H), 6.69 (d, J = 3.6 Hz, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 5.01-5.03 (m, 1H), 4.48 (d, J = 17.2 Hz, 1H), 4.34 (d, J = 17.2 Hz, 1H), 4.03 (dd, J = 11.5, 4.4 Hz, 2H), 3.57-3.55 (m, 2H), 2.92-2.94 (m, 1H), 2.61 (d, J = 17.3 Hz, 1H), 2.39-2.41 (m, 1H), 2.14-2.26 (m, 2H), 2.09 – 1.97 (m, 1H), 1.90-1.92 (m, 2H). [000848] Example 1.81. Synthesis of 1-(cyclobutylmethyl)-N-(2-(2,6-dioxopiperidin-3-yl)- 1-oxoisoindolin-5-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-122)
[000849] Synthesis of 1-(cyclobutylmethyl)-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000850] Into a 40mL round-bottom flask were added methyl 1H-pyrrolo[2,3-b]pyridine-5- carboxylate (300 mg, 1.703 mmol, 1 equiv) in DMF (10 mL) was treated with NaH (204.33 mg, 5.109 mmol, 3 equiv, 60%) for 10 min at room temperature under nitrogen atmosphere followed
by the addition of (iodomethyl)cyclobutane (500.72 mg, 2.554 mmol, 1.5 equiv) dropwise room temperature. The resulting mixture was stirred for 2h at room temperature. Desired product could be detected by LCMS. The reaction was quenched with Water/Ice and stirred for 2h at room temperature. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 30% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in 1-(cyclobutylmethyl)pyrrolo[2,3- b]pyridine-5-carboxylic acid (150 mg, 38.2%) as a brown solid. [000851] 1H NMR: (400 MHz, DMSO-d6) δ 12.85 (s, 1H), 8.81 (d, J = 2.0 Hz, 1H), 8.50 (d, J = 2.1 Hz, 1H), 7.67 (d, J = 3.6 Hz, 1H), 6.61 (d, J = 3.5 Hz, 1H), 4.32 (d, J = 7.4 Hz, 2H), 2.82 (p, J = 7.7 Hz, 1H), 2.00 – 1.86 (m, 2H), 1.90 – 1.71 (m, 4H). [000852] Synthesis of 1-(cyclobutylmethyl)-N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5- yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide
[000853] Into a 40mL round-bottom flask were added 1-(cyclobutylmethyl)pyrrolo[2,3- b]pyridine-5-carboxylic acid (150 mg, 0.651 mmol, 1 equiv) and 3-(5-amino-1-oxo-3H-isoindol- 2-yl)piperidine-2,6-dione (202.67 mg, 0.781 mmol, 1.2 equiv) in DMF (10 mL) were added HATU (371.54 mg, 0.977 mmol, 1.5 equiv) and DIEA (168.39 mg, 1.302 mmol, 2 equiv) in portions at room temperature under air atmosphere. The resulting mixture was stirred for overnight at 60°C under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 30% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in 1-(cyclobutylmethyl)-N-[2-(2,6-dioxopiperidin-3-yl)- 1-oxo-3H-isoindol-5-yl]pyrrolo[2,3-b]pyridine-5-carboxamide (77.5 mg, 25.11%) as a white solid. [000854] LC-MS (ES, m/z): [M+H]+=472.2 [000855] 1H NMR: (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 10.61 (s, 1H), 8.86 (d, J = 2.1 Hz, 1H), 8.59 (d, J = 2.2 Hz, 1H), 8.17 (d, J = 1.7 Hz, 1H), 7.86 (dd, J = 8.4, 1.8 Hz, 1H), 7.76 – 7.67
(m, 2H), 6.64 (d, J = 3.5 Hz, 1H), 5.11 (dd, J = 13.3, 5.1 Hz, 1H), 4.48 (d, J = 17.2 Hz, 1H), 4.39 – 4.30 (m, 3H), 3.00 – 2.78 (m, 2H), 2.66 – 2.57 (m, 1H), 2.39-2.41 (m, 1H), 2.07 – 1.73 (m, 7H). [000856] Example 1.82. Synthesis of 1-(sec-butyl)-N-(2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindolin-5-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-123)
[000857] Synthesis of 1-(sec-butyl)-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000858] Into a 50 mL round-bottom flask were added methyl 1H-pyrrolo[2,3-b]pyridine-5- carboxylate (500 mg, 2.838 mmol, 1 equiv) and DMF (5 mL) at room temperature. To the above mixture was added NaH (170.27 mg, 4.257 mmol, 1.5 equiv, 60%) in DMF (5 mL) dropwise at 0 °C. The resulting mixture was stirred for additional 1 h at room temperature. To the above mixture was added 2-bromobutane (583.31 mg, 4.257 mmol, 1.5 equiv) dropwise at room temperature. The resulting mixture was stirred for additional overnight at room temperature. The reaction was quenched with Water and stirred for 3 h at 0 °C. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 1-(sec-butyl)pyrrolo[2,3-b]pyridine-5-carboxylic acid (258 mg, 39.57%) as a yellow oil. [000859] LC-MS (ES, m/z): [M+ H]+:219.10 [000860] 1H NMR: (400 MHz, DMSO-d6) δ 8.80 (s, 1H), 8.50 (s, 1H), 7.76 (d, J = 3.6 Hz, 1H), 6.65 (d, J = 3.6 Hz, 1H), 4.91 (h, J = 7.0 Hz, 1H), 1.97-1.78 (m, J = 7.1 Hz, 2H), 1.47 (d, J = 6.8 Hz, 3H), 0.69 (t, J = 7.3 Hz, 3H). [000861] Synthesis of 1-(sec-butyl)-N-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)-1H- pyrrolo[2,3-b]pyridine-5-carboxamide
[000862] Into a 20 mL vial were added 1-(sec-butyl)pyrrolo[2,3-b]pyridine-5-carboxylic acid (100 mg, 0.458 mmol, 1 equiv), 3-(5-amino-1-oxo-3H-isoindol-2-yl)piperidine-2,6-dione (118.79 mg, 0.458 mmol, 1 equiv) and DMF (2 mL) at room temperature. To the above mixture were added DIEA (177.65 mg, 1.374 mmol, 3 equiv) and HATU (261.32 mg, 0.687 mmol, 1.5 equiv) in portions at room temperature. The resulting mixture was stirred for additional overnight at room temperature. Desired product could be detected by LCMS. The reaction was quenched with Water (15 mL) at room temperature. The aqueous layer was extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (2 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10mmol/L NH4HCO3), 10% to 50% gradient in 10 min; detector, UV 254 nm. The crude product (116 mg) was purified by Prep-HPLC with the following conditions (column, C18 silica gel; mobile phase, MeOH in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm) to afford N-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-1-(sec-butyl)pyrrolo[2,3- b]pyridine-5-carboxamide (44.0 mg, 20.52%) as an off-white solid. [000863] LC-MS (ES, m/z): [M+H]+: 460.10 [000864] 1H NMR: (400 MHz, DMSO-d6)δ10.99 (s, 1H), 10.60 (s, 1H), 8.85 (d, J = 2.1 Hz, 1H),8.59 (d, J = 2.1 Hz, 1H), 8.18 (s, 1H), 7.89-7.78 (m, 2H), 7.72(d, J = 8.3 Hz, 1H), 6.69 (d, J = 3.6 Hz, 2H), 5.11(dd, J = 13.2, 5.1 Hz, 1H), 4.93(h, J = 6.9 Hz, 1H), 4.48(d, J = 17.2 Hz, 1H), 4.34(d, J = 17.2 Hz, 1H), 2.93(ddd, J = 17.9, 13.5, 5.3 Hz, 1H), 2.65-2.57(m, 1H), 2.40(qd, J =12.9, 4.2 Hz, 1H), 2.06-1.99(m, 3H), 1.99-1.81(m, J = 7.0 Hz, 1H), 1.50(d, J = 6.8 Hz, 3H), 0.69(t, J = 7.3 Hz, 3H). [000865] Example 1.83. Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro- 1-oxoisoindolin-5-yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (I-94)
[000866] Synthesis of methyl 1-cyclobutyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylate
[000867] Into a 100 mL round-bottom flask were added methyl 1H-pyrrolo[2,3-b]pyridine-5- carboxylate (1000 mg, 5.676 mmol, 1 equiv) and DMF (20 mL) at room temperature. To the above mixture was added Cs2CO3 (7397.62 mg, 22.704 mmol, 4.00 equiv), bromocyclobutane (2298.92 mg, 17.028 mmol, 3 equiv) at room temperature. The resulting mixture was stirred for additional 2h at 80°C under nitrogen atmosphere .Desired product could be detected by LCMS. The reaction was quenched with Water(100 ml) at room temperature. The resulting mixture was extracted with EtOAc (3 x 100mL). The combined organic layers were washed with brine (2x100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (5:1) to afford methyl 1-cyclobutylpyrrolo[2,3-b]pyridine-5-carboxylate (1000 mg, 76.51%) as a yellow semi-solid. [000868] LC-MS (ES, m/z): [M+H]+: 231 [000869] 1H NMR: (400 MHz, DMSO-d6) δ8.82 (d, J=2.1 Hz, 1H), 8.54 (d, J=2.1 Hz, 1H), 7.97 (d, J=3.6 Hz, 1H), 6.69 (d, J=3.6 Hz, 1H), 5.44 –5.22 (m, 1H), 3.88 (s, 3H), 2.61 –2.53 (m, 2H),2.43 (dtd, J = 10.4, 8.2, 4.4 Hz, 2H), 1.91 – 1.80 (m, 2H). [000870] Synthesis of 1-cyclobutyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[000871] Into a 25mL round-bottom flask were added methyl 1-cyclobutylpyrrolo[2,3- b]pyridine-5-carboxylate (460 mg, 1.998 mmol, 1 equiv) and THF (5.00 mL) at room
temperature. To the above mixture was added LiOH in H2O (1M/L, 8.00 mL, 7.992 mmol, 4.00 equiv) at room temperature. The resulting mixture was stirred for additional 3h at room temperature. Desired product could be detected by LCMS. The mixture was acidified to pH 1 with conc. HCl. The resulting mixture was extracted with EtOAc (3 x 20mL). The combined organic layers were washed with brine (3x10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 1-cyclobutylpyrrolo[2,3- b]pyridine-5-carboxylic acid (400 mg, 92.60%) as a white solid. [000872] LC-MS (ES, m/z): [M+H]+ : 217 [000873] 1H NMR: (300 MHz, DMSO-d6) δ 12.89 (s, 1H), 8.81 (d, J = 2.0 Hz, 1H), 8.51 (d, J = 2.0 Hz, 1H), 7.94 (d, J = 3.6 Hz, 1H), 6.67 (d, J = 3.6 Hz, 1H), 5.49 – 5.22 (m, 1H), 2.65 – 2.52 (m, 2H), 2.49 – 2.34 (m, 2H), 1.86 (tt, J = 10.4, 6.1 Hz, 2H). [000874] Synthesis of tert-butyl (2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1-oxoisoindolin-5- yl)carbamate
[000875] Into a 40mL vial were added 3-(5-bromo-6-fluoro-1-oxo-3H-isoindol-2-yl)piperidine- 2,6-dione (500 mg, 1.466 mmol, 1 equiv), Cs2CO3 (1910.20 mg, 5.864 mmol, 4 equiv), 1,4- dioxane (15 mL), Pd(OAc)2 (131.62 mg, 0.586 mmol, 0.4 equiv), dicyclohexyl({2-[2,4,6- tris(propan-2-yl)phenyl]phenyl})phosphane (279.49 mg, 0.586 mmol, 0.4 equiv), tert-butyl carbamate (686.80 mg, 5.864 mmol, 4 equiv) at room temperature. The resulting mixture was stirred for additional 2h at room temperature under argon atmosphere. Desired product could be detected by LCMS. The reaction was quenched with sat. citric acid (aq.) at room temperature. The resulting mixture was extracted with EtOAc (3 x 50mL). The combined organic layers were washed with brine (2x50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1:9) to afford tert-butyl N-[2-(2,6-dioxopiperidin-3-yl)-6- fluoro-1-oxo-3H-isoindol-5-yl]carbamate)( 470 mg) as a yellow solid. [000876] LC-MS (ES, m/z): [M+H]+ : 378 [000877] 1H NMR: (300 MHz, DMSO-d6) δ 10.99 (s, 1H), 9.34 (d, J = 1.5 Hz, 1H), 8.00 (d, J = 6.7 Hz, 1H), 7.53 (d, J = 9.6 Hz, 1H), 5.09 (dd, J = 13.3, 5.1 Hz, 1H), 4.46 – 4.21 (m, 2H), 2.91
(ddd, J = 17.2, 13.5, 5.3 Hz, 1H), 2.57 (s, 1H), 2.37 (qd, J = 13.1, 4.4 Hz, 1H), 2.05 – 2.00 (m, 1H), 1.49 (s, 9H). [000878] Synthesis of 3-(5-amino-6-fluoro-1-oxoisoindolin-2-yl)piperidine-2,6-dione
[000879] Into a 25mL round-bottom flask were added tert-butyl N-[2-(2,6-dioxopiperidin-3- yl)-6-fluoro-1-oxo-3H-isoindol-5-yl]carbamate (250 mg, 0.662 mmol, 1 equiv) and DCM (5 mL) at room temperature. To the above mixture was added HCl(gas)in 1,4-dioxane (4M/L, 5 mL) at room temperature. The resulting mixture was stirred for additional 4h at room temperature. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water , 0% to 60% gradient in 30 min; detector, UV 254 nm.This resulted in 3-(5-amino-6-fluoro-1-oxo-3H- isoindol-2-yl)piperidine-2,6-dione (120 mg, 65.33%) as a white solid. [000880] LC-MS (ES, m/z): [M+H]+ : 278 [000881] 1H NMR: (300 MHz, DMSO-d6) δ 10.94 (s, 1H), 7.27 (d, J = 10.4 Hz, 1H), 6.85 (d, J = 7.8 Hz, 1H), 5.89 (s, 2H), 5.02 (dd, J = 13.3, 5.1 Hz, 1H), 4.27 (d, J = 16.7 Hz, 1H), 4.13 (d, J = 16.7 Hz, 1H), 2.90 (ddd, J = 17.2, 13.6, 5.4 Hz, 1H), 2.57 (d, J = 17.6 Hz, 1H), 2.32 (qd, J = 13.2, 4.5 Hz, 1H), 1.95 (ddq, J = 10.4, 5.3, 3.1, 2.7 Hz, 1H). [000882] Synthesis of 1-cyclobutyl-N-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1-oxoisoindolin-5- yl)-1H-pyrrolo[2,3-b]pyridine-5-carboxamide
[000883] Into a 25mL 2-necked round-bottom flask were added 1-cyclobutylpyrrolo[2,3- b]pyridine-5-carboxylic acid (70 mg, 0.324 mmol, 1 equiv) and 3-(5-amino-6-fluoro-1-oxo-3H- isoindol-2-yl)piperidine-2,6-dione (89.75 mg, 0.324 mmol, 1 equiv), Py (1 ml) at room temperature. To the above mixture was added POCl3 (99.26 mg, 0.648 mmol, 2 equiv) at 0°C under argon atmosphere. The resulting mixture was stirred at 0°C for additional 2h. Desired
product could be detected by LCMS. The resulting mixture was diluted with NMP (5mL). The reaction was quenched with Water at room temperature. The residue was purified by reversed- phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water , 0% to 60% gradient in 30 min; detector, UV 254 nm. This resulted in 1- cyclobutyl-N-[2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1-oxo-3H-isoindol-5-yl]pyrrolo[2,3- b]pyridine-5-carboxamide (22.9 mg, 14.70%) as a yellow solid. [000884] LC-MS (ES, m/z): [M+H]+ : 476 [000885] 1H NMR: (400 MHz, DMSO-d6) δ 11.03 (s, 1H), 10.40 (s, 1H), 8.87 (d, J = 2.1 Hz, 1H), 8.61 (d, J = 2.2 Hz, 1H), 8.05 (d, J = 6.5 Hz, 1H), 7.97 (d, J = 3.6 Hz, 1H), 7.66 (d, J = 9.1 Hz, 1H),6.70 (d, J = 3.6 Hz, 1H), 5.38 (p, J = 8.6 Hz, 1H), 5.14 (dd, J = 13.3, 5.1 Hz, 1H), 4.49 (d, J = 17.3 Hz, 1H), 4.35 (d, J = 17.3 Hz, 1H), 2.93 (ddd, J = 18.1, 13.6, 5.3 Hz, 1H), 2.65 – 2.52 (m, 3H), 2.49 – 2.29 (m, 3H), 2.06 – 1.99 (m, 1H), 1.87 (tt, J = 10.2, 5.5 Hz, 2H). [000886] Example 1.84. Synthesis of N-(1-cyclobutyl-1H-pyrrolo[2,3-b]pyridin-5-yl)-2- (2,6-dioxopiperidin-3-yl)-6-fluoro-1-oxoisoindoline-5-carboxamide (I-93)
[000887] Synthesis of 2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1-oxoisoindoline-5-carboxylic acid
[000888] A mixture of 3-(5-bromo-6-fluoro-1-oxo-3H-isoindol-2-yl)piperidine-2,6-dione (400 mg, 1.173 mmol, 1 equiv), Pd(OAc)2 (88 mg, 0.392 mmol, 0.33 equiv), H2O (124 mg, 6.883 mmol, 5.87 equiv), DIEA (133 mg, 1.029 mmol, 0.88 equiv) and DPPP (126 mg, 0.305 mmol, 0.26 equiv) in DMF (5 mL) was stirred at 90°C for overnight under carbon monoxide atmosphere. The reaction was monitored by LCMS and TLC. The reaction was quenched with Water/Ice at room temperature. The resulting mixture was extracted with CH2Cl2 (3 x 70 mL). The combined organic layers were washed with brine (3x30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This
resulted in 2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1-oxo-3H-isoindole-5-carboxylic acid (350 mg, crude, 87.72%) as a brown yellow solid. [000889] LC-MS (ES, m/z): [M+H]+: 307 [000890] 1H NMR: (400 MHz, DMSO-d6) δ 11.02 (s, 1H), 7.90 (d, J = 6.0 Hz, 1H), 7.80 (dd, J = 11.8, 7.4 Hz, 1H), 7.53 – 7.44 (m, 2H), 5.13 (dd, J = 13.3, 5.1 Hz, 1H), 4.46 (d, J = 17.3 Hz, 1H), 4.33 (d, J = 17.3 Hz, 1H), 3.38 – 3.32 (m, 1H), 3.07 (d, J = 8.0 Hz, 3H), 2.98 – 2.84 (m, 1H), 2.65 – 2.52 (m, 1H), 2.39 (qd, J = 13.3, 4.4 Hz, 1H), 2.10 – 1.96 (m, 1H), 1.25 (d, J = 6.9 Hz, 20H), 0.84 (td, J = 9.3, 8.3, 5.9 Hz, 1H). [000891] Synthesis of N-(1-cyclobutyl-1H-pyrrolo[2,3-b]pyridin-5-yl)-2-(2,6-dioxopiperidin-3- yl)-6-fluoro-1-oxoisoindoline-5-carboxamide
[000892] To a stirred solution/mixture of 2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1-oxo-3H- isoindole-5-carboxylic acid (150 mg, 0.490 mmol, 1 equiv), HATU (140 mg, 0.368 mmol, 0.75 equiv) and DIEA (95 mg, 0.735 mmol, 1.50 equiv) in DMF (3 mL) was added 1- cyclobutylpyrrolo[2,3-b]pyridin-5-amine (55 mg, 0.294 mmol, 0.60 equiv) dropwise/ in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred at 60°C for 2 h under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. The reaction was quenched with Water/Ice at room temperature. The resulting mixture was extracted with CH2Cl2 (3 x 70 mL). The combined organic layers were washed with brine (3x30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 5% to 50% gradient in 25 min; detector, UV 254 nm to afford N-{1-cyclobutylpyrrolo[2,3-b]pyridin-5-yl}- 2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1-oxo-3H-isoindole-5-carboxamide (86 mg, 36.26%) as a light yellow solid. [000893] LC-MS (ES, m/z): [M+H]+ : 476
[000894] 1H NMR: (300 MHz, DMSO-d6) δ 11.04 (s, 1H), 10.64 (s, 1H), 8.42 (dd, J = 17.3, 2.3 Hz, 2H), 7.96 (d, J = 5.8 Hz, 1H), 7.82 (d, J = 3.5 Hz, 1H), 7.71 (d, J = 8.7 Hz, 1H), 6.53 (d, J = 3.5 Hz, 1H), 5.45 – 4.96 (m, 2H), 4.68 – 4.21 (m, 2H), 2.93 (ddd, J = 17.0, 13.5, 5.3 Hz, 1H), 2.74 – 2.52 (m, 2H), 2.48 – 2.24 (m, 3H), 2.13 – 2.02 (m, 1H), 1.85 (td, J = 9.9, 5.5 Hz, 2H). [000895] Example 1.85. Synthesis of N-(4-cyclobutoxypyrimidin-2-yl)-2-(2,6- dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxamide (I-98)
[000896] Synthesis of 2-chloro-4-cyclobutoxypyrimidine
[000897] A solution of cyclobutanol (532.44 mg, 7.384 mmol, 1.1 equiv) in THF (10 mL) was treated with NaH (402.73 mg, 10.069 mmol, 1.5 equiv, 60%) for 5min at room temperature under nitrogen atmosphere followed by the addition of 2-chloro-6-fluoropyridine (300 mg, 2.281 mmol, 1 equiv) at room temperature. The resulting mixture was stirred for 2 h at room temperature under argon atmosphere. The reaction was quenched with Water at room temperature. The resulting mixture was extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (3:1) to afford 2-chloro-4-cyclobutoxypyrimidine (750 mg, 60.52%) as a colorless oil. [000898] LC-MS (ES, m/z): [M+H]+:185.10 [000899] 1H NMR: (400 MHz, Chloroform-d) δ 8.27 (d, J = 5.8 Hz, 1H), 6.61 (d, J = 5.7 Hz, 1H), 5.23 (dt, J = 24.9, 7.4 Hz, 1H), 2.48 (ddtt, J = 12.6, 7.9, 5.4, 2.7 Hz, 2H), 2.18 (dddd, J = 20.1, 12.8, 10.0, 5.0 Hz, 2H), 1.87 (tdd, J = 10.3, 6.5, 2.7 Hz, 1H), 1.68 (dddd, J = 18.4, 15.5, 10.4, 7.9 Hz, 1H).
[000900] Synthesis of 4-cyclobutoxypyrimidin-2-amine
[000901] A solution of 2-chloro-6-cyclobutoxypyridine (750 mg, 4.084 mmol, 1 equiv) in NH3(g) in MeOH (12.50 mL,7 M/L) was stirred for 2 days at 90 °C under argon atmosphere. Desired product could be detected by LCMS. The mixture was allowed to cool down to room temperature. The resulting mixture was concentrated under vacuum. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 5% to 50% gradient in 30 min; detector, UV 254 nm. This resulted in 6-cyclobutoxypyridin-2-amine (160 mg, 23.86%) as a white solid. [000902] LC-MS (ES, m/z): [M+H]+ : 166.15 [000903] 1H NMR: (300 MHz, DMSO-d6) δ 7.94 (d, J = 5.6 Hz, 1H), 6.45 (s, 2H), 5.94 (d, J = 5.6 Hz, 1H), 5.20 – 4.94 (m, 1H), 2.43 – 2.28 (m, 2H), 2.12 – 1.93 (m, 2H), 1.84 – 1.48 (m, 2H). [000904] Synthesis of N-(4-cyclobutoxypyrimidin-2-yl)-2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindoline-5-carboxamide
[000905] Into a 25 mL round-bottom flask were added 4-cyclobutoxypyrimidin-2-amine (80 mg, 0.484 mmol, 1 equiv) and 2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5-carboxylic acid (139.60 mg, 0.484 mmol, 1 equiv), pyridine (1 mL) at room temperature. To the above mixture was added POCl3 (148.50 mg, 0.968 mmol, 2 equiv) at 0 °C. The resulting mixture was stirred for additional 2 h at 0 °C. Desired product could be detected by LCMS. The resulting mixture was diluted with EtOAc (30 mL). The reaction was quenched by the addition of water (20 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 30mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 0% to 40% gradient in 30 min; detector, UV 254 nm. The
product was purified by Prep-HPLC to afford N-(4-cyclobutoxypyrimidin-2-yl)-2-(2,6- dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5-carboxamide (7.7 mg, 3.62%) as a white solid. [000906] LC-MS (ES, m/z): [M+H]+ : 436.15 [000907] 1H NMR: (300 MHz, DMSO-d6) δ 11.00 (s, 2H), 8.41 (d, J = 5.7 Hz, 1H), 8.12 (s, 1H), 8.04 – 7.96 (m, 1H), 7.83 (d, J = 8.0 Hz, 1H), 6.61 (d, J = 5.7 Hz, 1H), 5.24 – 4.91 (m, 2H), 4.60 –4.33 (m, 2H), 3.03 – 2.82 (m, 1H), 2.61 (d, J = 17.3 Hz, 1H), 2.43 (d, J = 12.3 Hz, 3H), 2.15 – 1.96 (m, 3H), 1.76 (q, J = 10.2 Hz, 1H), 1.66 – 1.48 (m, 1H). [000908] Example 1.86. Synthesis of N-(4-(cyclobutylamino)pyrimidin-2-yl)-2-(2,6- dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxamide (I-96)
[000909] Synthesis of N4-cyclobutylpyrimidine-2,4-diamine
[000910] A solution of 4-chloropyrimidin-2-amine (300 mg, 2.316 mmol, 1 equiv), cyclobutylamine (413 mg, 5.807 mmol, 2.51 equiv) and TEA (705 mg, 6.967 mmol, 3.01 equiv) in ACN (3 mL) was stirred for overnight at 85 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 5% to 40% gradient in 25 min; detector, UV 254 nm to afford N4- cyclobutylpyrimidine-2,4-diamine (236 mg, 62.06%) as a white solid. [000911] LC-MS (ES, m/z): [M+H]+:165.15 [000912] 1H NMR: (300 MHz, DMSO-d6) δ 7.58 (d, J = 5.8 Hz, 1H), 7.13 (s, 1H), 5.91 (s, 2H), 5.64 (d, J = 5.8 Hz, 1H), 2.20 (ddt, J = 13.1, 7.5, 3.6 Hz, 2H), 1.85 (tt, J = 11.5, 8.9 Hz, 2H), 1.73 – 1.50 (m, 2H). [000913] Synthesis of N-(4-(cyclobutylamino)pyrimidin-2-yl)-2-(2,6-dioxopiperidin-3-yl)-1- oxoisoindoline-5-carboxamide
[000914] To a stirred solution of N4-cyclobutylpyrimidine-2,4-diamine (50 mg, 0.304 mmol, 1 equiv) and 2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5-carboxylic acid (88 mg, 0.305 mmol, 1.00 equiv) in Pyridine (1 mL) was added POCl3 (93 mg, 0.607 mmol, 1.99 equiv) dropwise at 0 °C. The resulting mixture was stirred for 2 h at room temperature. The reaction was quenched with water at room temperature. The crude product was purified by Prep-HPLC with the following conditions (Column: Xselect CSH Prep C18 OBD, 30*150mm, 5um; Mobile Phase A: Water (0.1% TFA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 5% B to 19% B in 8 min; Wave Length: 254nm/220nm nm; RT1(min): 8.95) to afford N-[4- (cyclobutylamino)pyrimidin-2-yl]-2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5- carboxamide (36.7 mg, 27.66%) as a white solid. [000915] LC-MS (ES, m/z): [M+H]+ : 435.1 [000916] 1H NMR: (300 MHz, DMSO-d6) δ 12.49 (s, 1H), 11.05 (s, 1H), 9.70 (d, J = 7.4 Hz, 1H), 8.27 (s, 1H), 8.15 (dd, J = 8.0, 1.5 Hz, 1H), 7.95 (dd, J = 9.3, 7.6 Hz, 2H), 6.54 (d, J = 7.2 Hz, 1H), 5.18 (dd, J = 13.2, 5.1 Hz, 1H), 4.77 – 4.21 (m, 3H), 2.95 (ddd, J = 17.8, 13.4, 5.3 Hz, 1H), 2.66 (t, J = 14.4 Hz, 1H), 2.49 – 2.25 (m, 3H), 2.24 – 1.88 (m, 3H), 1.87 – 1.60 (m, 2H). [000917] Example 1.87. Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(7-methoxy-5,6,7,8- tetrahydroquinolin-3-yl)-1-oxoisoindoline-5-carboxamide (I-108)
[000918] Synthesis of 7-methoxy-3-nitro-5,6,7,8-tetrahydroquinoline
[000919] Into a 40 mL sealed tube were added 1-methyl-3,5-dinitropyridin-2-one (500 mg, 2.511 mmol, 1 equiv), 3-methoxycyclohexan-1-one (321.84 mg, 2.511 mmol, 1 equiv) and NH3(g) in MeOH (7 mL) at room temperature. The resulting mixture was stirred for 1 h at 65°C. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1:1) to afford 7-methoxy-3-nitro-5,6,7,8-tetrahydroquinoline (100 mg, 19.13%) as an orange oil. [000920] LC-MS (ES, m/z): [M+H]+: 209.10 [000921] 1H NMR: (400 MHz, Chloroform-d) δ 9.20 (d, J = 2.5 Hz, 1H), 8.20 (dd, J = 2.4, 1.1 Hz, 1H), 3.93 – 3.84 (m, 1H), 3.42 (d, J = 0.9 Hz, 3H), 3.29 – 3.14 (m, 2H), 3.14 – 3.01 (m, 1H), 2.91 – 2.80 (m, 1H), 2.16 – 2.04 (m, 1H), 2.04 – 1.93 (m, 1H). [000922] Synthesis of 7-methoxy-5,6,7,8-tetrahydroquinolin-3-amine
[000923] Into a 50 mL round-bottom flask were added 7-methoxy-3-nitro-5,6,7,8- tetrahydroquinoline (100 mg, 0.480 mmol, 1 equiv), Pd/C (5 mg, 5%) and MeOH (4 mL) at room temperature. The resulting mixture was stirred for overnight at room temperature under hydrogen atmosphere. Desired product could be detected by LCMS. The resulting mixture was filtered, the filter cake was washed with MeOH (2 x 10 mL). The filtrate was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. [000924] LC-MS (ES, m/z): [M+H]+: 179.05 [000925] Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(7-methoxy-5,6,7,8-tetrahydroquinolin-3- yl)-1-oxoisoindoline-5-carboxamide
[000926] Into a 40 mL vial were added 2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5- carboxylic acid (161.73 mg, 0.561 mmol, 1 equiv), 7-methoxy-5,6,7,8-tetrahydroquinolin-3- amine (100 mg, 0.561 mmol, 1 equiv), DIEA (435.09 mg, 3.366 mmol, 6 equiv), DMF (5 mL) and HATU (320.00 mg, 0.842 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 25% to 30% gradient in 10 min; detector, UV 254 nm. This resulted in 2-(2,6-dioxopiperidin-3-yl)-N-(7-methoxy-5,6,7,8-tetrahydroquinolin-3-yl) - 1-oxo-3H-isoindole-5-carboxamide (138.9 mg, 52.61%) as an off-white solid. [000927] LC-MS (ES, m/z): [M+H]+: 449.00 [000928] 1H NMR: (400 MHz, DMSO-d6) δ 11.03 (s, 1H), 10.51 (s, 1H), 8.66 (d, J = 2.4 Hz, 1H), 8.18 (s, 1H), 8.10 (dd, J = 8.0, 1.5 Hz, 1H), 7.93 (d, J = 2.4 Hz, 1H), 7.89 (d, J = 8.0 Hz, 1H), 5.17 (dd, J = 13.2, 5.2 Hz, 1H), 4.57 (d, J = 17.6 Hz, 1H), 4.44 (d, J = 17.6 Hz, 1H), 3.80 – 3.71 (m, 1H), 3.31 (s, 3H), 3.06 (dd, J = 17.2, 4.7 Hz, 1H), 3.00 – 2.89 (m, 1H), 2.89 – 2.67 (m, 3H), 2.67 – 2.58 (m, 1H), 2.49 – 2.37 (m, 1H), 2.10 – 2.01 (m, 1H), 2.01 – 1.90 (m, 1H), 1.90 – 1.78 (m, 1H). [000929] Example 1.88. Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(5-methoxy-5,6,7,8- tetrahydroquinolin-3-yl)-1-oxoisoindoline-5-carboxamide (I-107)
[000930] Synthesis of 5-methoxy-3-nitro-5,6,7,8-tetrahydroquinoline
[000931] Into a 40 mL sealed tube were added 1-methyl-3,5-dinitropyridin-2-one (500 mg, 2.511 mmol, 1 equiv), 3-methoxycyclohexan-1-one (321.84 mg, 2.511 mmol, 1 equiv) and NH3(g) in MeOH (7 mL) at room temperature. The resulting mixture was stirred for 1 h at 65°C under nitrogen atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1:1) to afford 5-methoxy-3-nitro-5,6,7,8- tetrahydroquinoline (20 mg, 3.83%) as lightless oil. [000932] LC-MS (ES, m/z): [M+H]+: 209.25 [000933] 1H NMR: (400 MHz, Chloroform-d) δ 9.26 (d, J = 2.4 Hz, 1H), 8.54 (t, J = 2.3 Hz, 1H), 4.41 (d, J = 7.6 Hz, 1H), 3.53 (d, J = 2.0 Hz, 3H), 3.17 – 3.03 (m, 2H), 2.19 – 2.06 (m, 2H), 2.00 – 1.81 (m, 2H). [000934] Synthesis of 5-methoxy-5,6,7,8-tetrahydroquinolin-3-amine
[000935] Into a 25 mL round-bottom flask were added 5-methoxy-3-nitro-5,6,7,8- tetrahydroquinoline (20 mg, 0.096 mmol, 1 equiv), Pd/C (2 mg, 10%) and MeOH (1 mL) at
room temperature. The resulting mixture was stirred for overnight at room temperature under hydrogen atmosphere. Desired product could be detected by LCMS. The resulting mixture was filtered, the filter cake was washed with MeOH (2 x 7 mL). The filtrate was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. [000936] LC-MS (ES, m/z): [M+H]+: 179.10 [000937] Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(5-methoxy-5,6,7,8-tetrahydroquinolin-3- yl)-1-oxoisoindoline-5-carboxamide
[000938] Into a 10 mL vial were added 2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5- carboxylic acid (40.43 mg, 0.140 mmol, 1 equiv), 5-methoxy-5,6,7,8-tetrahydroquinolin-3-amine (25 mg, 0.140 mmol, 1 equiv), DIEA (108.77 mg, 0.840 mmol, 6 equiv), DMF (3 mL) and HATU (80.00 mg, 0.210 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 15% to 22% gradient in 10 min; detector, UV 254 nm. This resulted in 2-(2,6-dioxopiperidin-3-yl)-N-(5- methoxy-5,6,7,8-tetrahydroquinolin-3-yl)-1 -oxo-3H-isoindole-5-carboxamide (36.1 mg, 55.84%) as a white solid. [000939] LC-MS (ES, m/z): [M+H]+: 449.05 [000940] 1H NMR: (400 MHz, DMSO-d6) δ 11.02 (s, 1H), 10.57 (s, 1H), 8.79 (d, J = 2.5 Hz, 1H), 8.20 (s, 1H), 8.18 – 8.08 (m, 2H), 7.89 (d, J = 7.9 Hz, 1H), 5.17 (dd, J = 13.3, 5.1 Hz, 1H), 4.57 (d, J = 17.6 Hz, 1H), 4.44 (d, J = 17.6 Hz, 1H), 4.37 (dd, J = 6.0, 3.6 Hz, 1H), 3.40 (s, 3H), 3.00 – 2.85 (m, 1H), 2.85 – 2.68 (m, 2H), 2.67 – 2.57 (m, 1H), 2.49 – 2.37 (m, 1H), 2.10 – 2.01 (m, 1H), 1.99 – 1.89 (m, 2H), 1.89 – 1.70 (m, 2H). [000941] Example 1.89. Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(4-methylpyridin-3-yl)- 1-oxoisoindoline-5-carboxamide (I-109)
[000942] Into a 40 mL vial were added 2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5- carboxylic acid (100 mg, 0.347 mmol, 1 equiv), 4-methylpyridin-3-amine (37.5 mg, 0.347 mmol, 1 equiv), DIEA (269.02 mg, 2.082 mmol, 6 equiv), DMF (4 mL) and HATU (197.86 mg, 0.520 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 2-(2,6-dioxopiperidin-3-yl)-N-(4-methylpyridin-3- yl)-1-oxo-3H-isoindole-5-carboxamide (26.0 mg, 19.49%) as an off-white solid. [000943] LC-MS (ES, m/z): [M+H]+: 378.95 [000944] 1H NMR: (400 MHz, DMSO-d6) δ 11.02 (s, 1H), 10.30 (s, 1H), 8.50 (s, 1H), 8.34 (d, J = 4.9 Hz, 1H), 8.21 (s, 1H), 8.13 (d, J = 8.0 Hz, 1H), 7.90 (d, J = 7.9 Hz, 1H), 7.35 (d, J = 4.9 Hz, 1H), 5.17 (dd, J = 13.3, 5.1 Hz, 1H), 4.57 (d, J = 17.6 Hz, 1H), 4.45 (d, J = 17.6 Hz, 1H), 3.00 – 2.87 (m, 1H), 2.71 – 2.57 (m, 1H), 2.49 – 2.37 (m, 1H), 2.27 (s, 3H), 2.10 – 2.01 (m, 1H). [000945] Example 1.90. Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(6-methyl-5,6,7,8- tetrahydroquinolin-3-yl)-1-oxoisoindoline-5-carboxamide (I-106)
[000946] Synthesis of 6-methyl-3-nitro-5,6,7,8-tetrahydroquinoline
[000947] Into a 25 mL sealed tube were added 1-methyl-3,5-dinitropyridin-2-one (500 mg, 2.511 mmol, 1 equiv), 4-methylcyclohexanone (281.67 mg, 2.511 mmol, 1 equiv) and NH3(g) in MeOH (5 mL) at room temperature. The resulting mixture was stirred at 65 °C for 2 h under nitrogen atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (2/1) to afford 6-methyl-3-nitro-5,6,7,8- tetrahydroquinoline (450 mg, 93.23%) as a yellow solid. [000948] LC-MS (ES, m/z): [M+H]+: 193.25 [000949] 1H NMR: (400 MHz, Chloroform-d) δ 9.17 (d, J = 2.6 Hz, 1H), 8.18 – 8.12 (m, 1H), 3.20 – 3.09 (m, 1H), 3.08 – 2.90 (m, 2H), 2.59 – 2.47 (m, 1H), 2.11 – 2.02 (m, 1H), 2.02 – 1.88 (m, 1H), 1.64 – 1.49 (m, 1H), 1.13 (d, J = 6.5 Hz, 3H). [000950] Synthesis of 6-methyl-5,6,7,8-tetrahydroquinolin-3-amine
[000951] Into a 25 mL round-bottom flask were added 6-methyl-3-nitro-5,6,7,8- tetrahydroquinoline (450 mg, 2.341 mmol, 1 equiv), Fe (522.95 mg, 9.364 mmol, 4 equiv), NH4Cl (500.90 mg, 9.364 mmol, 4 equiv), EtOH (5 mL) and H2O (5 mL) at room temperature. The resulting mixture was stirred at room temperature for overnight under nitrogen atmosphere. Desired product could be detected by LCMS. The resulting mixture was filtered and the filter cake was washed with MeOH (3 x 10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1/2) to afford 6-methyl-5,6,7,8-tetrahydroquinolin-3-amine (320 mg, 84.25%) as a yellow solid. [000952] LC-MS (ES, m/z): [M+H]+: 163.30 [000953] Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(6-methyl-5,6,7,8-tetrahydroquinolin-3- yl)-1-oxoisoindoline-5-carboxamide
[000954] Into a 25 mL vial were added 2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5- carboxylic acid (100 mg, 0.347 mmol, 1 equiv), 6-methyl-5,6,7,8-tetrahydroquinolin-3-amine (56.28 mg, 0.347 mmol, 1 equiv) dissolved in DMF (4 mL). DIEA (269.02 mg, 2.082 mmol, 6 equiv) was added dropwise to the reaction. Then, HATU (197.86 mg, 0.520 mmol, 1.5 equiv) was added to the reaction at room temperature. The resulting mixture was stirred at room temperature for overnight under nitrogen atmosphere. Desired product could be detected by LCMS. The reaction was quenched with water (3 mL). The reaction mixture was extracted with EA (30 mL). The organic layer was washed with water (3 x 10 mL), dried over anhydrous Na2SO4. After filtrated, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 25% to 30% gradient in 10 min; detector, UV 254 nm. This resulted in 2-(2,6-dioxopiperidin-3-yl)-N-(6-methyl-5,6,7,8- tetrahydroquinolin-3-yl)-1 -oxo-3H-isoindole-5-carboxamide (112.8 mg, 73.23%) as a white solid. [000955] LC-MS (ES, m/z): [M+H]+: 433.05 [000956] 1H NMR: (300 MHz, DMSO-d6) δ 11.03 (s, 1H), 10.51 (s, 1H), 8.65 (d, J = 2.4 Hz, 1H), 8.17 (d, J = 6.5 Hz, 1H), 8.10 (dd, J = 8.0, 1.5 Hz, 1H), 7.93 – 7.84 (m, 2H), 5.17 (dd, J = 13.2, 5.1 Hz, 1H), 4.57 (d, J = 17.7 Hz, 1H), 4.44 (d, J = 17.6 Hz, 1H), 3.03 – 2.76 (m, 4H), 2.68 – 2.56 (m, 1H), 2.48 – 2.34 (m, 2H), 2.10 – 1.97 (m, 1H), 1.97 – 1.75 (m, 2H), 1.56 – 1.36 (m, 1H), 1.05 (d, J = 6.4 Hz, 3H). [000957] Example 1.91. Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(1-methyl-1H- pyrrolo[2,3-b]pyridin-5-yl)-1-oxoisoindoline-5-carboxamide (I-110)
[000958] Into a 25 mL round-bottom flask were added 2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H- isoindole-5-carboxylic acid (100 mg, 0.347 mmol, 1 equiv), 1-methylpyrrolo[2,3-b]pyridin-5- amine (51.06 mg, 0.347 mmol, 1 equiv), DIEA (134.51 mg, 1.041 mmol, 3 equiv), DMF (5 mL, 64.608 mmol, 186.24 equiv) ) and HATU (197.86 mg, 0.520 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for 16 h at 60 °C under nitrogen atmosphere. The reaction was quenched with water (3 mL). The reaction mixture was extracted with EA (30 mL). The organic layer was washed with water (3 x 10 mL), dried over anhydrous Na2SO4. After filtrated, the filtrate was concentrated under reduced pressure. The mixture was purified by Prep- HPLC with the following conditions (Column: Xselect CSH C18 OBD Column 30*150mm 5μm, n; Mobile Phase A: Water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 7% B to 27% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 10.93) to afford 2-(2,6-dioxopiperidin-3-yl)-N-{1-methylpyrrolo[2,3-b]pyridin-5-yl}-1-oxo-3H- isoindole-5-carboxamide (71.6 mg, 47.47%) as an off-white solid. [000959] LC-MS (ES, m/z):[M+H]+: 418.00 [000960] 1H NMR: (400 MHz, DMSO-d6) δ 11.03 (s, 1H), 10.53 (s, 1H), 8.54 (d, J = 2.4 Hz, 1H), 8.40 (d, J = 2.3 Hz, 1H), 8.22 (s, 1H), 8.14 (dd, J = 7.9, 1.5 Hz, 1H), 7.89 (d, J = 8.0 Hz, 1H), 7.53 (d, J = 3.4 Hz, 1H), 6.49 (d, J = 3.4 Hz, 1H), 5.17 (m, 1H), 4.57 (d, J = 17.5 Hz, 1H), 4.45 (d, J = 17.5 Hz, 1H), 3.83 (s, 3H), 2.94 (m, 1H), 2.67 – 2.58 (m, 1H), 2.43 (m, 1H), 2.07 (d, J = 3.6 Hz, 1H). [000961] Example 1.92. Synthesis of N-(1-cyclobutyl-1H-pyrrolo[2,3-b]pyridin-5-yl)-2- (2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxamide (I-105)
[000962] Synthesis of 1-cyclobutyl-5-nitro-1H-pyrrolo[2,3-b]pyridine
[000963] To a stirred mixture of 5-nitro-1H-pyrrolo[2,3-b]pyridine (200 mg, 1.226 mmol, 1 equiv) and Cs2CO3 (798.89 mg, 2.452 mmol, 2 equiv) in DMF (4 mL) was added iodocyclobutane (245.44 mg, 1.349 mmol, 1.1 equiv) dropwise at room temperature under argon atmosphere. The resulting mixture was stirred at 90°C for overnight under argon atmosphere. The resulting mixture was filtered and the filter cake was washed with DMF (1 mL). The filtrate was combined and concentrated under reduced pressure. The residue was purified by reversed- phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 30% to 90% gradient in 30 min; detector, UV 254 nm. This resulted in 1-cyclobutyl-5-nitropyrrolo[2,3-b]pyridine (180 mg, 67.59%) was obtained as a yellow oil. [000964] 1H NMR: (400 MHz, Chloroform-d) δ 9.21 (d, J = 2.5 Hz, 1H), 8.75 (d, J = 2.4 Hz, 1H), 7.60 (d, J = 3.6 Hz, 1H), 6.69 (d, J = 3.6 Hz, 1H), 5.43 (m, 1H), 2.63 – 2.45 (m, 4H), 2.03 – 1.79 (m, 2H). [000965] Synthesis of 1-cyclobutyl-1H-pyrrolo[2,3-b]pyridin-5-amine
[000966] To a solution of 1-cyclobutyl-5-nitropyrrolo[2,3-b]pyridine (180 mg, 0.829 mmol, 1 equiv) in 10 mL MeOH was added Pd/C (10%, 36 mg) in a 50 mL round-bottom flask. The reaction was stirred at room temperature for 2 h under H2 atmosphere. Desired product was detected by LCMS. The reaction mixture was filtered through a Celite pad. The filtrate was combined and concentrated under reduced pressure. The crude product 1-cyclobutylpyrrolo[2,3- b]pyridin-5-amine (140 mg, 90.23%) was obtained as a brown solid. [000967] 1H NMR: (400 MHz, DMSO-d6) δ 7.73 (d, J = 2.5 Hz, 1H), 7.54 (d, J = 3.5 Hz, 1H), 7.07 (d, J = 2.5 Hz, 1H), 6.20 (d, J = 3.5 Hz, 1H), 5.14 (m, 1H), 4.68 (s, 2H), 2.49 – 2.42 (m, 2H), 2.37 (m, 2H), 1.85 – 1.72 (m, 2H). [000968] Synthesis of N-(1-cyclobutyl-1H-pyrrolo[2,3-b]pyridin-5-yl)-2-(2,6-dioxopiperidin-3- yl)-1-oxoisoindoline-5-carboxamide
[000969] To a stirred mixture of 1-cyclobutylpyrrolo[2,3-b]pyridin-5-amine (103.93 mg, 0.556 mmol, 2 equiv) and 2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5-carboxylic acid (80 mg, 0.278 mmol, 1.00 equiv) in DMF (2 mL) were added HATU (316.58 mg, 0.834 mmol, 3 equiv) and DIPEA (0.15 mL, 0.834 mmol, 3 equiv) at 0 °C under argon atmosphere. The resulting mixture was stirred for overnight at room temperature under argon atmosphere. Desired product could be detected by LCMS. The reaction was quenched with water (3 mL). The reaction mixture was extracted with EA (30 mL). The organic layer was washed with water (3 x 10 mL), dried over anhydrous Na2SO4. After filtrated, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 20% to 95% gradient in 30 min; detector, UV 254 nm. The product N-{1-cyclobutylpyrrolo[2,3-b]pyridin-5-yl}-2-(2,6- dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5-carboxamide (34.5 mg, 98.4%) was obtained as a white solid. [000970] LC-MS (ES, m/z):[M+H]+: 458.10 [000971] 1H NMR: (400 MHz, DMSO-d6) δ 11.02 (s, 1H), 10.52 (s, 1H), 8.51 (d, J = 2.3 Hz, 1H), 8.38 (d, J = 2.3 Hz, 1H), 8.21 (s, 1H), 8.17 – 8.10 (m, 1H), 7.89 (d, J = 7.9 Hz, 1H), 7.81 (d, J = 3.5 Hz, 1H), 6.53 (d, J = 3.5 Hz, 1H), 5.29 (m, 1H), 5.17 (m, 1H), 4.57 (d, J = 17.6 Hz, 1H), 4.45 (d, J = 17.6 Hz, 1H), 2.94 (m, 1H), 2.71 – 2.51 (m, 3H), 2.48 – 2.38 (m, 3H), 2.09 – 2.01 (m, 1H), 1.86 (m, 2H). [000972] Example 1.93. Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(1-isopropyl-1H- pyrrolo[2,3-b]pyridin-5-yl)-1-oxoisoindoline-5-carboxamide (I-104)
[000973] Synthesis of 1-isopropyl-5-nitro-1H-pyrrolo[2,3-b]pyridine
[000974] To a stirred mixture of 5-nitro-1H-pyrrolo[2,3-b]pyridine (500 mg, 3.065 mmol, 1 equiv) and Cs2CO3 (1497.92 mg, 4.598 mmol, 1.5 equiv) in DMF (12 mL) was added 2- iodopropane (573.12 mg, 3.372 mmol, 1.1 equiv) dropwise at room temperature under argon atmosphere. The resulting mixture was stirred for overnight at 80 °C. Desired product could be detected by LCMS. The reaction was quenched with H2O (30 mL). The reaction mixture was extracted with EtOAc (50 mL). The combined organic layers were washed with brine (3 x 20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was dissolved in DMF (1 mL) and purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water, 10% to 80% gradient in 25 min; detector, UV 254 nm to afford 1-isopropyl-5- nitropyrrolo[2,3-b]pyridine (500 mg, 79.49%) as an off-white solid. [000975] LC-MS (ES, m/z): [M +H]+: 206.05 [000976] 1H NMR: (400 MHz, DMSO-d6) δ 9.13 (d, J = 2.5 Hz, 1H), 8.88 (d, J = 2.5 Hz, 1H), 7.98 (d, J = 3.6 Hz, 1H), 6.80 (d, J = 3.6 Hz, 1H), 5.20-5.13 (m, 1H), 1.51 (d, J = 6.8 Hz, 6H). [000977] Synthesis of 1-isopropyl-1H-pyrrolo[2,3-b]pyridin-5-amine
[000978] Into a 25 mL round-bottom flask were added 1-isopropyl-5-nitropyrrolo[2,3- b]pyridine (200 mg, 0.975 mmol, 1 equiv) and MeOH (4 mL) at 25 °C. Then Pd/C (20.74 mg, 0.195 mmol, 0.2 equiv) was added. The resulting mixture was stirred for 2 hours at room temperature under hydrogen balloon atmosphere. Desired product was detected by LCMS. The mixture was filtered and the filter cake was washed with MeOH (10 mL). The organic layer was combined and concentrated under reduced pressure. The crude product was obtained and directly used for the next step. [000979] LC-MS (ES, m/z): [M +H]+:176.10 [000980] 1H NMR: (400 MHz, DMSO-d6) δ 7.74 (d, J = 2.5 Hz, 1H), 7.40 (d, J = 3.4 Hz, 1H), 7.08 (d, J = 2.5 Hz, 1H), 6.17 (d, J = 3.4 Hz, 1H), 4.92 (h, J = 6.8 Hz, 1H), 4.66 (s, 2H), 1.41 (d, J = 6.7 Hz, 6H). [000981] Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(1-isopropyl-1H-pyrrolo[2,3-b]pyridin-5- yl)-1-oxoisoindoline-5-carboxamide
[000982] A mixture of 1-isopropylpyrrolo[2,3-b]pyridin-5-amine (60 mg, 0.342 mmol, 1 equiv), 2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5-carboxylic acid (88.83 mg, 0.308 mmol, 0.90 equiv), HATU (195.29 mg, 0.513 mmol, 1.5 equiv) and DIEA (132.76 mg, 1.026 mmol, 3 equiv)in DMF (5 mL) was stirred 2 hours at 60°C under argon atmosphere. The reaction was monitored by LCMS. The resulting mixture was dissolved in DMF (5 mL). The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 10% to 80% gradient in 25 min; detector, UV 254 nm to afford to afford 2-(2,6-dioxopiperidin-3-yl)-N-{1-isopropylpyrrolo[2,3-b]pyridin-5-yl}-1-oxo- 3H-isoindole-5-carboxamide (115.2 mg, 70.77%) as a white solid. [000983] LC-MS (ES, m/z): [M -H]-: 444.20 [000984] 1H NMR: (400 MHz, DMSO-d6) δ 11.03 (s, 1H), 10.52 (s, 1H), 8.51 (d, J = 2.3 Hz, 1H), 8.37 (d, J = 2.3 Hz, 1H), 8.22 (s, 1H), 8.17 – 8.10 (m, 1H), 7.89 (d, J = 7.9 Hz, 1H), 7.67 (d, J = 3.5 Hz, 1H), 6.50 (d, J = 3.5 Hz, 1H), 5.17 (dd, J = 13.3, 5.1 Hz, 1H), 5.07 (hept, J = 6.9 Hz, 1H), 4.57 (d, J = 17.5 Hz, 1H), 4.45 (d, J = 17.6 Hz, 1H), 2.94 (ddd, J = 17.1, 13.5, 5.4 Hz, 1H),
2.62 (d, J = 17.1 Hz, 1H), 2.43 (td, J = 13.2, 4.5 Hz, 1H), 2.09 – 2.04 (m, 1H), 1.48 (d, J = 6.8 Hz, 6H). [000985] Example 1.94. Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(1-isopropyl-3-methyl- 1H-pyrazolo[3,4-d]pyrimidin-6-yl)-1-oxoisoindoline-5-carboxamide (I-102)
[000986] Synthesis of 1-isopropyl-3-methyl-1H-pyrazolo[3,4-d]pyrimidin-6-amine
[000987] Into a 8mL vial were added 6-chloro-1-isopropyl-3-methylpyrazolo[3,4-d]pyrimidine (100 mg, 0.475 mmol, 1 equiv) in NH3(g) in MeOH (2 mL). The resulting mixture was stirred for 1 overnight at 90°C. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. This resulted in1-isopropyl-3-methylpyrazolo[3,4- d]pyrimidin-6-amine (120 mg, crude) as a solid. [000988] LC-MS (ES, m/z): [M+H]+: 192.05 [000989] Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(1-isopropyl-3-methyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)-1-oxoisoindoline-5-carboxamide
[000990] Into a 8 mL vial were added 1-isopropyl-3-methylpyrazolo[3,4-d]pyrimidin-6-amine (60 mg, 0.314 mmol, 1 equiv), 2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5-carboxylic acid (90.44 mg, 0.314 mmol, 1 equiv) and Pyridine (2 mL) at room temperature. To the above mixture was added POCl3 (144.31 mg, 0.942 mmol, 3 equiv) dropwise at room temperature. The resulting mixture was stirred for additional 1 h at room temperature. Desired product could be detected by LCMS. The crude product was purified by Prep-HPLC with the following conditions
(Column: XBridge Prep Phenyl OBD Column 19*250 mm, 5m; Mobile Phase A: Water(0.1% FA), Mobile Phase B: MEOH; Flow rate: 60 mL/min mL/min; Gradient: 35% B to 50 % B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 10.281) to afford 2-(2,6-dioxopiperidin-3-yl)- N-{1-isopropyl-3-methylpyrazolo[3,4-d]pyrimidin-6-yl}-1-oxo-3H-isoindole-5-carboxamide (19.7 mg, 13.29%) as a white solid. [000991] LC-MS (ES, m/z): [M+H]+: 462.05 [000992] 1H NMR: (400 MHz, DMSO-d6) δ 11.28 (s, 1H), 11.02 (s, 1H), 9.19 (s, 1H), 8.16 (s, 1H), 8.04 (dd, J = 7.9, 1.5 Hz, 1H), 7.84 (d, J = 7.9 Hz, 1H), 5.16 (dd, J = 13.3, 5.1 Hz, 1H), 4.92 (hept, J = 6.6 Hz, 1H), 4.54 (d, J = 17.6 Hz, 1H), 4.42 (d, J = 17.6 Hz, 1H), 2.93 (ddd, J = 17.3, 13.6, 5.4 Hz, 1H), 2.62 (dd, J = 17.0, 3.7 Hz, 1H), 2.55 (s, 3H), 2.43 (td, J = 13.4, 4.8 Hz, 1H), 2.09 – 2.00 (m, 1H), 1.44 (d, J = 6.7 Hz, 6H). [000993] Example 1.95. Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(1-isopropyl-1H- pyrazolo[3,4-d]pyrimidin-6-yl)-1-oxoisoindoline-5-carboxamide (I-103)
[000994] Synthesis of 6-chloro-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidine
[000995] A solution of propan-2-ylhydrazine hydrochloride (343.67 mg, 3.108 mmol, 1.1 equiv) and TEA (857.66 mg, 8.475 mmol, 3 equiv) in MeOH (5 mL) was stirred for 10 min at 0°C under argon atmosphere. To the above mixture was added 2,4-dichloropyrimidine-5- carbaldehyde (500 mg, 2.825 mmol, 1 equiv) in MeOH (5 mL) dropwise at 0°C. The resulting
mixture was stirred for additional overnight at room temperature. Desired product could be detected by LCMS. The reaction was quenched with water at room temperature. The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (3:1) to afford 6-chloro-1-isopropylpyrazolo[3,4-d]pyrimidine (400 mg, 72.00%) as a yellow solid. [000996] LC-MS (ES, m/z) [M+H]+: 197.1 [000997] 1H NMR: (400 MHz, DMSO-d6) δ 9.26 (s, 1H), 8.45 (d, J = 0.6 Hz, 1H), 5.09 (hept, J = 6.6 Hz, 1H), 1.50 (d, J = 6.7 Hz, 6H). [000998] Synthesis of 1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-6-amine
[000999] Into a 20 mL sealed tube were added 6-chloro-1-isopropylpyrazolo[3,4-d]pyrimidine (200 mg, 1.017 mmol, 1 equiv) and NH3(g) in MeOH (5 mL) at room temperature. The mixture was stirred for overnight at 90°C under argon atmosphere. Desired product could be detected by LCMS. The resulting mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was dissolved in DMSO (2 mL). The residue was purified by reversed- phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 10% to 60% gradient in 20 min; detector, UV 254 nm. This resulted in 1- isopropylpyrazolo[3,4-d]pyrimidin-6-amine (150 mg, 83.22%) as a white solid. [0001000] LC-MS (ES, m/z) [M+H]+: 178.10 [0001001] 1H NMR: (400 MHz, DMSO-d6) δ 8.75 (s, 1H), 7.92 (d, J = 0.6 Hz, 1H), 6.82 (s, 2H), 4.92 – 4.79 (m, 1H), 1.42 (d, J = 6.7 Hz, 6H). [0001002] Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(1-isopropyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)-1-oxoisoindoline-5-carboxamide
[0001003] To a stirred mixture of 1-isopropylpyrazolo[3,4-d]pyrimidin-6-amine (60 mg, 0.339 mmol, 1 equiv) and 2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5-carboxylic acid (97.60 mg, 0.339 mmol, 1 equiv) in pyridine (2 mL) was added POCl3 (259.55 mg, 1.695 mmol, 5 equiv) dropwise at 0°C under argon atmosphere. The mixture was stirred for 2 h at room temperature under argon atmosphere. Desired product could be detected by LCMS. The resulting mixture was quenched with water. The reaction was extract with EA. The organic layer was combined and concentrated under reduced pressure. The residue was dissolved in DMF (2 mL). The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 30% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in 2-(2,6-dioxopiperidin-3-yl)-N-(1-isopropylpyrazolo[3,4- d]pyrimidin-6-yl-1-oxo-3H-isoindole-5-carboxamide (16.7 mg, 10.90%) as a white solid. [0001004] LC-MS (ES, m/z) [M+H]+: 448.05 [0001005] 1H NMR: (400 MHz, DMSO-d6) δ 11.33 (s, 1H), 11.02 (s, 1H), 9.22 (s, 1H), 8.29 (s, 1H), 8.17 (s, 1H), 8.05 (dd, J = 8.0, 1.5 Hz, 1H), 7.84 (d, J = 7.9 Hz, 1H), 5.16 (dd, J = 13.3, 5.1 Hz, 1H), 5.00 (hept, J = 6.6 Hz, 1H), 4.55 (d, J = 17.6 Hz, 1H), 4.42 (d, J = 17.6 Hz, 1H), 3.00 – 2.87 (m, 1H), 2.62 (dd, J = 16.5, 3.4 Hz, 1H), 2.43 (td, J = 13.1, 4.4 Hz, 1H), 2.05 (dd, J = 9.1, 3.8 Hz, 1H), 1.51 – 1.45 (m, 6H). [0001006] Example 1.96. Synthesis of N-(1-cyclobutyl-3-methyl-1H-pyrrolo[2,3-b]pyridin- 5-yl)-2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxamide (I-101)
[0001007] Synthesis of 1-cyclobutyl-3-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine
[0001008] Into a 40 mL vial were added 3-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine (300 mg, 1.693 mmol, 1 equiv), Cs2CO3 (827.59 mg, 2.540 mmol, 1.5 equiv), DMF (5 mL) and iodocyclobutane (369.84 mg, 2.032 mmol, 1.2 equiv) at room temperature. The resulting mixture was stirred for overnight at 80°C under nitrogen atmosphere. Desired product could be detected by LCMS. The reaction was quenched with water (3 mL). The reaction mixture was extracted with EA (30 mL). The organic layer was washed with water (3 x 10 mL), dried over anhydrous Na2SO4. After filtrated, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH4HCO3), 95% to 100% gradient in 10 min; detector, UV 254 nm. This resulted in 1-cyclobutyl-3-methyl-5-nitropyrrolo[2,3-b]pyridine (350 mg, 89.38%) as a yellow solid. [0001009] LC-MS (ES, m/z): [M+H]+: 232.10 [0001010] 1H NMR: (400 MHz, Chloroform-d) δ 9.17 (d, J = 2.4 Hz, 1H), 8.67 (dd, J = 2.5, 1.2 Hz, 1H), 7.36 (q, J = 1.2 Hz, 1H), 5.44 – 5.31 (m, 1H), 2.62 – 2.41 (m, 4H), 2.37 (d, J = 1.2 Hz, 3H), 2.03 – 1.86 (m, 2H). [0001011] Synthesis of 1-cyclobutyl-3-methyl-1H-pyrrolo[2,3-b]pyridin-5-amine
[0001012] Into a 25 mL round-bottom flask were added 1-cyclobutyl-3-methyl-5- nitropyrrolo[2,3-b]pyridine (300 mg, 1.297 mmol, 1 equiv), NH4Cl (277.56 mg, 5.188 mmol, 4 equiv), EtOH (5 mL), H2O (5 mL) and Fe (289.78 mg, 5.188 mmol, 4 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The resulting mixture was filtered and the filter cake was washed with MeOH (2 x 10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by
reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH4HCO3), 30% to 40% gradient in 10 min; detector, UV 254 nm. This resulted in 1-cyclobutyl-3-methylpyrrolo[2,3-b]pyridin-5-amine (240 mg, 91.92%) as a brown solid. [0001013] LC-MS (ES, m/z): [M+H]+: 202.05 [0001014] 1H NMR: (400 MHz, Chloroform-d) δ 7.91 (d, J = 2.5 Hz, 1H), 7.21 (d, J = 2.5 Hz, 1H), 7.13 (d, J = 1.2 Hz, 1H), 5.31 – 5.18 (m, 1H), 2.56 – 2.46 (m, 2H), 2.46 – 2.32 (m, 2H), 2.25 (d, J = 1.1 Hz, 3H), 1.92 – 1.77 (m, 2H). [0001015] Synthesis of N-(1-cyclobutyl-3-methyl-1H-pyrrolo[2,3-b]pyridin-5-yl)-2-(2,6- dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxamide
[0001016] Into a 40 mL vial were added 1-cyclobutyl-3-methylpyrrolo[2,3-b]pyridin-5-amine (220 mg, 1.093 mmol, 1 equiv), 2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5-carboxylic acid (315.08 mg, 1.093 mmol, 1 equiv), DIEA (847.64 mg, 6.558 mmol, 6 equiv), DMF (7 mL) and HATU (623.42 mg, 1.639 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for overnight at 60 °C under nitrogen atmosphere. The reaction was monitored by LCMS. Desired product could be detected by LCMS. The residue was purified by reversed- phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 50% to 60% gradient in 10 min; detector, UV 254 nm. This resulted in N-(1-cyclobutyl-3-methylpyrrolo[2,3-b]pyridin-5-yl-2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H - isoindole-5-carboxamide (110.3 mg, 20.82%) as a white solid. [0001017] LC-MS (ES, m/z): [M+H]+: 472.10 [0001018] 1H NMR: (400 MHz, DMSO-d6) δ 11.03 (s, 1H), 10.51 (s, 1H), 8.48 (d, J = 2.3 Hz, 1H), 8.34 (d, J = 2.3 Hz, 1H), 8.22 (s, 1H), 8.14 (d, J = 7.9 Hz, 1H), 7.89 (d, J = 7.9 Hz, 1H), 7.58 (s, 1H), 5.31 – 5.13 (m, 2H), 4.58 (d, J = 17.5 Hz, 1H), 4.45 (d, J = 17.6 Hz, 1H), 3.01 –
2.87 (m, 1H), 2.67 – 2.58 (m, 1H), 2.57 – 2.51 (m, 1H), 2.49 – 2.34 (m, 4H), 2.28 (s, 3H), 2.10 – 2.02 (m, 1H), 1.89 – 1.76 (m, 2H). [0001019] Example 1.97. Synthesis of N-(1-cyclobutyl-4-fluoro-3-methyl-1H-pyrrolo[2,3- b]pyridin-5-yl)-2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxamide (I-95)
[0001020] Synthesis of methyl 3-bromo-4-fluoro-1H-pyrrolo[2,3-b]pyridine-5-carboxylate
[0001021] Into a 30mL round-bottom flask were added methyl 4-fluoro-1H-pyrrolo[2,3- b]pyridine-5-carboxylate (1 g, 5.150 mmol, 1 equiv) in DMF (10 mL) was added NBS (1008.34 mg, 5.665 mmol, 1.1 equiv) in portions at room temperature under air atmosphere. The resulting mixture was stirred for 2h at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA),
30% to 70% gradient in 20 min; detector, UV 254 nm. This resulted in methyl 3-bromo-4-fluoro- 1H-pyrrolo[2,3-b]pyridine-5-carboxylate (850 mg, 60.44%) as a brown solid. [0001022] LC-MS (ES, m/z): [M+H]+: 273.1 [0001023] 1H NMR: (400 MHz, DMSO-d6) δ 12.77 (s, 1H), 8.73 (d, J = 9.2 Hz, 1H), 7.86 (d, J = 2.5 Hz, 1H), 3.89 (s, 3H). [0001024] Synthesis of methyl 3-bromo-1-cyclobutyl-4-fluoro-1H-pyrrolo[2,3-b]pyridine-5- carboxylate
[0001025] Into a 20 mL vial were added PPh3 (1440.84 mg, 5.494 mmol, 2 equiv) in THF (10 mL) was treated with DIAD (1110.80 mg, 5.494 mmol, 2 equiv) for 5 min at 0 °C under nitrogen atmosphere followed by the addition of methyl 3-bromo-4-fluoro-1H-pyrrolo[2,3-b]pyridine-5- carboxylate (750 mg, 2.747 mmol, 1 equiv) and cyclobutanol (198.05 mg, 2.747 mmol, 1 equiv) dropwise at 0 °C. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 30% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in methyl 3-bromo-1-cyclobutyl-4-fluoropyrrolo[2,3-b]pyridine-5-carboxylate (450 mg, 50.08%) as a yellow solid. [0001026] 1H NMR: (400 MHz, DMSO-d6) δ 8.66 (d, J = 9.0 Hz, 1H), 7.70 (s, 1H), 5.32 – 5.23 (m, 1H), 3.87 (s, 3H), 2.44 – 2.31 (m, 4H), 1.86 – 1.78 (m, 2H). [0001027] Synthesis of methyl 1-cyclobutyl-4-fluoro-3-methyl-1H-pyrrolo[2,3-b]pyridine-5- carboxylate
[0001028] Into a 40 mL vial were added methyl 3-bromo-1-cyclobutyl-4-fluoropyrrolo[2,3- b]pyridine-5-carboxylate (250 mg, 0.764 mmol, 1 equiv) and dimethylzinc (5.84 mg, 0.062
mmol, 2 equiv) in dimethylzinc (145.88 mg, 1.528 mmol, 2 equiv) was added Pd(dppf)Cl2 (13.42 mg, 0.018 mmol, 0.2 equiv) in portions at room temperature under air atmosphere. The resulting mixture was stirred for 4 h at 100 °C under nitrogen atmosphere. Desired product could be detected by LCMS. The resulting mixture was filtered and the filter cake was washed with MeOH (3 x 5 mL). The filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 30% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in methyl 1-cyclobutyl-4-fluoro-3-methylpyrrolo[2,3-b]pyridine-5- carboxylate (90 mg, 44.90%) as a brown solid. [0001029] LC-MS (ES, m/z): [M+H]+: 262.9 [0001030] 1H NMR: (400 MHz, DMSO-d6) δ 8.66 (d, J = 9.4 Hz, 1H), 7.70 (d, J = 1.5 Hz, 1H), 5.34 – 5.21 (m, 1H), 3.87 (s, 3H), 2.50 – 2.43 (m, 2H), 2.46 – 2.34 (m, 5H), 1.90 – 1.75 (m, 2H). [0001031] Synthesis of 1-cyclobutyl-4-fluoro-3-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid
[0001032] Into a 8mL vial were added methyl 1-cyclobutyl-4-fluoro-3-methylpyrrolo[2,3- b]pyridine-5-carboxylate (95 mg, 0.362 mmol, 1 equiv) and LiOH (43.37 mg, 1.810 mmol, 5 equiv) in THF (4.75 mL, 58.597 mmol, 161.87 equiv) was added H2O (4.75 mL, 263.522 mmol, 727.96 equiv) in portions at room temperature under air atmosphere. The resulting mixture was stirred for 4 h at room temperature under air. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. This resulted in 1-cyclobutyl-4- fluoro-3-methylpyrrolo[2,3-b]pyridine-5-carboxylic acid (80 mg, 88.97%) as a brown solid. The crude product used in the next step directly without further purification. [0001033] LC-MS (ES, m/z): [M+H]+: 248.9 [0001034] 1H NMR: (400 MHz, DMSO-d6) δ 13.11 (s, 1H), 8.65 (d, J = 9.5 Hz, 1H), 7.67 (d, J = 1.4 Hz, 1H), 5.33 – 5.20 (m, 1H), 2.47 (d, J = 2.5 Hz, 2H), 2.47 – 2.34 (m, 5H), 1.83-1.85 (m, 2H). [0001035] Synthesis of tert-butyl (1-cyclobutyl-4-fluoro-3-methyl-1H-pyrrolo[2,3-b]pyridin-5- yl)carbamate
[0001036] Into a 8 mL vial were added 1-cyclobutyl-4-fluoro-3-methylpyrrolo[2,3-b]pyridine-5- carboxylic acid (130 mg, 0.524 mmol, 1 equiv) and DPPA (288.22 mg, 1.048 mmol, 2 equiv) in t-BuOH (5 mL, 52.615 mmol, 100.48 equiv) was added TEA (105.98 mg, 1.048 mmol, 2 equiv) in portions at room temperature under air atmosphere. The resulting mixture was stirred for 2 h at 80 °C under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 30% to 70% gradient in 20 min; detector, UV 254 nm. This resulted in tert-butyl N-{1-cyclobutyl-4-fluoro-3- methylpyrrolo[2,3-b]pyridin-5-yl}carbamate (100 mg, 59.79%) as a white solid. [0001037] LC-MS (ES, m/z): [M+H]+: 320.2 [0001038] Synthesis of 1-cyclobutyl-4-fluoro-3-methyl-1H-pyrrolo[2,3-b]pyridin-5-amine
[0001039] Into a 8mL vial were added tert-butyl N-(1-cyclobutyl-4-fluoro-3-methylpyrrolo[2,3- b]pyridin-5-ylcarbamate (100 mg, 0.313 mmol, 1 equiv) in DCM (5 mL, 78.653 mmol, 251.20 equiv) was added TFA (1 mL, 13.463 mmol, 43.00 equiv) dropwise 0 °C under air atmosphere. The resulting mixture was stirred at room temperature for 2 h under nitrogen atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. This resulted in 1-cyclobutyl-4-fluoro-3- methylpyrrolo[2,3-b]pyridin-5-amine (50 mg, 72.83%) as a brown oil. The resulting mixture was used in the next step directly without further purification. [0001040] Synthesis of N-(1-cyclobutyl-4-fluoro-3-methyl-1H-pyrrolo[2,3-b]pyridin-5-yl)-2- (2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxamide
[0001041] Into a 8 mL vial were added 1-cyclobutyl-4-fluoro-3-methylpyrrolo[2,3-b]pyridin-5- amine (50 mg, 0.228 mmol, 1 equiv) and 2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5- carboxylic acid (78.88 mg, 0.274 mmol, 1.2 equiv) in DMF (5.00 mL, 64.597 mmol) were added HATU (130.06 mg, 0.342 mmol, 1.5 equiv) and DIEA (58.95 mg, 0.456 mmol, 2 equiv) in portions at room temperature under air atmosphere. The resulting mixture was stirred at 60 °C for 2 h under nitrogen atmosphere. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 30% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in N-(1-cyclobutyl-4-fluoro-3-methylpyrrolo[2,3-b]pyridin-5-yl-2- (2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5-carboxamide (11.2 mg, 9.64%) as a white solid. [0001042] LC-MS (ES, m/z): [M+H]+: 490.00 [0001043] 1H NMR: (400 MHz, DMSO-d6) δ 11.02 (s, 1H), 10.37 (s, 1H), 8.26 – 8.19 (m, 2H), 8.15 (d, J = 7.9 Hz, 1H), 7.90 (d, J = 7.9 Hz, 1H), 7.63 (s, 1H), 5.31 – 5.12 (m, 2H), 4.57 (d, J = 17.7 Hz, 1H), 4.45 (d, J = 17.6 Hz, 1H), 2.91-2.95 (m, 1H), 2.64 (s, 1H), 2.66 – 2.51 (m, 1H), 2.49 – 2.36 (m, 7H), 2.18 – 1.98 (m, 1H), 1.90 – 1.77 (m, 2H). [0001044] Example 1.98. Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(4-fluoro-1-isopropyl- 3-methyl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)-1-oxoisoindoline-5-carboxamide (I-99)
[0001045] Synthesis of 4-chloro-1H-pyrazolo[3,4-d]pyrimidin-6-amine
[0001046] Into a 1000 mL round-bottom flask were added 2-amino-4,6-dichloropyrimidine-5- carbaldehyde (20 g, 104.167 mmol, 1 equiv) and THF (315 mL) and triethylamine (21.08 g, 208.334 mmol, 2 equiv) and H2O (90 mL) and hydrazine hydrate (7.17 g, 114.584 mmol, 1.1 equiv, 80%) at room temperature. The resulting mixture was stirred for 4 h at 60 °C under nitrogen atmosphere. The reaction was monitored by LCMS. ~72% product. The mixture was allowed to cool down to room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by trituration with H2O (150 mL). The precipitated solids were collected by filtration and washed with water (2 x 20 mL). To afford 4-chloro-1H- pyrazolo[3,4-d]pyrimidin-6-amine (17 g, yield 91%) as a yellow solid. [0001047] LC-MS (ES, m/z): [M+H]+ 169.90 [0001048] 1H NMR: (300 MHz, DMSO-d6) δ 13.24 (s, 1H), 7.94 (s, 1H), 7.14 (s, 2H). [0001049] Synthesis of 4-chloro-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-6-amine
[0001050] Into a 250 mL round-bottom flask were added 4-chloro-1H-pyrazolo[3,4- d]pyrimidin-6-amine (6 g, 35.384 mmol, 1 equiv) and DMF (90 mL) and N-iodosuccinimide (15.92 g, 70.768 mmol, 2 equiv) at room temperature. The resulting mixture was stirred for 2 h at 80 °C under nitrogen atmosphere. The reaction was monitored by LCMS. No SM, ~75% product. The resulting mixture was concentrated under reduced pressure. The residue was purified by trituration with water/ACN=2:1 (120 mL). The precipitated solids were collected by filtration and washed with water (3 x 30 mL). To afford 4-chloro-3-iodo-1H-pyrazolo[3,4- d]pyrimidin-6-amine (9 g, 64%) as a brown solid. [0001051] LC-MS (ES, m/z): [M+H]+ 295.80 [0001052] 1H NMR: (300 MHz, DMSO-d6) δ 13.58 (s, 1H), 7.38 – 7.24 (m, 2H). [0001053] Synthesis of 4-chloro-3-iodo-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-6-amine
[0001054] Into a 500 mL round-bottom flask were added 4-chloro-3-iodo-1H-pyrazolo[3,4- d]pyrimidin-6-amine (8.8 g, 29.783 mmol, 1 equiv) and Cs2CO3 (19.41 g, 59.566 mmol, 2 equiv) and DMF (88 mL) and 2-iodopropane (5.57 g, 32.761 mmol, 1.1 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. No SM, ~15%+70% isomer+product. The resulting mixture was filtered, the filter cake was washed with DMF (88 mL)(2 x 5 mL). The filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18; mobile phase, MeCN in Water, 5% to 95% gradient in 30 min; detector, UV 220 nm. To afford 4-chloro-3-iodo-1- isopropylpyrazolo[3,4-d]pyrimidin-6-amine (4 g, yield 37%) as a yellow solid. [0001055] LC-MS (ES,m/z): [M+H]+ 337.90 [0001056] 1H NMR: (300 MHz, DMSO-d6) δ 7.40 (s, 2H), 4.81 (p, J = 6.6 Hz, 1H), 1.40 (d, J = 6.7 Hz, 6H). [0001057] Synthesis of 4-fluoro-3-iodo-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-6-amine
[0001058] Into a 250 mL round-bottom flask were added 4-chloro-3-iodo-1- isopropylpyrazolo[3,4-d]pyrimidin-6-amine (2 g, 5.925 mmol, 1 equiv) and THF (20 mL) and TBAF (5.93 mL, 5.925 mmol, 1 equiv) at room temperature. The resulting mixture was stirred for overnight at 80 °C under nitrogen atmosphere. The reaction was monitored by LCMS. ~43% product. The mixture was allowed to cool down to room temperature. The resulting mixture was diluted with EtOAc (100 mL). The resulting mixture was extracted with EtOAc (2 x 100 mL). The combined organic layers were washed with brine (3 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18; mobile phase, MeCN in Water, 5% to 95% gradient in 30 min; detector, UV 220 nm. To afford 4-fluoro-3-iodo-1-isopropylpyrazolo[3,4-d]pyrimidin-6-amine (670 mg, yield 31%) as a yellow solid. [0001059] LC-MS (ES, m/z): [M-H]- 320.00 [0001060] 1H NMR: (300 MHz, DMSO-d6) δ 7.38 (s, 2H), 4.80 (h, J = 6.6 Hz, 1H), 1.41 (d, J = 6.7 Hz, 6H). [0001061] 19F NMR: (282 MHz, DMSO) δ -65.14. [0001062] Synthesis of 4-fluoro-1-isopropyl-3-methyl-1H-pyrazolo[3,4-d]pyrimidin-6-amine
[0001063] Into a 50 mL round-bottom flask were added 4-fluoro-3-iodo-1- isopropylpyrazolo[3,4-d]pyrimidin-6-amine (640 mg, 1.993 mmol, 1 equiv) and methylboronic acid (357.93 mg, 5.979 mmol, 3 equiv) and K2CO3 (826.39 mg, 5.979 mmol, 3 equiv) and dioxane (11.2 mL) and H2O (1.4 mL) and Pd(dppf)Cl2 (291.68 mg, 0.399 mmol, 0.2 equiv) at room temperature. The resulting mixture was stirred for 6h at 80 °C under argon
atmosphere. The reaction was monitored by LCMS. The resulting mixture was diluted with EtOAc (200 mL). The resulting mixture was washed with brine (3 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18; mobile phase, MeCN in Water, 5% to 95% gradient in 25 min; detector, UV 220 nm. To afford 4-fluoro-1-isopropyl-3-methylpyrazolo[3,4-d]pyrimidin-6-amine (140 mg, yield 33%) as a yellow solid. [0001064] LC-MS (ES, m/z): [M+H]+ 210.05 [0001065] 1H NMR: (300 MHz, DMSO-d6) δ 7.15 (s, 2H), 4.77 (m, 1H), 2.39 (s, 3H), 1.39 (d, J = 6.7 Hz, 6H). [0001066] 19F NMR: (282 MHz, DMSO) δ -62.20. [0001067] Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(4-fluoro-1-isopropyl-3-methyl-1H- pyrazolo[3,4-d]pyrimidin-6-yl)-1-oxoisoindoline-5-carboxamide
[0001068] Into a 25 mL 2-necked round-bottom flask were added 4-fluoro-1-isopropyl-3- methylpyrazolo[3,4-d]pyrimidin-6-amine (50 mg, 0.239 mmol, 1 equiv) and 2-(2,6- dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5-carboxylic acid (75.77 mg, 0.263 mmol, 1.1 equiv) and pyridine (0.5 mL) at room temperature. To a stirred mixture and POCl3 (73.28 mL, 0.478 mmol, 2 equiv) was added dropwise at 0 °C under argon atmosphere. The resulting mixture was stirred for 3h at 0 °C under argon atmosphere. The reaction was monitored by LCMS. ~60% product. The resulting mixture was concentrated under vacuum. The reaction was quenched with Water at room temperature. The resulting mixture was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 5% to 95% gradient in 20 min; detector, UV 220 nm. The crude product was purified by Prep-HPLC chromatography with the following conditions: column: Xselect CSH Prep C18 OBD, 30*150mm, 5um; Mobile Phase A: Water(0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 20% B to 39% B in 8 min; Wave
Length: 254nm/220nm nm; RT1(min): 7.78. To afford 2-(2,6-dioxopiperidin-3-yl)-N-{4-fluoro- 1-isopropyl-3-methylpyrazolo[3,4-d]pyrimidin-6-yl}-1-oxo-3H-isoindole-5-carboxamide (28.5 mg, yield 24%) as a white solid. [0001069] LC-MS (ES, m/z): [M+H]+ 480.20 [0001070] 1H NMR: (300 MHz, DMSO-d6) δ 11.49 (s, 1H), 11.03 (s, 1H), 8.16 (s, 1H), 8.05 (m, 1H), 7.85 (d, J = 7.9 Hz, 1H), 5.17 (m, 1H), 4.93 (m, 1H), 4.63 – 4.37 (m, 2H), 3.06 – 2.84 (m, 1H), 2.55 (s, 4H), 2.44 (s, 1H), 2.12 – 1.99 (m, 1H), 1.46 (d, J = 6.7 Hz, 6H). [0001071] 19F NMR: (282 MHz, DMSO) δ -60.49. [0001072] Example 1.99. Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(4-fluoro-1-isopropyl- 1H-pyrazolo[3,4-d]pyrimidin-6-yl)-1-oxoisoindoline-5-carboxamide (I-100)
[0001073] Synthesis of 4-chloro-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-6-amine
[0001074] Into a 250 mL round-bottom flask were added 4-chloro-1H-pyrazolo[3,4- d]pyrimidin-6-amine (5 g, 29.486 mmol, 1 equiv) and DMF (100 mL) at room temperature. To the above mixture was added Cs2CO3 (19.21 g, 58.972 mmol, 2 equiv), 2-iodopropane (5.26 g, 30.960 mmol, 1.05 equiv) at room temperature. The resulting mixture was stirred for additional 2 h at room temperature. Desired product could be detected by LCMS. The reaction was quenched with water at room temperature. The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was
purified by silica gel column chromatography, eluted with PE / EA (5:1) to afford 4-chloro-1- isopropylpyrazolo[3,4-d]pyrimidin-6-amine (2 g, 32.05%) as a white solid. [0001075] LC-MS (ES, m/z): [M+H]+:212.10 [0001076] 1H-NMR: (300 MHz, DMSO-d6) δ 7.97 (s, 1H), 7.26 (s, 2H), 4.83 (hept, J = 6.7 Hz, 1H), 1.42 (d, J = 6.7 Hz, 6H). [0001077] Synthesis of 4-fluoro-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-6-amine
[0001078] Into a 100 mL round-bottom flask were added 4-chloro-1-isopropylpyrazolo[3,4- d]pyrimidin-6-amine (1.8 g, 8.505 mmol, 1 equiv) and THF (36.00 mL),TEA (1032.73 mg, 10.206 mmol, 1.2 equiv) at room temperature. To the above mixture was added TBAF (8.50 mL, 8.505 mmol, 1 equiv) at room temperature. The resulting mixture was stirred for additional overnight at 80 °C under nitrogen atmosphere. Desired product could be detected by LCMS. The reaction was quenched with Water at room temperature. The resulting mixture was extracted with EtOAc (3 x 30mL). The combined organic layers were washed with brine (3 x 30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (5:1) to afford 4-fluoro-1-isopropylpyrazolo[3,4-d]pyrimidin-6-amine (1 g, 60.24%) as a white solid. [0001079] LC-MS (ES, m/z): [M+H]+ :196.15 [0001080] 1H NMR: (300 MHz, DMSO-d6) δ 8.00 (s, 1H), 7.22 (s, 2H), 4.85 (p, J = 6.7 Hz, 1H), 1.43 (d, J = 6.7 Hz, 6H). [0001081] Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(4-fluoro-1-isopropyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)-1-oxoisoindoline-5-carboxamide
[0001082] Into a 25 mL round-bottom flask were added 4-fluoro-1-isopropylpyrazolo[3,4- d]pyrimidin-6-amine (100 mg, 0.512 mmol, 1 equiv) and 2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-
isoindole-5-carboxylic acid (147.67 mg, 0.512 mmol, 1 equiv) ,Pyridine(1ml) at room temperature.To the above mixture was added POCl3 (157.09 mg, 1.024 mmol, 2 equiv) at 0 °C. The resulting mixture was stirred for additional 2h at room temperature. Desired product could be detected by LCMS. The resulting mixture was diluted with EtOAc (50 mL). The reaction was quenched with water at room temperature. The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (3 x 50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water , 0% to 50% gradient in 30 min; detector, UV 254 nm. This resulted in 2-(2,6-dioxopiperidin-3-yl)-N-{4-fluoro-1-isopropylpyrazolo[3,4-d]pyrimidin-6- yl}-1-oxo-3H-isoindole-5-carboxamide (30.7 mg, 12.23%) as a white solid. [0001083] LC-MS (ES, m/z): [M+H]+ : 466.15 [0001084] 1H NMR: (300 MHz, DMSO-d6) δ 11.54 (s, 1H), 11.02 (s, 1H), 8.37 (s, 1H), 8.17 (s, 1H), 8.08 – 8.01 (m, 1H), 7.85 (d, J = 7.9 Hz, 1H), 5.35 – 4.87 (m, 2H), 4.69 – 4.23 (m, 2H), 3.02 – 2.83 (m, 1H), 2.62 (d, J = 16.7 Hz, 1H), 2.40 (dd, J = 13.4, 4.3 Hz, 1H), 2.13 – 1.95 (m, 1H), 1.49 (d, J = 6.7 Hz, 6H). [0001085] Example 1.100. Synthesis of N-(1-cyclobutyl-4-fluoro-3-methyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)-2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxamide (I-97)
[0001086] Synthesis of 4-chloro-1-cyclobutyl-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-6-amine
[0001087] Into a 100 mL vial were added 4-chloro-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-6- amine (2 g, 6.769 mmol, 1 equiv) and Cs2CO3 (8.82 g, 27.076 mmol, 4 equiv) and DMF (20 mL) and iodocyclobutane (2.46 g, 13.538 mmol, 2 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was monitored by LCMS. The mixture was allowed to cool down to room temperature. The resulting mixture was filtered, the filter cake was washed with DMF (2 x 2 mL). The filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18; mobile phase, MeCN in Water, 5% to 95% gradient in 30 min; detector, UV 220 nm. To afford 4-chloro-1-cyclobutyl-3- iodopyrazolo[3,4-d]pyrimidin-6-amine (450 mg, 17%) as a yellow solid. [0001088] LC-MS (ES, m/z): [M+H]+ 349.90 [0001089] 1H-NMR: (400 MHz, DMSO-d6) δ 7.41 (s, 2H), 5.07 (m, 1H), 2.59 (m, 2H), 2.37 – 2.24 (m, 2H), 1.83 (m, 2H). [0001090] Synthesis of 1-cyclobutyl-4-fluoro-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-6-amine
[0001091] Into a 20 mL sealed tube were added 4-chloro-1-cyclobutyl-3-iodopyrazolo[3,4- d]pyrimidin-6-amine (360 mg, 1.030 mmol, 1 equiv) and THF (3.6 mL) and TEA (125.06 mg, 1.236 mmol, 1.2 equiv) and TBAF (269.27 mg, 1.030 mmol, 1 equiv) at room temperature. The resulting mixture was stirred for 4 h at 80 °C under argon atmosphere. about 44% product. The mixture was allowed to cool down to room temperature. The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (3 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following
conditions: column, C18; mobile phase, MeCN in Water, 5% to 95% gradient in 30 min; detector, UV 220 nm. To afford 1-cyclobutyl-4-fluoro-3-iodopyrazolo[3,4-d]pyrimidin-6-amine (140 mg, 40%) as a yellow solid. [0001092] LC-MS (ES,m/z): [M-H]- 332.00 [0001093] 1H NMR: (300 MHz, DMSO-d6) δ 7.40 (s, 2H), 5.08 (m, 1H), 2.67 – 2.53 (m, 2H), 2.34 (d, J = 8.2 Hz, 2H), 1.92 – 1.76 (m, 2H). [0001094] 19F NMR: (282 MHz, DMSO) δ -64.97. [0001095] Synthesis of 1-cyclobutyl-4-fluoro-3-methyl-1H-pyrazolo[3,4-d]pyrimidin-6-amine
[0001096] Into a 50 mL round-bottom flask were added 1-cyclobutyl-4-fluoro-3- iodopyrazolo[3,4-d]pyrimidin-6-amine (120 mg, 0.360 mmol, 1 equiv) and methylboronic acid (107.82 mg, 1.800 mmol, 5 equiv) and K2CO3 (149.36 mg, 1.080 mmol, 3 equiv) and dioxane (1.6 mL) and H2O (0.8 mL) and Pd(dppf)Cl2 (105.44 mg, 0.144 mmol, 0.4 equiv) at room temperature. The resulting mixture was stirred at 100 °C for 2 h under nitrogen atmosphere. The reaction was monitored by LCMS. About 43% product. The mixture was allowed to cool down to room temperature. The resulting mixture was purified by reversed-phase flash chromatography with the following conditions: column, C18; mobile phase, MeCN in Water, 5% to 95% gradient in 10 min; detector, UV 220 nm. To afford 1-cyclobutyl-4-fluoro-3- methylpyrazolo[3,4-d]pyrimidin-6-amine(25 mg, 31%) as a yellow solid. [0001097] LC-MS (ES, m/z): [M+H]+ 222.05 [0001098] 1H NMR: (500 MHz, DMSO-d6) δ 7.16 (s, 2H), 5.04 (m, 1H), 2.62 – 2.55 (m, 2H), 2.41 (s, 3H), 2.35 – 2.27 (m, 2H), 1.86 – 1.77 (m, 2H). [0001099] 19F NMR: (471 MHz, DMSO) δ -61.99. [0001100] Synthesis of N-(1-cyclobutyl-4-fluoro-3-methyl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)- 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxamide
[0001101] Into a 25 mL round-bottom flask were added 1-cyclobutyl-4-fluoro-3- methylpyrazolo[3,4-d]pyrimidin-6-amine (25 mg, 0.113 mmol, 1 equiv) and 2-(2,6- dioxopiperidin-3-yl)-1-oxo-3H-isoindole-5-carboxylic acid (32.57 mg, 0.113 mmol, 1 equiv) and Pyridine (0.75 mL) at room temperature. To a stirred mixture and POCl3 (34.65 mg, 0.226 mmol, 2 equiv) was added dropwise at 0 °C under argon atmosphere. The final reaction mixture was irradiated with microwave radiation at 0 °C for 1 h. The reaction was monitored by LCMS. About 70% product. The resulting mixture was concentrated under vacuum. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18; mobile phase, MeCN in Water (0.1% FA), 5% to 95% gradient in 25 min; detector, UV 220 nm. The crude product was purified by reversed-phase flash chromatography with the following conditions: Column: F-phenyl Prep OBD C18 Column, 30*150 mm, 5μm; Mobile Phase A: Water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 26% B to 44% B in 8 min; Wave Length: 254nm/220nm nm; RT1(min): 7.42. To afford N-(1-cyclobutyl- 4-fluoro-3-methylpyrazolo[3,4-d]pyrimidin-6-yl-2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H- isoindole-5-carboxamide (12.1 mg, 20%) as a white solid. [0001102] LC-MS (ES, m/z): [M-H]- 489.90 [0001103] 1H-NMR: (300 MHz, DMSO-d6) δ 11.50 (s, 1H), 11.04 (s, 1H), 8.17 (s, 1H), 8.05 (d, J = 7.9 Hz, 1H), 7.85 (d, J = 7.9 Hz, 1H), 5.27 – 5.10 (m, 2H), 4.59 – 4.39 (m, 2H), 3.00 – 2.87 (m, 1H), 2.62 (d, J = 28.1 Hz, 6H), 2.45 – 2.26 (m, 3H), 2.06 (m, 1H), 1.87 (m, 2H). [0001104] 19F NMR: (282 MHz, DMSO) δ -60.32. [0001105] Example 1.101. Synthesis of N-(5-cyclobutoxypyrazin-2-yl)-2-(2,6- dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxamide (I-196)
[0001106] Step 1: 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxylic acid [0001107] To a solution of 3-(5-bromo-1-oxoisoindolin-2-yl)piperidine-2,6-dione (20 g, 62 mmol) in DMF (160 mL) under a N2 atmosphere was added Pd(OAc)2 (418 mg, 1.86 mmol), DCC (2.6 g, 12.6 mmol), triethylamine (12.6 g, 125 mmol), Xantphos (1.08 g, 1.86 mmol) and formic acid (10 g, 217 mmol) and the reaction heated at 100 °C for 3 h. The solvent was removed, the residue was treated with DCM (500 mL) and the precipitate that formed was collected by filtration, washed with water (20 mL x 2) and ethanol (20 mL x 2) and dried under reduced pressure to give 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxylic acid (11 g, 62 %) as a grey solid. LCMS m/z = 289.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 13.31 (s, 1H), 11.01 (s, 1H), 8.17 (s, 1H), 8.07 (d, J = 7.8 Hz, 1H), 7.83 (d, J = 7.8 Hz, 1H), 5.14 (dd, J = 13.2, 5.2 Hz, 1H), 4.63 – 4.33 (m, 2H), 2.97 – 2.85 (m, 1H), 2.66 – 2.56 (m, 1H), 2.47 – 2.35 (m, 1H), 2.09 – 1.97 (m, 1H). [0001108] Step 2: 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carbonyl chloride [0001109] A solution of 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxylic acid (2.0 g, 7 mmol) in SOCl2 (100 mL) was heated at reflux for 4 h. The solvent was removed to give 2-(2,6- dioxopiperidin-3-yl)-1-oxoisoindoline-5-carbonyl chloride (2.13 g, quant.) as a red solid. LCMS m/z = 303.0 [M+H]+. [0001110] Step 3: N-(5-cyclobutoxypyrazin-2-yl)-2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline- 5-carboxamide [0001111] To a solution of 5-cyclobutoxypyrazin-2-amine (40 mg, 0.24 mmol) in THF (2 mL) was added TEA (73.5 mg, 0.73 mmol) and 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5- carbonyl chloride (74 mg, 0.24 mmol). The mixture was stirred at RT overnight then concentrated and the residue obtained purified by prep-HPLC to afford N-(5- cyclobutoxypyrazin-2-yl)-2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxamide (4.5 mg, 4.3 %) as a white solid. LCMS m/z =436.10 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 11.05 (s,
2H), 8.90 (s, 1H), 8.23 (s, 1H), 8.17 – 8.09 (m, 2H), 7.85 (d, J = 7.8 Hz, 1H), 5.21 – 5.09 (m, 2H), 4.60 – 4.38 (m, 2H), 2.98 – 2.88 (m, 1H), 2.68 – 2.59 (m, 1H), 2.46 – 2.38 (m, 3H), 2.16 – 2.01 (m, 3H), 1.81 (q, J = 10.2 Hz, 1H), 1.67 (q, J = 9.2 Hz, 1H). [0001112] The following compounds were made in an analogous manner using appropriate starting materials.
[0001113] Example 1.102. Synthesis of 2-(2,6-dioxopiperidin-3-yl)-N-(4-(2- hydroxyethoxy)pyrimidin-2-yl)-1-oxoisoindoline-5-carboxamide (I-227) )
[0001114] To a solution of N-(4-(2-((tert-butyldimethylsilyl)oxy)ethoxy)pyrimidin-2-yl)-2-(2,6- dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxamide (120 mg, 0.22 mmol) in DCM (2 mL) was added TFA (1 mL) and the reaction stirred at room temperature for 2 h. The solvent was removed under vacuum and purified by prep-HPLC to afford 2-(2,6-dioxopiperidin-3-yl)-N-(4- (2-hydroxyethoxy)pyrimidin-2-yl)-1-oxoisoindoline-5-carboxamide (45 mg, 47 %) as a white solid. LCMS m/z = 426.40 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 11.08 – 10.99 (m, 2H), 8.42 (d, J = 5.8, 1H), 8.13 (s, 1H), 8.02 (d, J = 8.0, 1H), 7.83 (d, J = 7.8, 1H), 6.67 (d, J = 5.6, 1H), 5.18 – 5.12 (m, 1H), 4.85 (s, 1H), 4.56 – 4.37 (m, 2H), 4.31 (t, J = 5.0, 2H), 3.71 (t, J = 5.0, 2H), 2.99 – 2.87 (m, 1H), 2.65 – 2.56 (m, 1H), 2.47 – 2.37 (m, 1H), 2.08 – 2.00 (m, 1H).
[0001115] Example 1.103. Synthesis of 5-(benzofuran-2-yl)-N-((4'-fluoro-[1,1'-biphenyl]-4- yl)methyl)pyrimidin-4-amine (I-165)
[0001116] Step 1: 6-cyclobutoxypyridazin-3-amine [0001117] To a solution of Na (19.5 mg, 0.84 mmol) in cyclobutanol (2 mL) was added 6- chloropyridazin-3-amine (100 mg, 0.77 mmol) and TBAI (10 mg) and the reaction heated at 110 °C for 3 h. The solvent was removed under vacuum and the residue obtained purified by RP- column (26 % MeCN in water) to afford 6-cyclobutoxypyridazin-3-amine (80 mg, 62.9 %) as a yellow solid. LCMS m/z =166.05 [M+H]+; 1HNMR (400 MHz, DMSO-d6) δ 6.82 (d, J = 1.8 Hz, 2H), 5.84 (s, 2H), 5.07 (p, J = 7.4 Hz, 1H), 2.41 – 2.32 (m, 2H), 2.08 – 1.92 (m, 2H), 1.81 – 1.53 (m, 2H). [0001118] Step 2: 5-(benzofuran-2-yl)-N-((4'-fluoro-[1,1'-biphenyl]-4-yl)methyl)pyrimidin-4- amine [0001119] To a solution of 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carboxylic acid (90 mg, 0.31 mmol) in DMF (1 mL) was added HATU (118.7 mg, 0.31 mmol) and DIEA (121.0 mg, 0.94 mmol), after stirring at room temperature for 30 min, 6-cyclobutoxypyridazin-3-amine (90.0 mg, 0.31 mmol) was added and stirring continued for 2 h. The mixture was diluted with water (10 mL) and extracted with EtOAc (20 mL × 2), the combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by RP- column (40 % MeCN in water) to afford N-(6-cyclobutoxypyridazin-3-yl)-2-(2,6-dioxopiperidin- 3-yl)-1-oxoisoindoline-5-carboxamide (20 mg, 14.7 %) as a yellow solid. LCMS m/z = 436.15 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 11.40 (s, 1H), 11.01 (s, 1H), 8.25 (d, J = 9.7 Hz, 2H), 8.14 (d, J = 8.0 Hz, 1H), 7.86 (d, J = 7.9 Hz, 1H), 7.28 (d, J = 9.5 Hz, 1H), 5.33 – 5.12 (m, 2H), 4.59 – 4.39 (m, 2H), 2.98 – 2.88 (m, 1H), 2.66 – 2.58 (m, 1H), 2.46 – 2.37 (m, 2H), 2.20 – 1.55 (m, 6H). [0001120] The following compounds were made in an analogous manner using appropriate starting materials.
[0001121] Example 1.104. Synthesis of 2-(2,6-dioxopiperidin-3-yl)-1-oxo-N-(4-(pyrrolidin- 3-yloxy)pyrimidin-2-yl)isoindoline-5-carboxamide (I-225)
[0001122] To a solution of tert-butyl 3-((2-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5- carboxamido)pyrimidin-4-yl)oxy)pyrrolidine-1-carboxylate (25 mg, 0.045 mmol) in DCM (1 mL) was added TFA (0.5 mL). The mixture was stirred at room temperature for 2 h then the olvent was removed and the residue obtained purified by prep-HPLC to afford 2-(2,6- dioxopiperidin-3-yl)-1-oxo-N-(4-(pyrrolidin-3-yloxy)pyrimidin-2-yl)isoindoline-5-carboxamide (8.4 mg, 42 %) as a white solid. LCMS m/z =451.15 [M+H]+; 1H NMR (400 MHz, Methanol-d4) δ 8.53 (s, 1H), 8.43 (d, J = 5.6 Hz, 1H), 8.14 (s, 1H), 8.08 (d, J = 8.0 Hz, 1H), 7.95 (d, J = 7.8 Hz, 1H), 6.67 (d, J = 5.8 Hz, 1H), 5.69 (s, 1H), 5.20 (dd, J = 13.2, 5.2 Hz, 1H), 4.59 (m, 2H),
3.64 (s, 3H), 3.60 (d, J = 1.8 Hz, 1H), 3.53 – 3.46 (m, 1H), 3.45 – 3.37 (m, 1H), 2.93 (m, 1H), 2.80 (m, 1H), 2.53 (m, 1H), 2.39 (m, 2H), 2.25 – 2.17 (m, 1H). [0001123] Example 1.105. Synthesis of N-(4-cyclobutoxypyrimidin-2-yl)-3-(2,4- dioxotetrahydropyrimidin-1(2H)-yl)benzamide (I-231)
[0001124] Step 1: 3-((2-carboxyethyl)amino)benzoic acid [0001125] To a solution of 3-aminobenzoic acid (5.0 g, 36.46 mmol) in toluene (100 mL) was added acrylic acid (3.42 g, 47.4 mmol) and the reaction heated at reflux for 48 h. The reaction was cooled to room temperature and the solid precipitate collected by filtration, washed by toluene and dried to afford 3-((2-carboxyethyl)amino)benzoic acid (6.6 g, 87 %) as a white solid. LCMS m/z = 210.05 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 12.44 (s, 2H), 7.26 – 7.09 (m, 3H), 6.80 (d, J = 7.2 Hz, 1H), 5.90 (s, 1H), 3.27 (t, J = 6.8 Hz, 2H), 2.52 (s, 1H), 2.48 (s, 1H). [0001126] Step 2: 3-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzoic acid [0001127] To a solution of 3-((2-carboxyethyl)amino)benzoic acid (6.6 g, 31.55 mmol) in acetic acid (80 mL) under N2 was added urea (5.68 g, 94.65 mmol) and the reaction heated at reflux for 16 h. The solvent was removed under vacuum and the residue obtained resuspended in water (60 mL). The solid precipitate was collected by filtration, washed with water and dried under reduced pressure to afford 3-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzoic acid (4.5 g, 61 %) as a white solid. LCMS m/z = 235.05 [M+H]+; 11H NMR (400 MHz, DMSO-d6) δ 13.03 (s, 1H), 10.42 (s, 1H), 7.89 (s, 1H), 7.79 (d, J = 7.6 Hz, 1H), 7.63 – 7.47 (m, 2H), 3.83 (t, J = 6.6 Hz, 2H), 2.72 (t, J = 6.6 Hz, 2H). [0001128] Step 3: 3-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzoyl chloride [0001129] A solution of 3-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzoic acid (500 mg, 2.13 mmol) in SOCl2 (10 mL) was heated at reflux for 4 h. The solvent was removed under vacuumn to afford 3-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzoyl chloride (530 mg, 98 %) as yellow
solid.1H NMR (400 MHz, Chloroform-d) δ 8.07 – 7.99 (m, 2H), 7.70 (d, J = 8.2 Hz, 1H), 7.62 – 7.53 (m, 2H), 3.95 (t, J = 6.6 Hz, 2H), 2.89 (t, J = 6.6 Hz, 2H). [0001130] Step 4: N-(4-cyclobutoxypyrimidin-2-yl)-3-(2,4-dioxotetrahydropyrimidin-1(2H)- yl)benzamide [0001131] To a solution of 4-cyclobutoxypyrimidin-2-amine (100 mg, 0.61 mmol) in DCM (5 mL) under N2 was added TEA (612 mg, 6.05 mmol), DMAP (7.4 mg, 0.06 mmol) and 3-(2,4- dioxotetrahydropyrimidin-1(2H)-yl)benzoyl chloride (305 mg, 1.21 mmol). The reaction was stirred at room temperature for 2 h then the solvent was removed under vaccuum. The mixture was purified by RP-column (40% MeCN in water) and prep-HPLC to afford N-(4- cyclobutoxypyrimidin-2-yl)-3-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzamide (5 mg, 2.1 %) as a white solid. LCMS m/z = 382.05 [M+H]+; 1 H NMR (400 MHz, DMSO-d6) δ 10.88 (s, 1H), 10.43 (s, 1H), 8.41 (d, J = 5.6 Hz, 1H), 7.87 (s, 1H), 7.78 (d, J = 7.6 Hz, 1H), 7.61 – 7.47 (m, 2H), 6.61 (d, J = 5.8 Hz, 1H), 5.11 (s, 1H), 3.88 (t, J = 6.6 Hz, 2H), 2.74 (t, J = 6.6 Hz, 2H), 2.44 (d, J = 7.2 Hz, 2H), 2.06 (s, 2H), 1.81 – 1.55 (m, 2H). [0001132] The following compound was prepared in an analogous manner using appropriate starting materials.
[0001133] Example 1.106. Synthesis of N-(4-cyclobutoxypyrimidin-2-yl)-2-(2,6- dioxopiperidin-3-yl)-1,3-dioxoisoindoline-5-carboxamide (I-229)
[0001134] Step 1: 2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-5-carboxylic acid [0001135] To a solution of 3-aminopiperidine-2,6-dione hydrochloride (500 mg, 3.04 mmol) in AcOH (8 mL) was added sodium acetate (374 mg, 4.56 mmol) and 1,3-dioxo-1,3- dihydroisobenzofuran-5-carboxylic acid (584 mg, 3.04 mmol). The reaction mixture was heated at 90 °C for 5 h then poured into the water and the precipitate that formed collected by filtration and dried under reduced pressure to afford 2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-5- carboxylic acid (600 mg, quant.) as a purple solid which was used directly in the next step. LCMS m/z = 303.00 [M+H]+ ; 1H NMR (400 MHz, DMSO-d6) δ 11.15 (s, 1H), 8.40 (d, J = 7.6, 1H), 8.28 (s, 1H), 8.05 (d, J = 7.6, 1H), 5.24 – 5.14 (m, 1H), 2.94 – 2.86 (m, 1H), 2.68 – 2.54 (m, 2H), 2.11 – 1.95 (m, 2H). [0001136] Step 2: 2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-5-carbonyl chloride [0001137] A solution of 2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-5-carboxylic acid (250 mg, 0.83 mmol) in SOCl2 (4 mL) was heated at reflux for 4 h. The mixture was concentrated to afford 2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-5-carbonyl chloride (300 mg, quant.) as a pink solid which was used directly in the next step. [0001138] Step 3: N-(4-cyclobutoxypyrimidin-2-yl)-2-(2,6-dioxopiperidin-3-yl)-1,3- dioxoisoindoline-5-carboxamide [0001139] To a solution of 2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-5-carbonyl chloride (300 mg, 0.94 mmol) in DCM (6 mL) under a N2 atmosphere was added DMAP (12 mg, 0.094 mmol), 4-cyclobutoxypyrimidin-2-amine (155 mg, 0.94 mmol) and TEA (948 mg, 9.36 mmol). The reaction mixture was stirred at room temperature overnight, then was diluted with water (50 mL) and extracted with DCM (50 mL × 2). The combined organic layers were washed with
brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by pep- HPLC to afford N-(4-cyclobutoxypyrimidin-2-yl)-2-(2,6-dioxopiperidin-3-yl)-1,3- dioxoisoindoline-5-carboxamide (68 mg, 16 %) as a white solid. LCMS m/z = 450.15 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 11.23 (s, 1H), 11.15 (s, 1H), 8.42 (d, J = 5.8, 1H), 8.37 – 8.31 (m, 2H), 8.05 (d, J = 7.8, 1H), 6.64 (d, J = 5.8, 1H), 5.24 – 5.16 (m, 1H), 5.08 – 5.00 (m, 1H), 2.87 (d, J = 5.6, 1H), 2.58 (s, 2H), 2.42 (d, J = 9.4, 2H), 2.11 – 2.03 (m, 3H), 1.77 (d, J = 10.6, 1H), 1.62 – 1.54 (m, 1H). [0001140] Example 1.107. Synthesis of 3-(5-((4,6-dimethoxypyrimidin-2-yl)methoxy)-1- oxoisoindolin-2-yl)piperidine-2,6-dione (I-251)
[0001141] Step 1: 3-(1-oxo-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)isoindolin-2- yl)piperidine-2,6-dione [0001142] To a solution of methyl 3-(5-bromo-1-oxoisoindolin-2-yl)piperidine-2,6-dione (2.0 g, 6.19 mmol) in 1,4-dioxane (20 mL) was added 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2- dioxaborolane) (1.9 g 6.19 mmol), KOAc (1.8 g, 18.57mmol) and Pd(dppf)2CH2Cl2 (505 mg, 0.61 mmol). The reaction was heated at reflux for 3 h then was diluted with water (50 mL). The precipitate that formed was collected by filtration and dried under reduced pressure to afford 3- (1-oxo-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)isoindolin-2-yl)piperidine-2,6-dione (2 g, 90 %) as a grey solid. LCMS m/z =371.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 7.90 (s, 1H), 7.80 (d, J = 7.4 Hz, 1H), 7.74 (d, J = 0.8 Hz, 1H), 5.13 (dd, J = 13.2, 5.2 Hz, 1H), 4.51 – 4.28 (m, 2H), 2.97 – 2.84 (m, 1H), 2.65 – 2.55 (m, 1H), 2.44 – 2.34 (m, 1H), 2.05 – 1.96 (m, 1H), 1.32 (s, 12H). [0001143] Step 2: 3-(5-hydroxy-1-oxoisoindolin-2-yl)piperidine-2,6-dione
[0001144] To a solution of 3-(1-oxo-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)isoindolin- 2-yl)piperidine-2,6-dione (300 mg, 0.8 mmol) in DMF (3 mL) was added 30 % H2O2 (916 mg, 8.1 mmol). The reaction was stirred at room temperature for 4 h, then was diluted with water (30 mL) and extracted with EtOAc (50 mL x 2). The combined organic layers were dried over Na2SO4, filtered and concentrated to afford 3-(5-hydroxy-1-oxoisoindolin-2-yl)piperidine-2,6- dione(88 mg, 42 %) as a white solid. LCMS m/z = 261.1[M+H]+; 1H NMR (400 MHz, DMSO- d6) δ 11.02 – 10.92 (m, 1H), 7.99 – 7.65 (m, 3H), 5.18 – 5.07 (m, 1H), 4.55 – 4.32 (m, 2H), 2.69 – 2.55 (m, 2H), 2.41 – 2.30 (m, 1H), 2.08 – 1.93 (m, 2H). [0001145] Step 3: 3-(5-((4,6-dimethoxypyrimidin-2-yl)methoxy)-1-oxoisoindolin-2- yl)piperidine-2,6-dione [0001146] To a solution of 3-(5-hydroxy-1-oxoisoindolin-2-yl)piperidine-2,6-dione (88 mg, 0.34 mmol) in DMF (2 mL) was added 2-(bromomethyl)-4,6-dimethoxypyrimidine (94 mg, 0.41 mmol) and NaHCO3 (85 mg, 1.01mmol) and the reaction was heated at 60 °C for 3 h. The mixture was diluted with water (30 mL) and extracted with EtOAc (50 mL), dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep-HPLC to afford 3-(5-((4,6- dimethoxypyrimidin-2-yl)methoxy)-1-oxoisoindolin-2-yl)piperidine-2,6-dione (10 mg, 7 %) as a white solid. LCMS m/z = 413.1[M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 10.95 (s, 1H), 7.63 (d, J = 8.4 Hz, 1H), 7.21 (d, J = 2.2 Hz, 1H), 7.11 (dd, J = 8.4, 2.2 Hz, 1H), 6.19 (s, 1H), 5.22 (s, 2H), 5.06 (dd, J = 13.2, 5.2 Hz, 1H), 4.40 – 4.22 (m, 2H), 3.84 (s, 6H), 2.96 – 2.86 (m, 1H), 2.62 – 2.55 (m, 1H), 2.39 – 2.31 (m, 1H), 2.02 – 1.94 (m, 1H). [0001147] Example 1.108. Synthesis of 3-(5-(((4,6-dimethoxypyrimidin-2- yl)amino)methyl)-1-oxoisoindolin-2-yl)piperidine-2,6-dione (I-252)
[0001148] Step 1: 3-(5-(aminomethyl)-1-oxoisoindolin-2-yl)piperidine-2,6-dione [0001149] To a solution of 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoline-5-carbonitrile (200 mg, 0.743 mmol) in MeOH (3 mL) was added conc. HCl (0.5 mL) and PtO2 (84 mg). The reaction was stirred at room temperature under a H2 atmosphere overnight. The catalyst was removed by filtration and the filtrate concentrated. The residue obtained was triturated with a mixture of
DCM and MeOH (3:1, 20 mL) and dried to afford 3-(5-(aminomethyl)-1-oxoisoindolin-2- yl)piperidine-2,6-dione (100 mg, 49 %) as a brown solid. LCMS m/z = 274.1[M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 8.42 (s, 3H), 7.79 (d, J = 7.8 Hz, 1H), 7.71 (s, 1H), 7.62 (d, J = 8.0 Hz, 1H), 5.13 (dd, J = 13.2, 5.2 Hz, 1H), 4.51 – 4.32 (m, 2H), 4.19 – 4.13 (m, 2H), 2.97 – 2.88 (m, 1H), 2.64 – 2.58 (m, 1H), 2.45 – 2.36 (m, 1H), 2.05 – 1.99 (m, 1H). [0001150] Step 2: 3-(5-(((4,6-dimethoxypyrimidin-2-yl)amino)methyl)-1-oxoisoindolin-2- yl)piperidine-2,6-dione [0001151] To a solution of 3-(5-(aminomethyl)-1-oxoisoindolin-2-yl)piperidine-2,6-dione (60 mg, 0.22 mmol) in DMF (1 mL) was added TEA (90 mg, 0.89 mmol) and 2-chloro-4,6- dimethoxypyrimidine (40 mg, 0.23 mmol). The reaction mixture was heated at 120 °C for 1 h in a microwave reactor. The solvent was removed and the residue that was obtained was purified by prep-HPLC to afford 3-(5-(((4,6-dimethoxypyrimidin-2-yl)amino)methyl)-1-oxoisoindolin-2- yl)piperidine-2,6-dione (10.4 mg, 11 %) as a white solid. LCMS m/z = 412.05 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 10.96 (s, 1H), 7.79 (t, J = 6.2 Hz, 1H), 7.66 (d, J = 7.8 Hz, 1H), 7.54 (s, 1H), 7.47 (d, J = 8.4 Hz, 1H), 5.36 (s, 1H), 5.09 (dd, J = 13.2, 5.2 Hz, 1H), 4.55 (d, J = 6.2 Hz, 2H), 4.45 – 4.27 (m, 2H), 3.75 (s, 6H), 2.94 – 2.87 (m, 1H), 2.62 – 2.57 (m, 1H), 2.42 – 2.35 (m, 1H), 2.02 – 1.96 (m, 1H). [0001152] Example 1.109. Synthesis of 3-(5-([1,2,4]triazolo[4,3-a]pyridin-3-ylamino)-1- oxoisoindolin-2-yl)piperidine-2,6-dione (I-259)
[0001153] Step1: 3-(5-isothiocyanato-1-oxoisoindolin-2-yl)piperidine-2,6-dione [0001154] To a solution of 3-(5-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione (400 mg,1.54 mmol) in DMF (5 mL) was added 1,1'-thiocarbonyldiimidazole (413 mg, 2.32 mmol). The reaction was stirred at room temperature overnight. The solid precipidate that formed was
collected by filtration, washed with water (10 mL) and DCM (10 mL × 2) and dried under vacuum to afford 3-(5-isothiocyanato-1-oxoisoindolin-2-yl)piperidine-2,6-dione (260 mg, 55 %) as a grey solid. LCMS m/z =302.00 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 10.98 (s, 1H), 7.78 (d, J = 8.1 Hz, 1H), 7.71 (s, 1H), 7.58 – 7.51 (m, 1H), 5.11 (dd, J = 13.2, 5.2 Hz, 1H), 4.52 – 4.30 (m, 2H), 2.98 – 2.86 (m, 1H), 2.65 – 2.55 (m, 1H), 2.46 – 2.33 (m, 1H), 2.05 – 1.96 (m, 1H). [0001155] Step2: 3-(5-([1,2,4]triazolo[4,3-a]pyridin-3-ylamino)-1-oxoisoindolin-2- yl)piperidine-2,6-dione [0001156] To a solution of 3-(5-isothiocyanato-1-oxoisoindolin-2-yl)piperidine-2,6-dione (200 mg, 0.66 mmol) in DMF (1 mL) was added 2-hydrazinylpyridine (72 mg, 0.66 mmol) and TBAF (2.65 mL, 1 M, 2.65 mmol), and the mixture heated at 80 °C for 1 h in a microwave reactor. Iodine (168 mg, 0.66 mmol) and K2CO3 (183 mg, 1.33 mmol) were then added and the reaction stirred at room temperature for 3 h. The mixture was filtered and the filtrate concentrated, the residue obtained was purified by prep-HPLC to afford 3-(5-([1,2,4]triazolo[4,3-a]pyridin-3- ylamino)-1-oxoisoindolin-2-yl)piperidine-2,6-dione (45 mg, 18 %) as a yellow solid. LCMS m/z =377.00 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 10.96 (s, 1H), 9.72 (s, 1H), 8.40 (d, J = 7.0 Hz, 1H), 7.84 (s, 1H), 7.71 – 7.63 (m, 2H), 7.60 – 7.51 (m, 1H), 7.36 – 7.25 (m, 1H), 6.97 (t, J = 6.8 Hz, 1H), 5.08 (dd, J = 13.4, 5.2 Hz, 1H), 4.48 – 4.23 (m, 2H), 2.97 – 2.84 (m, 1H), 2.66 – 2.57 (m, 1H), 2.44 – 2.30 (m, 1H), 2.05 – 1.90 (m, 1H). [0001157] The following examples provide representative syntheses of intermediates. [0001158] Example 1.110. Synthesis of 5-cyclobutoxypyrazin-2-amine
[0001159] A solution of Na (53 mg, 2.32 mmol) in cyclobutanol (2.0 mL) was heated at 80 °C for 15 min. Then 5-bromopyrazin-2-amine (200 mg, 1.15 mmol) and TBAI (15 mg, cat.) were added. The reaction was heated at 140 °C for 5 h then concentrated under reduced pressure. The residue obtained was purified by RP-column (30 % MeCN in water) to afford 5- cyclobutoxypyrazin-2-amine (25 mg, 13 %) as a yellow solid. LCMS m/z =166.15 [M+H]+. [0001160] Example 1.111. Synthesis of 3-chloro-1-cyclobutyl-4-methyl-1H-pyrrolo[2,3- b]pyridin-5-amine
[0001161] Step 1: 4-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine [0001162] To a solution of 4-chloro-5-nitro-1H-pyrrolo[2,3-b]pyridine (300 mg, 1.52 mmol) in a mixtre of 1,4-dioxane (3 mL) and water (0.75 mL) was added trimethyl boroxine (2.2 mL, 7.6 mmol), Pd(PPh3)4 (17.6 mg, 0.0152 mmol) and potassium carbonate (420 mg, 3 mmol). The reaction was heated in a sealed tube at 130 °C for 2 h then was concentrated and diluted with water (10 mL) and extracted with EtOAc (5 mL × 6). The combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was washed with diethyl ether (20 mL x 2) to afford 4-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine (450 g, 56%) as a brown solid. LCMS m/z = 178.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 12.33 (s, 1H), 8.90 (s, 1H), 7.68 (d, J = 3.4 Hz, 1H), 6.84 (d, J = 3.6 Hz, 1H), 2.81 (s, 3H). [0001163] Step 2: 3-chloro-4-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine [0001164] To a solution of 4-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine (100 mg, 0.56 mmol) in DMF (3.0 mL) was added N-chlorosuccinimide (71.6 mg, 0.54 mmol) and the reaction heated at 50 °C for 3 h. The mixture was diluted with water (10 mL) and the solid that precipitated collected by filtration and dried under reduced pressure to afford 3-chloro-4-methyl-5-nitro-1H- pyrrolo[2,3-b]pyridine (100 mg, 84%) as a brown solid. LCMS m/z = 212.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.85 (s, 1H), 7.89 (d, J = 2.6 Hz, 1H), 2.95 (s, 3H). [0001165] Step 3: 3-chloro-1-cyclobutyl-4-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine [0001166] To a solution of 3-chloro-4-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine (100 mg, 0.47 mmol), cyclobutanol (50 mg, 0.69 mmol) and triphenylphosphine (186 mg, 0.71 mmol) in toluene (2 mL) at 0 °C was added DIAD (143 mg, 0.71 mmol). The reaction was heated at 80 °C
for 3 h then filtered concentrated, the residue obtained was purified by column chromatography on silica gel (eluent: Pet. ether: EtOAc = 5: 1, v/v) to afford 3-chloro-1-cyclobutyl-4-methyl-5- nitro-1H-pyrrolo[2,3-b]pyridine (100 mg, 80 %) as a yellow solid. LCMS m/z = 266.0 [M+H]+; 1H NMR (400 MHz, Chloroform-d) δ 8.90 (s, 1H), 7.48 (s, 1H), 5.42 – 5.33 (m, 1H), 3.05 (s, 3H), 2.62 – 2.51 (m, 2H), 2.49 – 2.35 (m, 2H), 2.00 – 1.88 (m, 2H). [0001167] Step 4: 3-chloro-1-cyclobutyl-4-methyl-1H-pyrrolo[2,3-b]pyridin-5-amine [0001168] To a solution of 3-chloro-1-cyclobutyl-4-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine (80 mg , 0.3 mmol) in a mixture of EtOH (5 mL) and water (1 mL) was added iron (84 mg, 1.5 mmol) and ammonium chloride (80 mg, 1.5 mmol) and the reaction heated at reflux for 3 h. The mixture was diluted with water (20 mL) and extracted with EtOAc (20 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated to give 3- chloro-1-cyclobutyl-4-methyl-1H-pyrrolo[2,3-b]pyridin-5-amine (40 mg, 56 %) as a brown oil. LCMS m/z = 236.1[M+H]+. [0001169] Example 1.112. Synthesis of 4-(3,3-difluorocyclobutoxy)pyrimidin-2-amine
[0001170] A solution of Na (35.5 mg, 1.54 mmol) in 3,3-difluorocyclobutan-1-ol (0.5 mL) was heated at 80 °C for 15 min, then 4-chloropyrimidin-2-amine (100 mg, 0.772 mmol) and TBAI (5 mg, cat.) were added. The reaction was heated at 140 °C for 5 h then the mixture was concentrated and purified by RP-column (50 % MeCN in water) to afford 4-(3,3- difluorocyclobutoxy)pyrimidin-2-amine (60 mg, 38.6 %) as a yellow solid. LCMS m/z = 202.1[M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.99 (d, J = 5.6 Hz, 1H), 6.57 (s, 2H), 6.02 (d, J = 5.6 Hz, 1H), 5.11 – 4.98 (m, 1H), 3.12 (d, J = 15.2 Hz, 2H), 2.76 – 2.67 (m, 2H). [0001171] Example 1.113. Synthesis of 4-ethoxypyrimidin-2-amine
[0001172] To a solution of 4-chloropyrimidin-2-amine ( 300 mg, 2.32 mmol) in EtOH (3.0 mL) was added sodium ethoxide (160 mg, 2.32 mmol) and TBAI (15 mg, cat.) and the reaction heated at reflux overnight. The mixture was concentrated and the residue obtained purified by RP-
column (30 % MeCN in water) to afford 4-ethoxypyrimidin-2-amine (98 mg, 30 %) as a white solid. LCMS m/z =140.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.93 (d, J = 5.6 Hz, 1H), 6.47 (s, 2H), 5.96 (d, J = 5.6 Hz, 1H), 4.24 (q, J = 7.1 Hz, 2H), 1.29 – 1.25 (m, 3H). [0001173] Example 1.114. Synthesis of 4-cyclopropoxypyrimidin-2-amine
[0001174] To a solution of 4-chloropyrimidin-2-amine (1.0 g, 7.72 mmol) in THF (10 mL) was added cyclopropanol (1.12 g, 19.30 mmol) and potassium tert-butoxide (1.13 g, 10.03 mmol). The reaction was heated at reflux overnight then cooled to room temperature, diluted with water (30 mL) and extracted with EtOAc (30 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The crude was purified by RP-column (21 % MeCN in water) to afford 4-cyclopropoxypyrimidin-2-amine (370 mg, 31.6 %) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 7.99 (d, J = 5.6 Hz, 1H), 6.52 (s, 2H), 6.06 (d, J = 5.6 Hz, 1H), 4.17 (dt, J = 6.2, 3.2 Hz, 1H), 0.75 – 0.64 (m, 4H). [0001175] Example 1.115. Synthesis of 4-(prop-2-yn-1-yloxy)pyrimidin-2-amine
[0001176] To a solution of prop-2-yn-1-ol (433 mg, 7.72 mmol) in 1,4-dioxane (5.0 mL) was added potassium tert-butoxide (1.1 g, 10.03 mmol) and the reaction stirred at room temperature for 30 min. a solution of 4-chloropyrimidin-2-amine (1.0 g, 7.72 mmol) in DMSO (5 mL) was added and the reaction heated at 60 °C overnight. The mixture was concentrated and purified by RP-column (25 % MeCN in water) to afford 4-(prop-2-yn-1-yloxy)pyrimidin-2-amine (410 mg, 35 %) as a yellow solid. LCMS m/z =150.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.99 (d, J = 5.6 Hz, 1H), 6.61 (s, 2H), 6.03 (d, J = 5.6 Hz, 1H), 4.92 (d, J = 2.4 Hz, 2H), 3.53 (t, J = 2.4 Hz, 1H). [0001177] Example 1.116. Synthesis of 4-(cyclopentyloxy)pyrimidin-2-amine
[0001178] To a solution of 4-chloropyrimidin-2-amine (1 g, 7.7 mmol) in THF (15 mL) was added cyclopentanol (1.3 g, 15.4 mmol) and potassium tert-butoxide (1.3 g, 11.6 mmol,1.5 eq) and the reaction heated at reflux overnight. The reaction was diluted with water (70 mL) and extracted with EtOAc (150 mL x 3), the combined organic layers were dried over sodium sulfate filtered and concentrated to give 4-(cyclopentyloxy)pyrimidin-2-amine (1.4 g, 100 % ) as a yellow oil. LCMS m/z =180.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.91 (d, J = 5.6 Hz, 1H), 6.43 (s, 2H), 5.92 (d, J = 5.6 Hz, 1H), 5.34 – 5.30 (m, 1H), 1.94 – 1.86 (m, 2H), 1.69 – 1.54 (m, 6H). [0001179] Example 1.117. Synthesis of 4-phenoxypyrimidin-2-amine
[0001180] To a solution of 4-chloropyrimidin-2-amine (100 mg, 0.772 mmol) in DMF (2.0 mL) was added phenol (109 mg, 1.158 mmol) and potassium tert-butoxide (260 mg, 2.236 mmol) and the reaction heated at 110 °C overnight. The mixture was purified by RP-column (30 % MeCN in water) to afford 4-phenoxypyrimidin-2-amine (50 mg, 34.5%) as a yellow solid. LCMS m/z =188.10 [M+H]+; 1H NMR (400 MHz, CD3OD) δ 8.06 (d, J = 5.8 Hz, 1H), 7.43 (d, J = 8.0 Hz, 2H), 7.25 (d, J = 7.4 Hz, 1H), 7.13 (d, J = 7.4 Hz, 2H), 6.14 (d, J = 5.8 Hz, 1H). [0001181] Example 1.118. Synthesis of 4-(sec-butoxy)pyrimidin-2-amine
[0001182] To a solution of butan-2-ol (572 mg, 7.72 mmol) in 1,4-dioxane (5.0 mL) was added potassium tert-butoxide (1.2 g, 10.03 mmol) and the reaction stirred at room temperature for 30 min. A solution of 4-chloropyrimidin-2-amine (1.0 g, 7.72 mmol) in DMSO (5 mL) was added and the reaction heated to 60 °C and stirred overnight. The mixture was concentrated and the residue obtained purified by RP-column (25 % MeCN in water) to afford 4-(sec- butoxy)pyrimidin-2-amine (450 mg, 34 %) as a yellow oil. LCMS m/z = 168.2[M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.91 (d, J = 5.6 Hz, 1H), 6.42 (s, 2H), 5.92 (d, J = 5.7 Hz, 1H), 5.10 – 5.04 (m, 1H), 1.64 – 1.55 (m, 2H), 1.21 (d, J = 6.2 Hz, 3H), 0.87 (t, J = 7.4 Hz, 3H). [0001183] Example 1.119. Synthesis of tert-butyl 3-((2-aminopyrimidin-4- yl)oxy)pyrrolidine-1-carboxylate
[0001184] To a solution of tert-butyl 3-hydroxypyrrolidine-1-carboxylate (1.44 g, 0.71 mmol) in a mixture of 1,4-dioxane (5 mL) and DMSO (5 mL) was added potassium tert-butoxide (1.13g, 10.04 mmol) and the mixture was stirred at room temperature for 30 min.4-chloropyrimidin-2- amine (1.0 g, 7.72 mmol) was added and the reaction heated at 100 °C overnight. The reaction was diluted with water (30 mL) and extracted with EtOAc (30 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by column chromatography on silica gel (eluent: Pet. Ether: EtOAc = 3: 1, v/v) to afford tert-butyl 3-((2-aminopyrimidin-4-yl)oxy)pyrrolidine-1-carboxylate (1.9 g, 87 %) as a yellow solid. LCMS m/z =281.05[M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.96 (d, J = 5.6 Hz, 1H), 6.54 (s, 2H), 5.99 (d, J = 5.6 Hz, 1H), 5.43 (d, J = 13.2 Hz, 1H), 3.61 – 3.50 (m, 1H), 3.41 (m, 1H), 3.25 (d, J = 7.8 Hz, 2H), 2.22 – 1.96 (m, 2H), 1.39 (d, J = 3.6 Hz, 9H). [0001185] Example 1.120. Synthesis of 4-(pyridin-3-yloxy)pyrimidin-2-amine
[0001186] A solution of 4-chloropyrimidin-2-amine (1 g, 7.72 mmol), pyridin-3-ol (880 mg, 9.26 mmol) and potassium tert-butoxide (1.3 g, 11.6 mmol) in a mixture of 1,4-dioxane (6 mL) and DMSO (6 mL) was heated at 90 °C overnight. The reaction was diluted with water (30 mL) and extracted with EtOAc (30 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The crude product was purified by column chromatography on silica gel (eluent: Pet. ether: EtOAc = 5:1 to 2:1) to afford 4-(pyridin-3- yloxy)pyrimidin-2-amine (460 mg, 32 %) as a yellow solid. LCMS m/z = 189.05 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.52 – 8.41 (m, 2H), 8.15 (d, J = 5.6, 1H), 7.72 – 7.63 (m, 1H), 7.48 (dd, J = 8.4, 4.8, 1H), 6.67 (s, 2H), 6.23 (d, J = 5.6, 1H). [0001187] Example 1.121. Synthesis of 4-((tetrahydrofuran-3-yl)oxy)pyrimidin-2-amine
[0001188] To a solution of 4-chloropyrimidin-2-amine (1 g, 7.72 mmol) in MeCN (10 mL) was added tetrahydrofuran-3-ol (1.02 g, 11.58 mmol) and Cs2CO3 (5.03 g, 15.44 mmol). The reaction mixture was heated at 70 °C overnight then was was filtered through Celite and concentrated. The residue obtained was purified by column chromatography on silica (eluent: Pet. ether: EtOAc = 1: 1) to afford 4-((tetrahydrofuran-3-yl)oxy)pyrimidin-2-amine (700 mg, 50 %) as a white solid. LCMS m/z = 182.15 [M+H]+ ; 1H NMR (400 MHz, DMSO-d6) δ 7.95 (d, J = 5.6 Hz, 1H), 6.51 (s, 2H), 5.98 (d, J = 5.6 Hz, 1H), 5.49 – 5.41 (m, 1H), 3.91 – 3.69 (m, 4H), 2.26 – 2.13 (m, 1H), 2.00 – 1.93 (m, 1H). [0001189] Example 1.122. Synthesis of 4-(4-methoxyphenoxy)pyrimidin-2-amine
[0001190] To a solution of 4-chloropyrimidin-2-amine (100 mg, 0.77 mmol) in DMF (4 mL) was added 4-methoxyphenol (144 mg, 1.16 mmol) and t-BuOK (260 mg, 2.31 mmol). The reaction mixture was heated at 110 °C overnight then was diluted with water (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep-TLC (eluent: DCM: MeOH=20:1) to afford 4-(4-methoxyphenoxy)pyrimidin-2-amine (30 mg, 17 %) as a white solid. LCMS m/z =218.10 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.07 (d, J = 5.4 Hz, 1H), 7.12 – 7.04 (m, 2H), 6.99 – 6.94 (m, 2H), 6.57 (s, 2H), 6.03 (d, J = 5.4 Hz, 1H), 3.76 (d, J = 2.2 Hz, 3H). [0001191] Example 1.123. Synthesis of 4-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzoyl chloride
[0001192] Step 1: 4-((2-carboxyethyl)amino)benzoic acid [0001193] To a solution of 4-aminobenzoic acid (2.0 g, 14.58 mmol) in a mixture of AcOH (4 mL) and water (20 mL) was added acrylic acid (3.16 g, 43.74 mmol). The reaction was heated at 110 °C overnight then was cooled to room temperature. The precipitate that formed was collected by filtration, washed with water (10 mL) and dried to afford 4-((2- carboxyethyl)amino)benzoic acid (847 mg, 27 %) as a yellow solid. LCMS m/z =210.05 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 12.17 (s, 2H), 7.66 (d, J = 8.8 Hz, 2H), 6.57 (d, J = 8.8 Hz, 2H), 6.45 (t, J = 5.6 Hz, 1H), 3.62 (t, J = 7.2 Hz, 2H), 3.28 (d, J = 6.2 Hz, 2H). [0001194] Step 2: 4-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzoic acid [0001195] To a solution of 4-((2-carboxyethyl)amino)benzoic acid (840 mg, 4.02 mmol) in AcOH (5 mL) was added urea (723 mg, 12.05 mmol) and the reaction heated at 120 °C overnight. The reaction concentrated then resuspended in water the precipitate collected by filtration, washed with water (2 mL) and dried to afford 4-(2,4-dioxotetrahydropyrimidin-1(2H)- yl)benzoic acid (258 mg, 27 %) as a yellow solid. LCMS m/z =235.05 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 12.84 (s, 1H), 10.48 (s, 1H), 7.94 (d, J = 8.6 Hz, 2H), 7.46 (d, J = 8.6 Hz, 2H), 3.86 (t, J = 6.6 Hz, 2H), 2.72 (t, J = 6.6 Hz, 2H). [0001196] Step 3: 4-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzoyl chloride [0001197] A solution of 4-(2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzoic acid (150 mg, 0.64 mmol) in SOCl2 (3 mL) was heated at reflux for 4 h. The reaction was concentrated to afford 4- (2,4-dioxotetrahydropyrimidin-1(2H)-yl)benzoyl chloride (160 mg, quant.) as a yellow oil which was used in next step without further purification. [0001198] Example 1.124. Synthesis of 4-methoxy-6-propoxypyrimidin-2-amine
[0001199] To a solution of 4-bromo-6-methoxypyrimidin-2-amine (350 mg, 1.72 mmol) in THF (4.0 mL) was added t-BuOK (288 mg,2.57 mmol) and propan-1-ol (103 mg, 1.72 mmol) and the reaction heated at reflux overnight. The mixture was concentrated and purified by RP-column (30 % MeCN in water) to afford 4-methoxy-6-propoxypyrimidin-2-amine (86 mg, 27 %) as a yellow solid. LCMS m/z = 184.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 6.49 (s, 2H), 5.32 (s, 1H), 4.10 (t, J = 6.6 Hz, 2H), 3.75 (s, 3H), 1.65 (q, J = 7.2 Hz, 2H), 0.92 (t, J = 7.4 Hz, 3H). [0001200] Example 1.125. Synthesis of 4-cyclobutoxy-6-methylpyrimidin-2-amine
[0001201] To a solution of 4-chloro-6-methylpyrimidin-2-amine (1.0 g, 6.97 mmol) in THF (10.0 mL) was added t-BuOK (2.3 g, 20.9 mmol) and cyclobutanol (1.5 g, 20.9 mmol) and the reaction heated at reflux. The mixture was concentrated and the residue obtained purified by RP- column (30 % MeCN in water) to afford 4-cyclobutoxy-6-methylpyrimidin-2-amine (500 mg, 41 %) as a yellow solid. LCMS m/z = 180.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 6.34 (s, 2H), 5.82 (s, 1H), 5.09 – 5.02 (m, 1H), 2.39 – 2.31 (m, 2H), 2.11 (s, 3H), 2.03 – 1.96 (m, 2H), 1.79 – 1.69 (m, 1H), 1.64 – 1.54 (m, 1H). [0001202] Example 1.126. Synthesis of 3,5-dimethoxy-4-methylaniline
[0001203] Step 1: tert-butyl (3,5-dimethoxy-4-methylphenyl)carbamate [0001204] To a solution of 3,5-dimethoxy-4-methylbenzoic acid (200 mg, 1.02 mmol) in a mixture of toluene (3 mL) and t-butanol (3 mL) was added TEA (0.31 mL, 2.24 mmol) and DPPA (0.24 mL, 1.12 mmol). The reaction was heated at 85 °C under N2 overnight then was diluted with water (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by chromatography on silica gel (eluent: Pet.ether:EtOAc=4:1) to afford tert-butyl (3,5-
dimethoxy-4-methylphenyl)carbamate (18 mg, 6.6 %) as a colorless oil.1H NMR (400 MHz, DMSO-d6) δ 9.19 (s, 1H), 6.81 (s, 2H), 3.70 (s, 6H), 1.90 (s, 3H), 1.47 (s, 9H). [0001205] Step 2: 3,5-dimethoxy-4-methylaniline [0001206] To a solution of tert-butyl (3,5-dimethoxy-4-methylphenyl)carbamate (70 mg, 0.26 mmol) in DCM (2 mL) was added 4 N HCl in 1,4-dioxane (1 mL). The reaction mixture was stirred at room temperature for 2 h then the solvent was removed to afford 3,5-dimethoxy-4- methylaniline (44 mg, quant.) as a yellow oil which was used for next step without further purification. LCMS m/z =168.01 [M+H]+. [0001207] Example 1.127. Synthesis of 4-cyclobutoxy-6-methoxypyridin-2-amine
[0001208] Step 1: 4-chloro-6-methoxypyridin-2-amine [0001209] To a solution of 4,6-dichloropyridin-2-amine (3.0 g, 18.52 mmol) in NMP (30 mL) under a N2 atmosphere was added sodium methoxide (5.0 g, 92.6 mmol) and the reaction heated at 120 °C overnight. The mixture was diluted with water (50 mL) and extracted with EtOAc (50 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The crude product was purified column chromatography on silica gel (eluent: Pet. ether: EtOAc = 5: 1 to 2: 1) to afford 4-chloro-6-methoxypyridin-2-amine (800 mg, 28 %) as a brown oil. LCMS m/z =159.05 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 6.23 (s, 2H), 6.03 (d, J = 1.6 Hz, 1H), 5.93 (d, J = 1.6 Hz, 1H), 3.74 (s, 3H); further elution provided 6-chloro-4- methoxypyridin-2-amine (HL-3087-052-2, 1.0 g, 34 %) as a brown oil. LCMS m/z =159.05[M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 6.25 (s, 2H), 6.16 (d, J = 1.8 Hz, 1H), 5.89 (d, J = 1.8 Hz, 1H), 3.72 (s, 3H). [0001210] Step 2: 4-cyclobutoxy-6-methoxypyridin-2-amine [0001211] To a solution of cyclobutanol (546 mg, 7.60 mmol) in a mixture of 1,4-Dioxane (4.0 mL) and DMSO (4.0 mL) was added t-BuOK (1.13 g, 10.13 mmol) and the mixture was stirred at room temperature for 30 min.4-chloro-6-methoxypyridin-2-amine (400 mg, 2.53 mmol) was added and the reaction heated at 110 °C overnight. The mixture was concentrated and purified by column chromatography on silica gel (eluent: Pet. ether: EtOAc = 3: 1, v/v) to afford 4- cyclobutoxy-6-methoxypyridin-2-amine (170 mg, 34 %) as a yellow solid.1H NMR (400 MHz,
DMSO-d6) δ 5.75 (d, J = 1.2 Hz, 2H), 5.51 (d, J = 1.8 Hz, 1H), 5.39 (d, J = 1.8 Hz, 1H), 4.65 – 4.55 (m, 1H), 3.68 (s, 3H), 2.37 – 2.31 (m, 2H), 2.04 – 1.95 (m, 2H), 1.80 – 1.71 (m, 1H), 1.67 – 1.57 (m, 1H). [0001212] Example 1.128. Synthesis of 6-cyclobutoxy-4-methoxypyridin-2-amine
[0001213] To a solution of cyclobutanol (546 mg, 7.60 mmol) in a mixture of 1,4-Dioxane (4.0 mL) and DMSO (4.0 mL) was added t-BuOK (1.13 g, 10.13 mmol) and the mixture stirred at room temperature for 30 min.6-chloro-4-methoxypyridin-2-amine (400 mg, 2.53 mmol) was added and the reaction heated at 110 °C overnight. The mixture was concentrated and purified by column chromatography on silica gel (eluent: Pet. ether: EtOAc = 3: 1, v/v) to afford 6- cyclobutoxy-4-methoxypyridin-2-amine (110 mg, 22 %) as a yellow solid. LCMS m/z =195.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 5.68 (s, 2H), 5.56 (d, J = 1.8 Hz, 1H), 5.43 (d, J = 1.8 Hz, 1H), 5.03 – 4.92 (m, 1H), 3.66 (s, 3H), 2.36 – 2.28 (m, 2H), 2.00 – 1.92 (m, 2H), 1.75 – 1.66 (m, 1H), 1.60 – 1.51 (m, 1H). [0001214] Example 1.129. Synthesis of 4-cyclobutoxy-5-fluoropyrimidin-2-amine
[0001215] To a solution of cyclobutanol (150 mg, 0.67 mmol) in 1,4-dioxane (3 mL) was added t-BuOK (300 mg, 0.88 mmol) and the reaction stirred at room temperature for 30 mins. A solution of 4-chloro-5-fluoropyrimidin-2-amine (300 mg, 0.68 mmol) in DMSO (3 mL) was added and the reaction heated at 60 °C overnight. The mixture was diluted with water (50 mL) and extracted with EtOAc (50 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep-TLC (DCM: MeOH = 20: 1) to afford 4-cyclobutoxy-5-fluoropyrimidin-2-amine (150 mg, 40 %) as a yellow oil. LCMS m/z = 184.10 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.99 (d, J = 3.2, 1H), 6.44 (s, 2H), 5.14 (t, J = 7.4, 1H), 2.43 – 2.35 (m, 2H), 2.15 – 2.05 (m, 2H), 1.83 – 1.59 (m, 2H). [0001216] Example 1.130. Synthesis of 4-isopropoxypyrimidin-2-amine
[0001217] To a solution of propan-2-ol (280 mg, 4.65 mmol) in 1,4-dioxane (4 mL) was added t- BuOK (339 mg, 3.02 mmol) and the reaction stirred at room temperature for 30 mins. A solution of 4-chloropyrimidin-2-amine (300 mg, 2.32 mmol) in DMSO (4 mL) was added and the reaction was heated at 60 °C overnight. The mixture was diluted with water (50 mL) and extracted with EtOAc (50 mL × 2), the combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep-TLC (DCM: MeOH = 20: 1) to afford 4-cyclobutoxy-5-fluoropyrimidin-2-amine (350 mg, 90 %) as a white solid. LCMS m/z = 154.15 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.91 (d, J = 5.6, 1H), 6.43 (s, 2H), 5.91 (d, J = 5.6, 1H), 5.22 (p, J = 6.2, 1H), 1.25 (d, J = 6.2, 6H). [0001218] Example 1.131. Synthesis of 4-(cyclohexyloxy)pyrimidin-2-amine
[0001219] To a solution of cyclohexanol (679 mg, 6.78 mmol) in 1,4-dioxane (12 mL) was added t-BuOK (988 mg, 8.81 mmol) and the reaction stirred at room temperature for 30 min. A solution of 4-chloropyrimidin-2-amine (1.0 g, 6.78 mmol) in DMSO (12 mL) was added and the reaction was heated at 60 °C overnight. The mixture was diluted with water (80 mL) and extracted with EtOAc (80 mL × 2), the combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by RP-column (72% acetonitrile in water) to afford 4-(cyclohexyloxy)pyrimidin-2-amine (490 mg, 37 %) as a yellow solid. LCMS m/z = 194.15 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.91 (d, J = 5.6, 1H), 6.42 (s, 2H), 5.92 (d, J = 5.6, 1H), 4.98 (s, 1H), 1.91 (d, J = 5.4, 2H), 1.75 – 1.66 (m, 2H), 1.57 – 1.42 (m, 2H), 1.37 – 1.23 (m, 4H). [0001220] Example 1.132. Synthesis of 4-(but-2-yn-1-yloxy)pyrimidin-2-amine
[0001221] To a solution of but-2-yn-1-ol (543 mg, 7.75 mmol) in 1,4-dioxane (12 mL) was added t-BuOK (1.13 g, 10.08 mmol)and the reaction was stirred at room temperature for 30 min.
A solution of 4-chloropyrimidin-2-amine (1.0 g, 7.75 mmol) in DMSO (12 mL) was added and the reaction was heated to 60 °C overnight. The mixture was diluted with water (80 mL) and extracted with EtOAc (80 mL × 2), The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by RP-column (47 % acetonitrile in water) to afford 4-(but-2-yn-1-yloxy)pyrimidin-2-amine (800 mg, 63 %) as a yellow solid. LCMS m/z = 164.10 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.98 (d, J = 5.6, 1H), 6.58 (s, 2H), 6.01 (d, J = 5.6, 1H), 4.88 (d, J = 2.4, 2H), 1.83 (t, J = 2.4, 3H). [0001222] Example 1.133. Synthesis of 1-cyclobutyl-4-cyclopropyl-1H-pyrrolo[2,3- b]pyridin-5-amine
[0001223] Step 1: 4-cyclopropyl-5-nitro-1H-pyrrolo[2,3-b]pyridine [0001224] To a solution of 4-chloro-5-nitro-1H-pyrrolo[2,3-b]pyridine (500 mg, 2.54 mmol) in THF (12 mL) was added Pd(dppf)Cl2 (178 mg, 0.25 mmol), potassium phosphate tribasic (1.07 g, 5.08 mmol) and cyclopropylboronic acid (436 mg, 5.08 mmol). The reaction was heated at 100 °C in a Microwave reactor for 8 h. The mixture was diluted with water (40 mL) and extracted with EtOAc (40 mL × 2), the combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep-TLC (DCM: MeOH = 20:1) to afford 4-cyclopropyl-5-nitro-1H-pyrrolo[2,3-b]pyridine (160 mg, 31 %) as a yellow solid. LCMS m/z = 204.05 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 12.30 (s, 1H), 8.72 (s, 1H), 7.66 (d, J = 2.8, 1H), 6.73 – 6.68 (m, 1H), 1.18 – 1.12 (m, 2H), 0.96 – 0.90 (m, 2H). [0001225] Step 2: 1-cyclobutyl-4-cyclopropyl-5-nitro-1H-pyrrolo[2,3-b]pyridine [0001226] To a solution of 4-cyclopropyl-5-nitro-1H-pyrrolo[2,3-b]pyridine (160 mg, 0.79 mmol) in DMF (2 mL) was added cesium carbonate (513 mg, 1.58 mmol) and bromocyclobutane (160 mg, 1.18 mmol). The reaction was heated at 100 °C overnight in a sealed tube then was diluted with water (40 mL) and extracted with EtOAc (40 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was
purified by prep-TLC (eluent: Pet. ether: EtOAc = 10:1) to afford 1-cyclobutyl-4-cyclopropyl-5- nitro-1H-pyrrolo[2,3-b]pyridine (100 mg, 49 %) as a white solid. LCMS m/z = 258.10 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.75 (s, 1H), 8.02 (d, J = 3.6, 1H), 6.79 (d, J = 3.8, 1H), 5.31 (t, J = 8.6, 1H), 2.54 (d, J = 9.6, 2H), 2.43 (t, J = 5.0, 2H), 2.02 – 1.95 (m, 1H), 1.89 – 1.80 (m, 2H), 1.18 – 1.13 (m, 2H), 0.95 – 0.89 (m, 2H). [0001227] Step 3: 1-cyclobutyl-4-cyclopropyl-1H-pyrrolo[2,3-b]pyridin-5-amine [0001228] To a solution of 1-cyclobutyl-4-cyclopropyl-5-nitro-1H-pyrrolo[2,3-b]pyridine (100 mg, 0.39 mmol) in a mixture of ethanol (5 mL) and water (1 mL) was added Fe power (163 mg, 2.92 mmol) and NH4Cl (31 mg, 0.58 mmol). The reaction was heated at reflux for 2 h. the reaction was filtered and the filtrate diluted with water (30 mL) and extracted with EtOAc (30 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated to afford 1-cyclobutyl-4-cyclopropyl-1H-pyrrolo[2,3-b]pyridin-5-amine (88 mg, quant.) as a red oil which was used directly in the next step. LCMS m/z = 228.15 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.74 (s, 1H), 7.50 (d, J = 3.6, 1H), 6.32 (d, J = 3.4, 1H), 5.13 (t, J = 8.6, 1H), 4.55 (s, 2H), 2.47 – 2.41 (m, 2H), 2.39 – 2.33 (m, 2H), 2.02 – 1.95 (m, 1H), 1.79 (d, J = 4.8, 2H), 1.00 (dd, J = 8.4, 2.0, 2H), 0.71 (dd, J = 5.6, 1.8, 2H). [0001229] Example 1.134. Synthesis of 4-ethyl-6-methoxypyrimidin-2-amine
[0001230] To a solution of 4-chloro-6-ethylpyrimidin-2-amine (500 mg, 3.18 mmol) in THF (6 mL) was added sodium methoxide solution (5.4 M in MeOH, 1.6 mL, 7.96 mmol). The reaction heated at 60 °C for 3 h then was diluted with water (60 mL) and extracted with EtOAc (60 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated to afford 4-ethyl-6-methoxypyrimidin-2-amine (480 mg, 99 %) as yellow oil which was used directly in the next step. LCMS m/z = 154.15 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 6.42 (s, 2H), 5.87 (s, 1H), 3.77 (s, 3H), 2.45 – 2.38 (m, 2H), 1.12 (t, J = 7.6, 3H). [0001231] Example 1.135. Synthesis of 4-((1-methylpyrrolidin-3-yl)oxy)pyrimidin-2-amine
[0001232] To a solution of 4-chloropyrimidin-2-amine (1.0 g, 7.72 mmol) in THF (20 mL) was added 1-methylpyrrolidin-3-ol (780 mg, 7.72 mmol) and potassium tert-butoxide (1.13 g, 10.03 mmol). The reaction was heated at reflux overnight, then diluted with EtOAc (20 mL) and washed with water (20 mL × 2). The organic layer was dried over Na2SO4, filtered and concentrated to give 4-((1-methylpyrrolidin-3-yl)oxy)pyrimidin-2-amine (1.0 g, 66.7 %) as a yellow solid. LCMS m/z = 195.15 [M+H]+; 1HNMR (400 MHz, DMSO-d6) δ 7.93 (d, J = 5.6 Hz, 1H), 5.94 (dd, J = 5.6, 1.0 Hz, 1H), 5.33 – 5.22 (m, 1H), 2.81 – 2.70 (m, 1H), 2.68 – 2.59 (m, 1H), 2.55 – 2.52 (m, 1H), 2.38 – 2.18 (m, 5H), 1.84 – 1.66 (m, 1H). [0001233] Example 1.136. Synthesis of 4-(methoxymethyl)pyrimidin-2-amine
[0001234] A solution of 2-chloro-4-(methoxymethyl)pyrimidine (200 mg, 1.26 mmol) in a solution of NH3 in MeOH (2 mL) was heated at 100 °C for 1 h in a sealed tube. The mixture was diluted with EtOAc (20 mL) and washed with water (20 mL× 2), dried over Na2SO4, filtered and concentrated to give 4-(methoxymethyl)pyrimidin-2-amine (120 mg, 68.6 %) as a white solid. LCMS m/z = 140.09 [M+H]+; 1HNMR (400 MHz, DMSO-d6) δ 8.20 (d, J = 5.0 Hz, 1H), 6.56 (d, J = 4.2 Hz, 3H), 4.24 (s, 2H), 3.34 (s, 3H). [0001235] Example 1.137. Synthesis of 4-cyclobutoxy-6-methoxypyrimidin-2-amine
[0001236] Step 1: 4-bromo-6-methoxypyrimidin-2-amine [0001237] To a solution of 4,6-dibromopyrimidin-2-amine (200 mg, 0.79 mmol) in MeOH (2 mL) was added sodium methoxide (5.4 M, 0.15 mL, 0.81 mmol) and the reaction stirred at room temperature overnight. The solvent was removed and the residue diluted with water (30 mL) and extracted with EtOAc (30 mL). The organic layer was washed with brine, dried over Na2SO4, filtered and concentrated to afford 4-bromo-6-methoxypyrimidin-2-amine (120 mg, 74.5 %) as a yellow solid. LCMS m/z = 203.95 [M+H]+; 1HNMR (400 MHz, DMSO-d6) δ 7.16 – 7.00 (m, 2H), 6.24 (s, 1H), 3.80 (s, 3H). [0001238] Step 2: 4-cyclobutoxy-6-methoxypyrimidin-2-amine
[0001239] To a solution of 4-bromo-6-methoxypyrimidin-2-amine (100 mg, 0.49 mmol) in THF (2 mL) was added cyclobutanol (42 mg, 0.588 mmol) and potassium tert-butoxide (165 mg, 1.47 mmol) and the reaction heated at reflux overnight. The mixture was diluted with EtOAc (20 mL) and washed with water (20 mL × 2) and the organic layer dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep-TLC (eluent: DCM: MeOH=20:1)) to afford 4-cyclobutoxy-6-methoxypyrimidin-2-amine (80 mg, 84 %) as a white solid. LCMS m/z = 196.15 [M+H]+; 1HNMR (400 MHz, DMSO-d6) δ 6.46 (s, 2H), 5.28 (s, 1H), 5.03 – 4.99 (m, 1H), 3.74 (s, 3H), 2.35 – 2.31 (m, 3H), 2.02 – 1.97 (m, 3H). [0001240] Example 1.138. Synthesis of 4-(sec-butoxy)-6-methoxypyrimidin-2-amine
[0001241] To a solution of 4-bromo-6-methoxypyrimidin-2-amine (200 mg, 0.98 mmol) in THF (3 mL) was added butan-2-ol (220 mg, 1.96 mmol) and potassium tert-butoxide (145 mg, 1.96 mmol) and the reaction heated at reflux overnight. The mixture was diluted with EtOAc (20 mL) and washed with water (20 mL× 2). The organic layer was dried over Na2SO4, filtered and concentrated and the residue obtained purified by prep-TLC (eluent: DCM: MeOH=20:1)) to afford 4-(sec-butoxy)-6-methoxypyrimidin-2-amine (60 mg, 30 %) as colorless oil. LCMS m/z = 198.15 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 6.45 (s, 2H), 5.28 (s, 1H), 5.06 – 4.97 (m, 1H), 3.74 (s, 3H), 1.64 – 1.49 (m, 2H), 1.19 (d, J = 6.2 Hz, 3H), 0.86 (t, J = 7.4 Hz, 3H). [0001242] Example 1.139. Synthesis of 6-cyclobutoxypyridin-2-amine
[0001243] To a solution of 6-chloropyridin-2-amine (200 mg, 0.16 mmol) in THF (3 mL) was added cyclobutanol (134 mg, 1.86 mmol) and potassium tert-butoxide (350 mg, 3.12 mmol) and the reaction heated at reflux overnight. The mixture was diluted with EtOAc (20 mL), washed with water (20 mL × 2) and the organic layer dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep-TLC (eluent: DCM: MeOH=20:1)) to afford 6- cyclobutoxypyridin-2-amine (120 mg, 46.9 %) as a colorless oil. LCMS m/z = 165.15 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.24 (t, J = 7.8 Hz, 1H), 5.96 (d, J = 7.8 Hz, 1H), 5.79 (d, J =
7.8 Hz, 1H), 5.74 (d, J = 3.2 Hz, 2H), 5.04 – 4.92 (m, 1H), 2.38 – 2.30 (m, 2H), 2.00 – 1.94 (m, 2H), 1.78 – 1.67 (m, 1H), 1.61 – 1.52 (m, 1H). [0001244] Example 1.140. Synthesis of 4-ethylpyrimidin-2-amine
[0001245] A solution of 2-chloro-4-ethylpyrimidine (100 mg, 0.70 mmol) in a solution of NH3 in MeOH (1 mL) was heated at 100 °C for 1 h in sealed tube. The mixture was diluted with EtOAc (20 mL), washed with water (20 mL× 2) and the organic layer dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep-TLC (eluent: Per ether : EtOAc = 1 : 1) to afford 4-ethylpyrimidin-2-amine (60 mg, 69 %) as a white solid. LCMS m/z = 124.15 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.09 (d, J = 5.0 Hz, 1H), 6.50 – 6.34 (m, 3H), 2.49 – 2.44 (m, 2H), 1.14 (t, J = 7.6 Hz, 3H). [0001246] Example 1.141. Synthesis of N2-cyclobutyl-N2-methylpyridine-2,5-diamine
[0001247] Step 1: N-cyclobutyl-N-methyl-5-nitropyridin-2-amine [0001248] To a solution of N-methylcyclobutanamine hydrochloride (182 mg, 1.51 mmol) in DCM (3 mL) was added DIPEA (382 mg, 3.78 mmol) and 2-chloro-5-nitropyridine (200 mg, 1.26 mmol) and the reaction stirred at room temperature for 2 h. The mixture was diluted with EtOAc (20 mL), washed with water (20 mL × 2) and the organic layer dried over Na2SO4, filtered and concentrated to afford N-cyclobutyl-N-methyl-5-nitropyridin-2-amine (180 mg, 69 %) as a colorless oil. LCMS m/z = 208.20 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.96 (d, J = 2.8 Hz, 1H), 8.25 – 8.18 (m, 1H), 6.74 (d, J = 9.6 Hz, 1H), 3.11 (s, 3H), 4.94 (s, 1H) , 2.28 – 2.14 (m, 4H), 1.74 – 1.62 (m, 2H). [0001249] Step 2: N2-cyclobutyl-N2-methylpyridine-2,5-diamine [0001250] To a solution of N-cyclobutyl-N-methyl-5-nitropyridin-2-amine (150 mg, 0.72 mmol) in a mixture of EtOH (3 mL) and water (0.5 mL) was added Fe (202 mg, 3.62 mmol) and NH4Cl (135 mg, 1.09 mmol) and the reaction heated at 80 °C for 3 h. The mixture was filtered and the filtrate concentrated to afford N2-cyclobutyl-N2-methylpyridine-2,5-diamine (110 mg, 64.7 %)
which was used directly in the next step. LCMS m/z = 178.24 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.57 (d, J = 2.8 Hz, 1H), 6.92 – 6.84 (m, 1H), 6.43 (d, J = 8.8 Hz, 1H), 4.56 – 4.31 (m, 3H), 2.77 (s, 3H), 2.12 – 1.99 (m, 4H), 1.66 – 1.53 (m, 2H). [0001251] Example 1.142. Synthesis of 4-(allyloxy)pyrimidin-2-amine
[0001252] To a solution of 4-chloropyrimidin-2-amine (1.0 g, 7.7 mmol) in THF (15 mL) was added prop-2-en-1-ol (900 mg, 15.4 mmol) and potassium tert-butoxide (1.3 g, 11.6 mmol) and the reaction heated at reflux for 24 h. The solvent was removed, the residue resuspended in DCM (50 mL) and filtered, the filtrate was concentrated to give 4-(allyloxy)pyrimidin-2-amine (900 mg, 77 %) as a white solid. LCMS m/z = 152.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.96 (d, J = 5.6 Hz, 1H), 6.51 (s, 2H), 6.10 – 5.95 (m, 2H), 5.35 (dd, J = 17.2, 1.8 Hz, 1H), 5.23 (dd, J = 10.5, 1.7 Hz, 1H), 4.75 (d, J = 5.4 Hz, 2H). [0001253] Example 1.143. Synthesis of (4-methoxypyrimidin-2-yl)methanamine
[0001254] To a solution of 4-methoxypyrimidine-2-carbonitrile (100 mg, 0.74 mmol) in MeOH (5.0 mL) was added 10% Pd/C (20 mg) and the reaction stirred under a hydrogen atmosphere at 50 °C for 3 h. The catalyst was removed by filtration through Celite and the filtrate concentrated to give (4-methoxypyrimidin-2-yl)methanamine (103 mg, quant.) as a brown solid. LCMS m/z = 140.2 [M+H]+. [0001255] Example 1.144. Synthesis of 1-(4-methoxypyrimidin-2-yl)ethan-1-amine
[0001256] Step 1: 1-(4-methoxypyrimidin-2-yl)ethan-1-one
[0001257] To a solution of 2-iodo-4-methoxypyrimidine (1.62 g, 6.9 mmol) in THF (20 mL) at - 10 °C was added isopropylmagnesium chloride (2 M in THF, 3.45 mL, 6.9 mmol) and the reaction stirred for 1 hr N-methoxy-N-methylacetamide (776 mg, 7.5 mmol) was added and the reaction allowed to warm to RT and stirred overnight. The mixture was diluted with water (20 mL) and extracted with DCM (20 mL × 2). The combined organic layers were washed with water and brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by column chromatography on silica gel (eluent: Pet. ether: EtOAc = 4: 1, v/v) to afford 1-(4- methoxypyrimidin-2-yl)ethan-1-one (460 mg, 44 %) as a yellow solid. LCMS m/z = 153.0 [M+H]+; 1H NMR (400 MHz, Chloroform-d) δ 8.58 (d, J = 5.6 Hz, 1H), 6.85 (d, J = 5.6 Hz, 1H), 4.07 (s, 3H), 2.74 (s, 3H). [0001258] Step 2 1-(4-methoxypyrimidin-2-yl)ethan-1-ol [0001259] To a solution of 1-(4-methoxypyrimidin-2-yl)ethan-1-one (200 mg, 1.31 mmol) in MeOH (5.0 mL) at 0 °C was added NaBH4 (149 mg, 3.94 mmol) portionwise., The reaction was stirred at room temperature for 2 h then diluted with water (20 mL) and extracted with DCM (20 mL × 2). The combined organic layers were washed with water, brine, dried over Na2SO4, filtered and concentrated to afford 1-(4-methoxypyrimidin-2-yl)ethan-1-ol (200 mg, 50 %) as a yellow oil. LCMS m/z = 155.0 [M+H]+; 1H NMR (400 MHz, Chloroform-d) δ 8.39 (d, J = 5.8 Hz, 1H), 6.61 (d, J = 5.8 Hz, 1H), 4.82 (q, J = 6.8 Hz, 1H), 4.00 (s, 3H), 1.55 (d, J = 6.6 Hz, 3H). [0001260] Step 3: 1-(4-methoxypyrimidin-2-yl)ethyl methanesulfonate [0001261] To a solution of 1-(4-methoxypyrimidin-2-yl)ethan-1-ol (100 mg, 0.65 mmol) in DCM (4.0 mL) at 0 °C was added TEA (197 mg, 1.95 mmol) and MsCl (96 mg, 0.84 mmol) and the reaction stirred at room temperature for 5 h. The mixture was diluted with water (20 mL) and extracted with DCM (20 mL × 2). The combined organic layers were washed with water, brine, dried over Na2SO4, filtered and concentrated to afford 1-(4-methoxypyrimidin-2-yl)ethyl methanesulfonate (150 mg, quant.) as a yellow oil. LCMS m/z = 233.0 [M+H]+; 1H NMR (400 MHz, Chloroform-d) δ 8.44 (d, J = 5.8 Hz, 1H), 6.68 (d, J = 5.8 Hz, 1H), 5.73 (q, J = 6.6 Hz, 1H), 4.00 (s, 3H), 3.08 (s, 3H), 1.78 (d, J = 6.6 Hz, 3H). [0001262] Step 4: 2-(1-azidoethyl)-4-methoxypyrimidine [0001263] To a solution of 1-(4-methoxypyrimidin-2-yl)ethyl methanesulfonate (270 mg, 1.16 mmol) in DMF (4.0 mL) was added sodium azide (226 mg, 3.49 mmol) and the reaction stirred at room temperature for 24 h. The mixture was diluted with water (20 mL) and extracted with
EtOAc (20 mL × 2). The combined organic layers were washed with water, brine, dried over Na2SO4, filtered and concentrated to afford 2-(1-azidoethyl)-4-methoxypyrimidine (130 mg, 62 %) as yellow oil. LCMS m/z = 180.0 [M+H]+; 1H NMR (400 MHz, Chloroform-d) δ 8.40 (d, J = 5.8 Hz, 1H), 6.65 (d, J = 5.8 Hz, 1H), 4.46 (q, J = 6.8 Hz, 1H), 4.02 (s, 3H), 1.64 (dd, J = 6.8, 1.0 Hz, 3H). [0001264] Step 5: 1-(4-methoxypyrimidin-2-yl)ethan-1-amine [0001265] To a solution of 2-(1-azidoethyl)-4-methoxypyrimidine (110 mg, 0.61 mmol) in MeOH (3.0 mL) was added 10% Pd/C (20 mg) and the reaction stirred under a hydrogen atmosphere for 5 h. The catalyst was removed by filtration through Celite and the filtrate concentrated give 1-(4-methoxypyrimidin-2-yl)ethan-1-amine ( 94 mg, quant.) as a yellow oil. LCMS m/z = 154.0 [M+H]+. [0001266] Example 1.145. Synthesis of 5-cyclobutoxypyrimidin-2-amine
[0001267] To a solution of 2-aminopyrimidin-5-ol (500 mg, 4.5 mmol) in toluene (5.0 mL) at 0 °C was added cyclobutanol (486 mg, 6.75 mmol), triphenylphosphine (1.77 g, 6.75 mmol) and DIAD (1.37 g, 6.75 mmol) and the reaction was heated at 80 °C for 3 h. The mixture was partitioned between 0.5 M HCl (20 mL) diethyl ether (20 mL) and extracted with diethyl ether (20 mL x 2). The pH of the aqueous was adjusted ~ 12 by addition of 6 M NaOH and extracted with EtOAc (20 mL x 3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated to afford 5-cyclobutoxypyrimidin-2-amine (253 mg, 34 %) as a yellow oil. LCMS m/z = 166 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.94 (s, 2H), 6.17 (s, 2H), 4.70 – 4.45 (m, 1H), 2.39 – 2.26 (m, 2H), 2.06 – 1.91 (m, 2H), 1.80 – 1.48 (m, 2H). [0001268] Example 1.146. Synthesis of 4-propoxypyrimidin-2-amine
[0001269] A solution of sodium metal (89 mg, 3.87 mmol) in propan-1-ol (2 mL) was heated at 80 °C for 15 min.4-chloropyrimidin-2-amine (250 mg, 1.93 mmol) and TBAI (12.5 mg, cat.) were added and the reaction was heated at reflux for 6 h. The solvent was concentrated and
purified by RP-column (45% MeCN in water) to afford 4-propoxypyrimidin-2-amine (150 mg, 51%) as a yellow solid. LCMS m/z = 154.1 [M+H]+. [0001270] Example 1.147. Synthesis of 4-(neopentyloxy)pyrimidin-2-amine
[0001271] To a solution of 4-chloropyrimidin-2-amine (1.0 g, 7.72 mmol) in THF (10 mL) was added t-BuOK (1.125 g, 10.03 mmol) and 2,2-dimethylpropan-1-ol (681 mg, 7.72 mmol) and the reaction heated at reflux overnight. The solvent was removed under reduced pressure and the residue obtained purified by RP-column (50% MeCN in water) to afford 4- (neopentyloxy)pyrimidin-2-amine (700 mg, 50 %) as a yellow solid. LCMS m/z =182.15 [M+H]+. [0001272] Example 1.148. Synthesis of 4-(2-((tert-butyldimethylsilyl)oxy)ethoxy) pyrimidin-2-amine
[0001273] Step 1: 2-((2-aminopyrimidin-4-yl)oxy)ethan-1-ol [0001274] To a solution of ethane-1,2-diol (1.92 g, 31.00 mmol) in 1,4-dioxane (12 mL) was added t-BuOK (1.31 g, 11.63 mmol) and the reaction stirred at room temperature for 30 min. A solution of 4-chloropyrimidin-2-amine (1.0 g, 7.75 mmol) in DMSO (12 mL) was added and the reaction was heated at 60 °C overnight. The mixture was filtered and the filtrate was concentrated, the residue obtained was purified by reverse column (12% acetonitrile in water) to afford 2-((2-aminopyrimidin-4-yl)oxy)ethan-1-ol (1.0 g, 83 %) as a white solid. LCMS m/z = 156.10 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.94 (d, J = 5.6, 1H), 6.48 (s, 2H), 5.98 (d, J = 5.6, 1H), 4.23 – 4.18 (m, 2H), 3.66 (t, J = 5.2, 2H). [0001275] Step 2: 4-(2-((tert-butyldimethylsilyl)oxy)ethoxy)pyrimidin-2-amine [0001276] To a solution of 2-((2-aminopyrimidin-4-yl)oxy)ethan-1-ol (900 mg, 5.80 mmol) in DMF (8 mL) was added imidazole (593 mg, 8.71 mmol), DMAP (1.06 g, 8.71 mmol) and TBSCl (1.3 g, 8.71 mmol) and the reaction stirred at room temperature overnight. The mixture was diluted with water (80 mL) and extracted with DCM (80 mL × 2), the combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was
purified by reverse column (87 % acetonitrile in water) to afford 4-(2-((tert- butyldimethylsilyl)oxy)ethoxy)pyrimidin-2-amine (700 mg, 45 %) as a pink solid. LCMS m/z = 270.10 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.94 (d, J = 5.6, 1H), 6.49 (s, 2H), 5.96 (d, J = 5.6, 1H), 4.25 (dd, J = 5.8, 4.0, 2H), 3.86 (dd, J = 5.8, 4.2, 2H), 0.85 (s, 9H), 0.04 (s, 6H). [0001277] Example 1.149. Synthesis of 4-((tetrahydrofuran-3-yl)methyl)pyrimidin-2-amine
[0001278] Step 1: 4-(bromomethyl)pyrimidin-2-amine [0001279] To a solution of (2-aminopyrimidin-4-yl)methanol (900 mg, 7.19 mmol) in THF (10 mL) was added CBr4 (2.86 g, 8.63 mmol) and PPh3 (2.26 g, 8.63 mmol) and the reaction stirred at room temperature for 2 h. The mixture was filtered and concentrated to afford 4- (bromomethyl)pyrimidin-2-amine (540 mg, 40 %) as a white solid. LCMS m/z = 188.05 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.24 (d, J = 5.0 Hz, 1H), 6.73 (s, 2H), 6.68 (d, J = 5.0 Hz, 1H), 4.36 (s, 2H). [0001280] Step 2: diethyl ((2-aminopyrimidin-4-yl)methyl)phosphonate [0001281] To a solution of 4-(bromomethyl)pyrimidin-2-amine (50 mg, 0.27 mmol) in toluene (3 mL) was added triethyl phosphite (133 mg, 0.8 mmol) and the reaction heated at reflux for 3 h under a N2 atmosphere. The mixture was concentrated to afford diethyl ((2-aminopyrimidin-4- yl)methyl)phosphonate (65 mg, quant.) which was used directly in the next step without purification. LCMS m/z = 246.05 [M+H]+. [0001282] Step 3: (E)-4-((dihydrofuran-3(2H)-ylidene)methyl)pyrimidin-2-amine [0001283] To a solution of diethyl ((2-aminopyrimidin-4-yl)methyl)phosphonate (130 mg, 0.53 mmol) in anhydrous THF (3 mL) at 0 °C under a N2 atmosphere was added sodium hydride (60 %, 42 mg, 1.05 mmol) and the reaction stirred for 0.5 h. Dihydrofuran-3(2H)-one (46 mg, 0.53 mmol) was added and the reaction allowed to warm to room temperature and stirred for another 2 h. The mixture was diluted with water (20 mL) and extracted with EtOAc (30 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep-TLC (DCM: MeOH = 10: 1) to afford (E)-4-
((dihydrofuran-3(2H)-ylidene)methyl)pyrimidin-2-amine (60 mg, 64 %) as a yellow oil. LCMS m/z = 178.15[M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.11 (d, J = 5.0 Hz, 1H), 6.49 (s, 2H), 6.43 (d, J = 5.0 Hz, 1H), 6.32 – 6.28 (m, 1H), 4.23 (t, J = 9.6 Hz, 2H), 3.23 (s, 2H), 2.47 – 2.41 (m, 2H). [0001284] Step 4: 4-((tetrahydrofuran-3-yl)methyl)pyrimidin-2-amine [0001285] To a solution of (E)-4-((dihydrofuran-3(2H)-ylidene)methyl)pyrimidin-2-amine (50 mg, 0.286 mmol) in a mixture of MeOH (1.5 mL) and DCM (0.5 mL) was added 10% Pd/C (25 mg) and the reaction stirred under a hydrogen atmosphere for 2 h. The catalyst was removed by filtration through Celite and the filtrate concentrated to afford 4-((tetrahydrofuran-3- yl)methyl)pyrimidin-2-amine (40 mg, 79 %) which was used directly in the next step without further purification. LCMS m/z = 180.15[M+H]+. [0001286] Example 1.150. Synthesis of cyclobutyl-3-fluoro-4-methyl-1H-pyrrolo[2,3- b]pyridin-5-amine
[0001287] Step 1: 4-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine [0001288] To a solution of 4-chloro-5-nitro-1H-pyrrolo[2,3-b]pyridine (200 mg, 1.01mmol) in a mixture of 1,4-dioxane (4 mL) and water (1 mL) was added trimethylboroxine (1.44 mL, 5.05 mmol), K2CO3 (279 mg, 2.02 mmol) and Pd(PPh3)4 (58 mg, 0.05 mmol) and the reaction heated at reflux for 3 h. The mixture was diluted with water (20 mL) and extracted with EtOAc (20 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated to afford 4-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine (130 mg, 72 %) as a brown solid.1H NMR (400 MHz, DMSO-d6) δ 12.34 (s, 1H), 8.90 (s, 1H), 7.70 – 7.66 (m, 1H), 6.87 – 6.83 (m, 1H), 2.81 (s, 3H). [0001289] Step 2: 3-fluoro-4-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine
[0001290] To a solution of 4-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine (150 mg, 0.847 mmol) in a mixture of MeCN (21 mL) and AcOH (4.5 mL) was added selectfluor (450 mg, 1.27 mmol) and the reaction heated at 80 °C overnight. The reaction was concentrated, diluted with water (20 mL) and extracted with EtOAc (20 mL× 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep- TLC (Pet. ether/EtOAc = 5/1) to afford 3-fluoro-4-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine (60 mg, 36 %) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ 12.47 – 12.32 (m, 1H), 9.14 – 9.11 (m, 1H), 8.92 – 8.89 (m, 1H), 7.82 – 7.79 (m, 1H). LCMS m/z =196.05 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 12.24 (s, 1H), 8.91 (s, 1H), 7.71 (s, 1H), 2.87 (s, 3H). [0001291] Step 3: 1-cyclobutyl-3-fluoro-4-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine [0001292] To a solution of 3-fluoro-4-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine (60 mg, 0.307 mmol) in DMF (2 mL) was added cyclobutanol (33 mg, 0.461 mmol), PPh3 (161 mg, 0.615 mmol) and DIAD (124 mg, 0.615 mmol) and the reaction heated at 80 °C overnight. The mixture was diluted with water (10 mL) and extracted with EtOAc (20 mL × 3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep-TLC (Pet. Ether/EtOAc = 5/1) to afford 1-cyclobutyl-3-fluoro-4- methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine (30 mg, 39 %) as a yellow solid. LCMS m/z = 250.1 [M+H]+ [0001293] Step 4: 1-cyclobutyl-3-fluoro-4-methyl-1H-pyrrolo[2,3-b]pyridin-5-amine [0001294] To a solution of 1-cyclobutyl-3-fluoro-4-methyl-5-nitro-1H-pyrrolo[2,3-b]pyridine (5 mg, 0.02 mmol) in a mixture of EtOH (2 mL) and water (0.5 mL) was added iron powder (6 mg, 0.107 mmol) and NH4Cl (6 mg, 0.112 mmol) and the reaction stirred at room temperature overnight. The mixture was filtered and the filtrate was diluted with water (10 mL) and extracted with EtOAc (20 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4 and concentrated to afford 1-cyclobutyl-3-fluoro-4-methyl-1H-pyrrolo[2,3-b]pyridin-5- amine (3 mg, 68 %) as a yellow solid. LCMS m/z = 220.1 [M+H]+. [0001295] Example 1.151. Synthesis of 4,6-dicyclobutoxypyrimidin-2-amine
[0001296] To a solution of 4,6-dibromopyrimidin-2-amine (200 mg, 0.79 mmol) in THF (5 mL) under a N2 atmosphere was added t-BuOK (266 mg, 2.37 mmol) and cyclobutanol (171 mg, 2.37 mmol) and the reaction heated at 80 °C overnight. The mixture was concentrated and purified by RP-column (50% MeCN in water) to afford 4,6-dicyclobutoxypyrimidin-2-amine (149 mg, 80 %) as a yellow solid. LCMS m/z = 236.10 [M+H]+. [0001297] Example 1.152. Synthesis of 4-(cyclobutylmethyl)pyrimidin-2-amine
[0001298] Step 1: 4-(bromomethyl)pyrimidin-2-amine [0001299] To a solution of (2-aminopyrimidin-4-yl)methanol (1.0 g, 8.0 mmol) in THF (10 mL) was added CBr4 (3.18 g, 9.6 mmol) and PPh3 (2.52 g, 9.6 mmol) and the reaction stirred at room temperature for 2 h. The mixture was filtered and concentrated to afford 4- (bromomethyl)pyrimidin-2-amine (700 mg, 47 %) as a white solid. LCMS m/z = 188.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.23 (d, J = 5.0 Hz, 1H), 6.73 (s, 2H), 6.67 (d, J = 5.0 Hz, 1H), 4.36 (s, 2H). [0001300] Step 2: diethyl ((2-aminopyrimidin-4-yl)methyl)phosphonate [0001301] To a solution of 4-(bromomethyl)pyrimidin-2-amine (700 mg, 3.72 mmol) in toluene (15 mL) was added triethyl phosphite (1.86 g, 11.17 mmol) and the reaction heated at reflux for 3 h under a N2 atmosphere. The mixture was concentrated to afford diethyl ((2-aminopyrimidin- 4-yl)methyl)phosphonate (913 mg, 100 %) which was used directly in the next step. LCMS m/z = 246.10 [M+H]+. [0001302] Step 3: 4-(cyclobutylidenemethyl)pyrimidin-2-amine [0001303] To a solution of diethyl ((2-aminopyrimidin-4-yl)methyl)phosphonate (400 mg, 1.63 mmol) in anhydrous THF (5 mL) at 0 °C was added sodium hydride (130 mg, 3.25 mmol) and the reaction stirred at 0 °C for 30 min. Cyclobutanone (126 mg, 1.8 mmol) was added and the reaction allowed to warm to room temperature and stirred for another 2 h. The mixture was diluted with water (20 mL) and extracted with EtOAc (30 mL × 3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was
purified by prep-TLC (DCM/MeOH = 10/1) to afford 4-(cyclobutylidenemethyl)pyrimidin-2- amine (80 mg, 30 %) as a yellow solid. LCMS m/z = 162.15 [M+H]+. [0001304] Step 4: 4-(cyclobutylmethyl)pyrimidin-2-amine [0001305] To a solution of 4-(cyclobutylidenemethyl)pyrimidin-2-amine (80 mg, 0.50 mmol) in a mixture of MeOH (1.5mL) and DCM (0.5 mL) was added 10% Pd/C (40 mg) and the reaction stirred under a H2 atmosphere for 2 h. The catalyst was removed by filtration and the filtrate concentrated to afford 4-(cyclobutylmethyl)pyrimidin-2-amine (50 mg, 62 %) which was used directly in the next step without further purification. LCMS m/z = 164.15 [M+H]+. [0001306] Example 1.153. Synthesis of 4-((tetrahydro-2H-pyran-4-yl)methyl)pyrimidin-2- amine
[0001307] Step 1: 4-((tetrahydro-4H-pyran-4-ylidene)methyl)pyrimidin-2-amine [0001308] To a solution of diethyl ((2-aminopyrimidin-4-yl)methyl)phosphonate (400 mg, 1.63 mmol) in anhydrous THF (5 mL) at 0 °C under a N2 atmosphere was added sodium hydride (60 %, 130 mg, 3.25 mmol) and the reaction stirred at 0 °C for 30 min. Tetrahydro-4H-pyran-4-one (163 mg, 1.63 mmol) was added and the reaction allowed to warm to room temperature and stirred for another 2 h. The mixture was diluted with water (20 mL) and extracted with EtOAc (30 mL × 3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep-TLC (DCM: MeOH = 10: 1) to afford 4-((tetrahydro-4H-pyran-4-ylidene)methyl)pyrimidin-2-amine (90 mg, 29 %) as a yellow solid. LCMS m/z = 191.10 [M+H]+. [0001309] Step 2: 4-((tetrahydro-2H-pyran-4-yl)methyl)pyrimidin-2-amine [0001310] To a solution of 4-((tetrahydro-4H-pyran-4-ylidene)methyl)pyrimidin-2-amine (80 mg, 0.418 mmol) in a mixture of MeOH (1.5 mL) and DCM (0.5 mL) was added 10% Pd/C (40 mg) and the reaction stirred under a hydrogen atmosphere for 2 h. The catalyst was removed by filtration through Celite and the filtrate concentrated to afford 4-((tetrahydro-2H-pyran-4- yl)methyl)pyrimidin-2-amine (80 mg, 99 %) which was used directly in the next step without further purification. LCMS m/z = 194.10[M+H]+. [0001311] Example 1.154. Synthesis of 4,6-dimethoxypyridin-2-amine
[0001312] To a solution of 4,6-dichloropyridin-2-amine (500 mg, 3.07 mmol) in NMP (10 mL) under a N2 atmosphere was added sodium methoxide (1.66 g, 30.7 mmol) and the reaction heated at 120 °C overnight. The mixture was diluted with water (20 mL) and extracted with EtOAc (30 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated to afford 4,6-dimethoxypyridin-2-amine (340 mg, 72 %) as a brown solid. LCMS m/z = 155.15[M+H]+. [0001313] Example 1.155. Synthesis of 1-cyclobutyl-3-fluoro-1H-pyrrolo[2,3-b]pyridin-5- amine
[0001314] Step 1: 3-fluoro-5-nitro-1H-pyrrolo[2,3-b]pyridine [0001315] To a solution of 5-nitro-1H-pyrrolo[2,3-b]pyridine (1.8 g, 11.03 mmol) in a mixture of MeCN (260 mL) and AcOH (54 mL) was added selectfluor (5.9 g, 16.55 mmol) and the mixture heated at 80 °C overnight. The reaction was concentrated, diluted with water (200 mL) and extracted with EtOAc (150 mL× 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by column chromatography on silica gel (Pet. ether/EtOAc = 5/1) to afford 3-fluoro-5-nitro-1H- pyrrolo[2,3-b]pyridine (1.3 g, 65 %) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ 12.40 (s, 1H), 9.13 (d, J = 2.5 Hz, 1H), 8.91 (s, 1H), 7.81 (s, 1H). [0001316] Step 2: 1-cyclobutyl-3-fluoro-5-nitro-1H-pyrrolo[2,3-b]pyridine [0001317] To a solution of 3-fluoro-5-nitro-1H-pyrrolo[2,3-b]pyridine (100 mg, 0.55 mmol) in DMF (3 mL) was added bromocyclobutane (112 mg, 0.83 mmol) and Cs2CO3 (360 mg, 1.10 mmol) and the reaction heated at 100 °C overnight. The mixture was filtered, concentrated and the residue obtained was purified by Prep-TLC (eluent: Pet. ether: EtOAc = 2: 1) to afford 1- cyclobutyl-3-fluoro-5-nitro-1H-pyrrolo[2,3-b]pyridine (50 mg, 38 %) as a yellow solid. LCMS m/z = 236 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 9.15 (d, J = 2.4 Hz, 1H), 8.91 (d, J = 2.4 Hz, 1H), 8.23 (d, J = 1.8 Hz, 1H), 5.47 – 5.35 (m, 1H), 2.49 – 2.35 (m, 4H), 1.88 – 1.81 (m, 2H).
[0001318] Step 3: 1-cyclobutyl-3-fluoro-1H-pyrrolo[2,3-b]pyridin-5-amine [0001319] To a solution of 1-cyclobutyl-3-fluoro-5-nitro-1H-pyrrolo[2,3-b]pyridine (50 mg, 0.21 mmol) in MeOH (3.0 mL) was added 10% Pd/C (10 mg) and the reaction stirred under a hydrogen atmosphere for 5 h. The catalyst was removed by filtration through Celite and the filtrate concentrated to give 1-cyclobutyl-3-fluoro-1H-pyrrolo[2,3-b]pyridin-5-amine ( 44 mg, quant.) as a yellow oil. LCMS m/z = 206 [M+H]+ [0001320] Example 1.156. Synthesis of 4-cyclobutoxypyridin-2-amine
[0001321] To a solution of 4-chloropyridin-2-amine (1.0 g, 7.78 mmol) in THF (10 mL) was added t-BuOK (1.13 g, 10.11 mmol) and cyclobutanol (673 mg, 9.33 mmol) and the reaction heated at 80 °C overnight. The mixture was concentrated under reduced pressure and the residue obtained purified by RP-column (50 % MeCN in water) to afford 4-cyclobutoxypyridin-2-amine (60 mg, 4.7 %) as a white solid. LCMS m/z = 165.15 [M+H]+. [0001322] Example 1.157. Synthesis of (R)-1-(3-chloropyridin-2-yl)-2,2,2-trifluoroethan-1- amine
[0001323] Step 1: (S)-N-((3-chloropyridin-2-yl)methylene)-2-methylpropane-2-sulfinamide [0001324] To a solution of 3-chloropicolinaldehyde (5.0 g, 35.32 mmol) in DCM (100 mL) was added (S)-2-methylpropane-2-sulfinamide (4.28 g, 35.32 mmol) and Cs2CO3 (13.8 g, 42.38 mmol) and the reaction stirred at room temperature overnight. The mixture was diluted with water (260 mL) and extracted with DCM (250 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated to afford (S)-N-((3- chloropyridin-2-yl)methylene)-2-methylpropane-2-sulfinamide (8.6 g, quant.) as a yellow oil. LCMS m/z = 245.1 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 9.06 – 9.04 (m, 1H), 8.70 – 8.68 (m, 1H), 7.83 – 7.79 (m, 1H), 7.38 – 7.33 (m, 1H), 1.30 (s, 9H). [0001325] Step 2: (S)-N-((R)-1-(3-chloropyridin-2-yl)-2,2,2-trifluoroethyl)-2-methylpropane-2- sulfinamide
[0001326] To a solution of (S)-N-((3-chloropyridin-2-yl)methylene)-2-methylpropane-2- sulfinamide (4.0 g, 16.34 mmol) and tetrabutylammonium difluorotriphenylsilicate (1.8 g, 3.27 mmol) in anhydrous THF (50 mL) at -78 °C was added (trifluoromethyl)trimethylsilane (4.7 g, 32.68 mmol) and the reaction stirred for 2 h. The reaction was allowed to warm to -10 °C and stirring continued for 2 h then the mixture was diluted with EtOAc (400 mL) and washed with water (200 mL × 2), dried over Na2SO4, filtered and concentrated. The residue obtained was purified by column chromatography on silica gel (eluent: Pet. Ether: EtOAc = 10:1 to 1:1) to afford (S)-N-((R)-1-(3-chloropyridin-2-yl)-2,2,2-trifluoroethyl)-2-methylpropane-2-sulfinamide (2.9 g, 56 %) as a yellow solid. LCMS m/z =315.0 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.69 – 8.63 (m, 1H), 8.12 – 8.08 (m, 1H), 7.59 – 7.53 (m, 1H), 6.07 – 6.02 (m, 1H), 5.60 – 5.51 (m, 1H), 1.14 (s, 9H). [0001327] Step 3: (R)-1-(3-chloropyridin-2-yl)-2,2,2-trifluoroethan-1-amine [0001328] To a solution of (S)-N-((R)-1-(3-chloropyridin-2-yl)-2,2,2-trifluoroethyl)-2- methylpropane-2-sulfinamide (2.2 g, 6.99 mmol) in DCM (20 mL) was added HCl (4 M in 1,4- dioxane, 20 mL) and the reaction stirred at room temperature for 3 h. The solvent was removed under vacuum to afford (R)-1-(3-chloropyridin-2-yl)-2,2,2-trifluoroethan-1-amine (1.4 g, quant.) as a yellow solid. LCMS m/z = 211.1 [M+H]+. [0001329] Example 1.158. Synthesis of 2-(bromomethyl)-4,6-dimethoxypyrimidine
[0001330] To a solution of 4,6-dimethoxy-2-methylpyrimidine (200 mg, 1.3 mmol) in carbon tetrachloride (3 mL) was added N-bromosuccinimide (231 mg, 1.3 mmol) and 2,2'-Azobis(2- methylpropionitrile) (11 mg, 64.9 mmol) and the reaction heated at 100 °C overnight. The mixture was diluted with EtOAc (30 mL) and washed with water (20 mL × 2), dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep-TLC (eluent: Pet. Ether: EtOAc = 10:1,v/v) to afford 2-(bromomethyl)-4,6-dimethoxypyrimidine (50 mg, 16 %) as a yellow oil. LCMS m/z =233 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 6.19 (s, 1H), 4.47 (s, 2H), 3.89 (s, 6H). [0001331] Example 1.159. Synthesis of 6-cyclobutoxy-4-ethylpyridin-3-amine
[0001332] Step 1: 4-chloro-2-cyclobutoxy-5-nitropyridine [0001333] To a solution of 2,4-dichloro-5-nitropyridine (50 mg, 0.26 mmol) in MeCN (2 mL) was added cesium carbonate (254 mg, 0.78 mmol) and cyclobutanol (0.3 mL, 3.83 mmol) and the reaction stirred at room temperature overnight. The mixture was filtered and concentrated and the residue obtained purified by prep-TLC (Pet. ether : EtOAc = 5:1) to afford 4-chloro-2- cyclobutoxy-5-nitropyridine (45 mg, 76 %) as a white solid.1H NMR (400 MHz, Methanol-d4) δ 8.78 (s, 1H), 7.21 (s, 1H), 5.07 – 4.95 (m, 1H), 2.60 – 2.52 (m, 2H), 2.30 – 2.20 (m, 2H), 1.99 – 1.89 (m, 1H), 1.85 – 1.74 (m, 1H). [0001334] Step 2: 2-cyclobutoxy-4-ethyl-5-nitropyridine [0001335] To a solution of 4-chloro-2-cyclobutoxy-5-nitropyridine (40 mg, 0.14 mmol) in DMF (1 mL) was added triethylborane (1.0 M in THF, 0.14 mL, 0.14 mmol), Pd(dppf)Cl2.DCM (11 mg, 0.014 mmol) and cesium carbonate (136 mg, 0.42 mmol). The reaction was stirred at room temperature for 3 h under N2 atmosphere then was diluted with water (10 mL) and extracted with EtOAc (10 mL × 3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue was purified by prep-TLC (Pet. ether : EtOAc = 5:1) to afford 2-cyclobutoxy-4-ethyl-5-nitropyridine (15 mg, 48 %) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ 8.83 (s, 1H), 7.00 (s, 1H), 5.05 – 4.96 (m, 1H), 2.83 (q, J = 7.6 Hz, 2H), 2.60 – 2.53 (m, 2H), 2.29 – 2.19 (m, 2H), 1.98 – 1.90 (m, 1H), 1.84 – 1.75 (m, 1H), 1.29 (d, J = 7.6 Hz, 3H). [0001336] Step 3: 6-cyclobutoxy-4-ethylpyridin-3-amine [0001337] To a solution of 2-cyclobutoxy-4-ethyl-5-nitropyridine (600 mg, 2.7 mmol) in methanol (12 mL) was added 10 % Pd/C (120 mg) and the reaction stirred at room temperature for 2 h under H2 atmosphere. The catalyst was removed by filtration through Celite and the filtrate concentrated to afford 6-cyclobutoxy-4-ethylpyridin-3-amine (519 mg, 100 %) as a brown solid. LCMS m/z = 193.28 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.74 (s, 1H), 6.49
(s, 1H), 4.79 – 4.69 (m, 1H), 4.53 (s, 2H), 2.56 – 2.51 (m, 2H), 2.47 – 2.40 (m, 2H), 2.09 – 2.00 (m, 2H), 1.83 – 1.75 (m, 1H), 1.69 – 1.61 (m, 1H), 1.13 (t, J = 7.6 Hz, 3H). [0001338] Example 1.160. Synthesis of 6-cyclopropylpyridazin-3-amine
[0001339] Step 1: tert-butyl (6-cyclopropylpyridazin-3-yl)carbamate [0001340] To a solution of 3-chloro-6-cyclopropylpyridazine (200 mg, 1.29 mmol) in 1,4- dioxane (6 mL) was added cesium carbonate (843 mg, 2.59 mmol), tert-butyl carbamate (227 mg, 1.94 mmol) and Pd(OAc)2 (29 mg, 0.13 mmol). The reaction was heated at 80 °C for 2 h then was diluted with water (50 mL) and extracted with EtOAc (60 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep-TLC (Petroleum ether: EtOAc, 5:1) to afford tert-butyl (6- cyclopropylpyridazin-3-yl)carbamate (250 mg, 82 %) as a white solid. LCMS m/z = 236.10 [M+H]+ ; 1H NMR (400 MHz, DMSO-d6) δ 10.20 (s, 1H), 7.89 (d, J = 9.2 Hz, 1H), 7.41 (d, J = 9.2 Hz, 1H), 2.17 (s, 1H), 1.47 (s, 9H), 1.02 (d, J = 5.8 Hz, 2H), 0.94 (d, J = 4.8 Hz, 2H). [0001341] Step 2: 6-cyclopropylpyridazin-3-amine [0001342] To a solution of tert-butyl (6-cyclopropylpyridazin-3-yl)carbamate (250 mg, 1.06 mmol) in DCM (2 mL) was added TFA (1 mL) and the reaction stirred at room temperature for 1 h. The solvent was removed under reduced pressure to afford 6-cyclopropylpyridazin-3-amine (144 mg, 100 %) which was used directly in the next step. LCMS
= 136.15 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.69 (d, J = 9.4 Hz, 1H), 7.39 (d, J = 9.4 Hz, 1H), 7.28 – 6.98 (m, 2H), 2.16 – 2.07 (m, 1H), 1.09 – 1.04 (m, 2H), 0.91 (d, J = 7.2 Hz, 2H). [0001343] Example 1.161. Synthesis of 5-methoxypyridazin-3-amine
[0001344] Step 1: tert-butyl (5-methoxypyridazin-3-yl)carbamate [0001345] To a solution of 3-chloro-5-methoxypyridazine (300 mg, 2.1 mmol) in 1,4-dioxane (6 mL) was added Pd(OAc)2 (47.2 mg, 0.21 mmol), Cs2CO3 (1.35 g, 4.17 mmol), NH2Boc (244 mg, 2.1 mmol) and Xantphos (120 mg, 0.21 mmol) and the reaction heated at 80 °C for 2 h. The
mixture was diluted with EtOAc (20 mL) and washed with water (10 mL × 2), the organic layer was dried over Na2SO4, filtered and concentrated. The residue obtained was purified by column chromatography on silica gel (eluent: Pet. ether : EtOAc = 10:1 to 3:1) to afford tert-butyl (5- methoxypyridazin-3-yl)carbamate (180 mg, 65 %) as a white solid.1HNMR (400 MHz, DMSO- d6) δ 10.33 (s, 1H), 8.64 (d, J = 2.6 Hz, 1H), 7.61 (d, J = 2.6 Hz, 1H), 3.89 (s, 3H), 1.49 (s, 9H). [0001346] Step 2: 5-methoxypyridazin-3-amine [0001347] To a solution of tert-butyl (5-methoxypyridazin-3-yl)carbamate (110 mg, 0.48 mmol) in a mixture of methanol (0.5 mL) and DCM (2 mL) was added a solution of HCl in dioxane (4 M in 1,4-dioxane, 2 mL) and the reaction stirred at room temperature for 3 h. The solvent was removed under vacuum to afford 5-methoxypyridazin-3-amine (70 mg, 80 %) as a white solid. LCMS m/z =126.03 [M+H]+. [0001348] Example 1.162. Synthesis of 5-cyclobutoxypyridazin-3-amine
[0001349] To a solution of 5-chloropyridazin-3-amine (100 mg, 0.77 mmol) in THF (2 mL) was added t-BuOK (433 mg, 3.86 mmol) and cyclobutanol (278 mg, 3.86 mmol) and the reaction heated at 80 °C overnight. The mixture was filtered and concentrated to afford 5- cyclobutoxypyridazin-3-amine (60 mg, 47 %) as a white solid. LCMS m/z = 166.15 [M+H]+; 1H NMR (400 MHz, Methanol-d4) δ 7.99 (d, J = 2.6 Hz, 1H), 5.99 (d, J = 2.6 Hz, 1H), 4.72 – 4.63 (m, 1H), 2.43 – 2.36 (m, 2H), 2.09 – 2.01 (m, 2H), 1.83 – 1.75 (m, 1H), 1.71 – 1.62 (m, 1H). [0001350] Example 1.163. Synthesis of 6-ethylpyridazin-3-amine
[0001351] Step 1: 6-vinylpyridazin-3-amine [0001352] To a solution of 6-bromopyridazin-3-amine (100 mg, 0.57 mmol) in a mixture of 1,4- dioxane (4 mL) and water (1 mL) was added 4,4,5,5-tetramethyl-2-vinyl-1,3,2-dioxaborolane (89 mg, 0.57 mmol), Pd(dppf)Cl2 (42 mg, 0.06 mmol) and K3PO4 (366 mg, 1.72 mmol) and the reaction heated at 110 °C for 3 h. The mixture was diluted with water (10 mL) and extracted with EtOAc (10 mL × 3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated to afford 6-vinylpyridazin-3-amine (63 mg, 90 %) as a black oil which
was used in the next step without further purification. LCMS m/z =122.15 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.57 – 7.54 (m, 1H), 6.84 – 6.78 (m, 1H), 6.77 – 6.72 (m, 1H), 6.44 (s, 2H), 5.90 (d, J = 17.8 Hz, 1H), 5.34 (d, J = 11.2 Hz, 1H). [0001353] Step 2: 6-ethylpyridazin-3-amine [0001354] To a solution of 6-vinylpyridazin-3-amine (60 mg, 0.50 mmol) in methanol (4 mL) was added 10% Pd/C (24 mg) and the reaction stirred at room temperature overnight under a hydrogen atmosphere. The catalyst was removed by filtration and the filtrate concentrated to afford 6-ethylpyridazin-3-amine (60 mg, quant.) as a black oil which was used without further purification. LCMS m/z =124.20 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.14 (d, J = 9.0 Hz, 1H), 6.71 (d, J = 9.0 Hz, 1H), 6.07 (s, 2H), 2.69 – 2.64 (m, 2H), 1.19 – 1.16 (m, 3H). [0001355] Example 1.164. Synthesis of 6-cyclobutoxy-4-methylpyridin-3-amine
[0001356] Step 1: 2-cyclobutoxy-4-methyl-5-nitropyridine [0001357] To a solution of 2-fluoro-4-methyl-5-nitropyridine (1.0 g, 6.41 mmol) in acetonitrile (10 mL) was added cyclobutanol (0.7 g, 9.61 mmol) and Cs2CO3 (4.2 g, 12.81 mmol) and the reaction stirred at room temperature for 20 h. The mixture was filtered and the filtrate concentrated to afford 2-cyclobutoxy-4-methyl-5-nitropyridine (1.26 g, 95 %) which was used in the next step without further purification. LCMS m/z = 209.1 [M+H]+; 1H NMR (400 MHz, CD3OD) δ 8.83 (s, 1H), 6.73 (s, 1H), 5.27 – 5.19 (m, 1H), 2.58 (s, 3H), 2.51 – 2.43 (m, 2H), 2.19 – 2.09 (m, 2H), 1.89 – 1.83 (m, 1H), 1.78 – 1.67 (m, 1H). [0001358] Step 2: 6-cyclobutoxy-4-methylpyridin-3-amine [0001359] To a solution of 2-cyclobutoxy-4-methyl-5-nitropyridine (500 mg, 1.68 mmol) in methanol (5 mL) was added 10% Pd/C (200 mg) and the reaction stirred for 24 h under a hydrogen atmosphere. The catalyst was removed by filtration and the filtrate concentrated to afford 6-cyclobutoxy-4-methylpyridin-3-amine (400 mg, 80 %) which was used in the next step without further purification. LCMS m/z = 179.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.43 (s, 1H), 6.39 (s, 1H), 4.98 – 4.90 (m, 1H), 4.48 (s, 2H), 2.34 – 2.27 (m, 2H), 2.04 (s, 3H), 1.98 – 1.91 (m, 2H), 1.77 – 1.67 (m, 1H), 1.62 – 1.52 (m, 1H). [0001360] Example 1.165. Synthesis of 6-isopropylpyridazin-3-amine
[0001361] Step 1: 6-(prop-1-en-2-yl)pyridazin-3-amine [0001362] To a solution of 6-bromopyridazin-3-amine (500 mg, 2.89 mmol) in a mixture of 1,4- dioxane (8 mL) and water (2 mL) was added 4,4,5,5-tetramethyl-2-(prop-1-en-2-yl)-1,3,2- dioxaborolane (729 mg, 4.34 mmol), cesium carbonate (2.82 mg, 8.67 mmol) and Pd(dppf)Cl2 (211 mg, 0.289 mmol) and the reaction was heated at 80 °C overnight. The mixture was diluted with water (50 mL) and extracted with EtOAc (50 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (eluent: Pet. ether: EtOAc = 3:1 to 1:1) to afford 6-(prop-1- en-2-yl)pyridazin-3-amine (320 mg, 82 %) as a white solid. LCMS m/z =136.12 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.61 (d, J = 9.2 Hz, 1H), 6.74 (d, J = 9.2 Hz, 1H), 6.39 (s, 2H), 5.60 (s, 1H), 5.24 – 5.16 (m, 1H), 2.14 (d, J = 1.2 Hz, 3H). [0001363] Step 2: 6-isopropylpyridazin-3-amine [0001364] To a solution of 6-(prop-1-en-2-yl)pyridazin-3-amine (100 mg, 0.74 mmol) in methanol (2 mL) was added 10% Pd/C (20 mg) and the reaction was stirred at room temperature for 2 h under a hydrogen atmosphere. The catalyst was removed by filtration and the filtrate concentrated to afford 6-isopropylpyridazin-3-amine (101 mg, 100 %) which was used directly in the next step without further purification.1H NMR (400 MHz, DMSO-d6) δ 7.18 (d, J = 9.0 Hz, 1H), 6.72 (d, J = 9.2 Hz, 1H), 6.08 (s, 2H), 3.06 – 2.93 (m, 1H), 1.19 (d, J = 7.0 Hz, 6H). [0001365] Example 1.166. Synthesis of 6-methoxypyridazin-3-amine
[0001366] A solution of 6-chloropyridazin-3-amine (300 mg, 2.32 mmol) in a solution of sodium methoxide in MeOH (5.4 M in methanol, 8 mL, 43.2 mmol) was heated at 90 °C for 72 h in a sealed tube. The mixture was diluted with water (20 mL) and extracted with EtOAc (10 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep-TLC (eluent: DMC/MeOH = 10/1) to
afford 6-methoxypyridazin-3-amine (160 mg, 55 %) as a yellow oil. LCMS m/z =126.15 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 6.93 – 6.78 (m, 2H), 5.89 (s, 2H), 3.84 (s, 3H). [0001367] Example 1.167. Synthesis of 5-cyclobutoxypyridin-2-amine
[0001368] Step 1: 5-cyclobutoxy-2-nitropyridine [0001369] To a solution of 6-nitropyridin-3-ol (300 mg, 2.14 mmol) in toluene (3 mL) at 0 °C was added cyclobutanol (232 mg, 3.21 mmol) and PPh3 (843 mg, 3.21 mmol) and the reaction stirred for 20 min. DIAD (0.6 mL, 3.21 mmol) was added and the reaction heated at 80 °C for 3 h. The mixture was diluted with water (30 mL) and extracted with EtOAc (10 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep-TLC (eluent: Pet. Ether: EtOAc = 5:1) to afford 5- cyclobutoxy-2-nitropyridine (110 mg, 26 % yield) as a yellow solid. LCMS m/z =195.10 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 8.30 (d, J = 9.0 Hz, 1H), 8.24 (d, J = 2.8 Hz, 1H), 7.60 (dd, J = 9.0, 2.8 Hz, 1H), 4.99 – 4.90 (m, 1H), 2.55 – 2.51 (m, 2H), 2.16 – 2.04 (m, 2H), 1.89 – 1.78 (m, 1H), 1.74 – 1.61 (m, 1H). [0001370] Step 2: 5-cyclobutoxypyridin-2-amine [0001371] To a solution of 5-cyclobutoxy-2-nitropyridine (110 mg, 0.57 mmol) in methanol (4 mL) was added 10 % Pd/C (35 mg) and the reaction was stirred at room temperature for 3 h under a hydrogen atmosphere. The catalyst was removed by filtration and the filtrate concentrated to afford 5-cyclobutoxypyridin-2-amine (90 mg, 96 %) as a yellow solid. LCMS m/z =165.15 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.53 (d, J = 3.0 Hz, 1H), 7.01 (dd, J = 8.8, 3.0 Hz, 1H), 6.39 (d, J = 8.8 Hz, 1H), 5.41 (s, 2H), 4.55 – 4.43 (m, 1H), 2.37 – 2.23 (m, 2H), 2.05 – 1.89 (m, 2H), 1.79 – 1.67 (m, 1H), 1.64 – 1.50 (m, 1H). [0001372] Example 1.168. Synthesis of 1-(oxetan-3-yl)-1H-pyrrolo[2,3-b]pyridin-5-amine
[0001373] Step 1: 5-nitro-1-(oxetan-3-yl)-1H-pyrrolo[2,3-b]pyridine
[0001374] To a solution of 5-nitro-1H-pyrrolo[2,3-b]pyridine (300 mg, 1.84 mmol) in DMF (6 mL) was added 3-iodooxetane (1.18 g, 6.44 mmol) and Cs2CO3 (1.2 g, 3.68 mmol) and the reaction was heated at 130 °C overnight. The mixture was diluted with water (60 mL) and extracted with EtOAc (20 mL × 2). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue obtained was purified by prep-TLC (eluent: Pet. Ether: EtOAc = 3:1) to afford 5-nitro-1-(oxetan-3-yl)-1H-pyrrolo[2,3-b]pyridine (260 mg, 64 %) as a yellow solid. LCMS m/z =220.05 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 9.12 (d, J = 2.6 Hz, 1H), 8.92 (d, J = 2.4 Hz, 1H), 8.24 (d, J = 3.6 Hz, 1H), 6.89 (d, J = 3.6 Hz, 1H), 6.09 – 5.97 (m, 1H), 5.07 – 4.96 (m, 4H). [0001375] Step 2: 1-(oxetan-3-yl)-1H-pyrrolo[2,3-b]pyridin-5-amine [0001376] To a solution of 5-nitro-1-(oxetan-3-yl)-1H-pyrrolo[2,3-b]pyridine (100 mg, 0.46 mmol) in methanol (4 mL) was added 10 % Pd/C (30 mg) and the reaction stirred at room temperature overnight under a hydrogen atmosphere. The catalyst was removed by filtration and the filtrate concentrated to afford 1-(oxetan-3-yl)-1H-pyrrolo[2,3-b]pyridin-5-amine (85 mg, 98 %) as a brown oil. LCMS m/z =190.05 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ 7.75 (d, J = 2.6 Hz, 1H), 7.69 (d, J = 3.6 Hz, 1H), 7.11 (d, J = 2.4 Hz, 1H), 6.29 (d, J = 3.6 Hz, 1H), 5.87 – 5.76 (m, 1H), 4.98 – 4.93 (m, 4H), 4.74 (s, 2H). [0001377] Example 2. Biological Assays [0001378] Example 2.1. CK1α Degradation Assay [0001379] Various compounds described herein were assessed for degradation of CK1α protein in vitro. Compounds were assessed using a HiBiT assay as described herein. In brief, an 11 amino acid HiBiT tag (VSGWRLFKKIS) was fused to the C terminus of CK1α by knocking in a construct encoding the HiBiT tag using specifically designed oligonucleotides for homology- directed repair (HDR), a targeted sgRNA, a SF Cell Line 4D Nucleofector X Kit S (Lonza), and a 4-D Nucleofector X Unit (Lonza) in accordance with the Lonza protocol. The assay was originally run with pooled HT-29 cells. Pool HT-29 cells were cloned out with the serial dilution method and single cell clones were identified, with one clone selected for assay purposes. Once a single clone was identified, the assay conditions were modified to use the single HT-29 clone instead of the pool. Depending on the assay, pooled or single clonal cells were plated at 5,000 cells/well in 40 ul of culture media in 384 well plates (Corning). Culture media comprised McCoys 5A medium (ATCC) and 10% Fetal Bovine Serum (FBS) (Clontech). Plates were
incubated for 24 hours at 37°C and 5% CO2 to allow cells to attach prior to compound treatment. After 24 hours, compounds were prepared as 10 mM stock solutions in 0.1% DMSO and dosed in duplicates with 10 uM top dose and a 9-point dose-response curve using an automated dispenser. Cells were incubated with compounds for 24 hours at 37°C and 5% CO2. After 24 hours, cells were removed from incubator and allowed to equilibrate to room temperature for 15 minutes. Nano-Glo HiBiT Lytic Reagent was prepared by adding LgBiT Protein and substrate to the buffer as described by manufacturer’s Nano-Glo HiBiT Lytic Detection System protocol (Promega). Reagent was added 1:1 to plate volume (40 ul) and plates were incubated at room temperature for 15 minutes while shaking. After 15 minutes, luminescence was read using GloMax Discover plate reader (Promega) according to manufacturer’s protocol. Resulting data was analyzed in CDD Vault software by normalizing to a DMSO control and using Levenberg- Marquardt algorithm to fit a hill equation to dose-response data. The concentration at which 50% CK1α degradation occurs (DC50) and the maximum percentage of CK1α degradation (Dmax) was calculated. Certain results are presented in the table below. [0001380] HiBiT Pool Results
[0001382] In the table above, for DC50, the symbol “++++” indicates a DC50 of ≤ 100 nM, the symbol “+++” indicates a DC50 of > 100 nM and ≤ 500 nM, the symbol “++” indicates a DC50 of > 500 nM and ≤ 5000 nM, and the symbol “+” indicates a DC50 of > 5000 nM. [0001383] As depicted in the tables above, various compounds can provide degradation of target proteins, e.g., CK1α proteins. In some embodiments, compounds can provide efficient
degradation of CK1α. In some embodiments, compounds can provide high levels of degradation of CK1α. In some embodiments, a compound, e.g., a compound of Table 1, can provide an increased level of degradation of CK1α as compared to a reference compound. In some embodiments, a compound, e.g., a compound of Table 1, can provide increased efficiency of degradation of CK1α as compared to a reference compound. In some embodiments, a reference compound is a compound of Table 2. [0001384] Example 2.2. Wnt Pathway Activation Assay [0001385] Various compounds described herein were assessed for Wnt pathway activation. Compounds were assessed using a TOPflash assay as described herein. As reported in the literature, TOPflash is a luciferase reporter plasmid that contains six tandem repeats of TCF binding elements. In brief, a HT-29 TOPflash stable cell line was generated by overexpression of a lentiviral construct, TCF/LEF Luciferase Report Lentivirus (blasticidin) (Vector Builder). A control cell line was also generated to assay whether any examined compounds interfered with the luciferase reporter or were cytotoxic. The control cell line was a HT-29 Luc2 stable cell line generated by overexpression of a lentiviral construct, Firefly Luciferase Lentivirus (blasticidin) (Vector Builder). Cells were selected with their respective antibiotics and pool cells were cloned out with the serial dilution method. Single cell clones were identified, with one selected for assay purposes. For the TOPflash/Luc2 assay, pool or clonal cells were plated 5,000 cells/well in 40 ul of culture media in 384 well plates or 10,000 cells/well in 100 ul of culture media in 96 well plates. Culture media comprised McCoys 5A medium (ATCC) and 10% FBS (Clontech). Plates were then incubated for 24 hours at 37°C and 5% CO2 to allow cells to attach prior to compound treatment. After 24 hours, compounds were prepared as 10 mM stocks in 0.1% DMSO and dosed in duplicate or triplicate with a 10 uM top dose and a 9-point dose-response curve. Cells were then incubated with compounds for 24 hours at 37°C and 5% CO2. After 24 hours, cells were removed from the incubator and allowed to equilibrate to room temperature for 15 minutes. To assay luciferase expression, either One-Lite Luciferase Assay System (Vazyme) or ONE-Glo Luciferase Assay System (Promega) was prepared according to manufacturer’s protocol. Prepared reagent was added 1:1 to wells of plates, and plates were placed on a shaker at room temperature for 15 minutes that was protected from light. Following reagent incubation period, luminescence was read using a plater reader according to the manufacturer’s protocol (using 0.3 integration time). Resulting data was analyzed by calculating % fold change
compared to DMSO control and by using the Levenberg-Marquardt algorithm to fit a hill equation to dose-response data. Certain results are presented in the table below.
[0001386] In the table above, for % Fold Change, the symbol “++++” indicates a fold change of ≥ 5, the symbol “+++” indicates a fold change of < 5 and ≥ 2, the symbol “++” indicates a fold change of < 2 and ≥ 1.5, and the symbol “+” indicates a fold change of < 1.5. In the table above, for EC50, the symbol “++++” indicates an EC50 of ≤ 1.5 uM, the symbol “+++” indicates an EC50 of > 1.5 uM and ≤ 4 uM, the symbol “++” indicates an EC50 of > 4 uM and ≤ 10, and the symbol “+” indicates an EC50 of > 10 uM. [0001387] As depicted in the table above, various compounds can provide activation of cellular pathways, e.g., Wnt pathway. In some embodiments, compounds can provide efficient Wnt pathway activation. In some embodiments, compounds can provide high levels of Wnt pathway activation. In some embodiments, a compound, e.g., a compound of Table 1, can provide an increased level of Wnt pathway activation as compared to a reference compound. In some embodiments, a compound, e.g., a compound of Table 1, can provide increased efficiency of
Wnt pathway activation as compared to a reference compound. In some embodiments, a reference compound is a compound of Table 2. [0001388] While various embodiments have been described and illustrated herein, those of ordinary skill in the art will readily envision a variety of other means and/or structures for performing the functions and/or obtaining the results and/or one or more of the advantages described in the present disclosure, and each of such variations and/or modifications is deemed to be included. More generally, those skilled in the art will readily appreciate that all parameters, dimensions, materials, and configurations described herein are meant to be example and that the actual parameters, dimensions, materials, and/or configurations may depend upon the specific application or applications for which the teachings of the present disclosure is/are used. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the embodiments of the present disclosure. It is, therefore, to be understood that the foregoing embodiments are presented by way of example only and that, within the scope of the appended claims and equivalents thereto, claimed technologies may be practiced otherwise than as specifically described and claimed. In addition, any combination of two or more features, systems, articles, materials, kits, and/or methods, if such features, systems, articles, materials, kits, and/or methods are not mutually inconsistent, is included within the scope of the present disclosure.
Claims
We claim: 1. A compound having the structure of formula I”:
or a pharmaceutically acceptable salt thereof, wherein: Ring A is selected from a 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, a 10-membered bicyclic aryl ring, a 3- to 7-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, an 8- to 10-membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 9- to 10-membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; each Rx is independently selected from oxo, halogen, -NO2, -(CH2)yR, –CN, –OR, -N(R)2, -SR, and -(CH2)zY; or: two instances of Rx, together with the atoms to which they are attached, form a 5- to 6- membered saturated, partially unsaturated or aromatic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; Y is selected from –OR, -N(R)2, -O(CH2)2OR, -C(O)R, -CO2R, -OC(O)R, -C(O)N(R)2, - N(R)C(O)R, -SO2R, -SO2N(R)2, and -N(R)SO2R; or: L is a C1-4 aliphatic chain wherein one or more methylene units of the aliphatic chain are optionally and independently replaced by a group selected from –N(Ry)-, -C(=O)-, -O-, - S(O)2-, and -CF2-; each Ry is selected from hydrogen, C1-3 alkyl and lower haloalkyl; Ring C is a bivalent group comprising
, wherein Ring C is substituted with 0-3 instances of Rx;
each R is independently hydrogen or an optionally substituted group selected from C1-6 aliphatic, a 3- to 7-membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, a 3- to 7-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5- to 6- membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; m is 0-3; y is 0-2; and z is 0-1.
6. The compound according to any one of claims 1-5, wherein Ring A is selected from:
7. The compound according to any one of claims 1-6, wherein the compound is not a compound set forth in Table 2.
8. The compound according to claim 1, wherein the compound is selected from those in Table 1, or a pharmaceutically acceptable salt thereof.
9. A pharmaceutical composition comprising a compound according to any one of claims 1-8, or a pharmaceutically acceptable salt thereof, to a patient in need thereof.
10. A method of treating a cancer, the method comprising administering a compound according to any one of claims 1-8, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 9.
11. The method according to claim 10, wherein the cancer is a CK1α-mediated cancer.
12. The method according to claim 11, wherein the cancer is a WNT-mediated cancer.
13. The method according to claim 11, wherein the cancer is a p53-mediated cancer.
14. A method of modulating an oncogenic pathway, the method comprising contacting a biological sample or administering to a patient in need thereof a compound according to any one of claims 1-8, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 9.
Applications Claiming Priority (6)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202363588628P | 2023-10-06 | 2023-10-06 | |
| US63/588,628 | 2023-10-06 | ||
| US202463637121P | 2024-04-22 | 2024-04-22 | |
| US63/637,121 | 2024-04-22 | ||
| US202463684009P | 2024-08-16 | 2024-08-16 | |
| US63/684,009 | 2024-08-16 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025076284A1 true WO2025076284A1 (en) | 2025-04-10 |
Family
ID=95283895
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2024/049873 Pending WO2025076284A1 (en) | 2023-10-06 | 2024-10-04 | Compounds, pharmaceutical compositions thereof, and methods of using the same |
Country Status (2)
| Country | Link |
|---|---|
| TW (1) | TW202530195A (en) |
| WO (1) | WO2025076284A1 (en) |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2025232888A1 (en) * | 2024-05-10 | 2025-11-13 | Shanghai Degron Biomedical Technology Co., Ltd. | Wee1 degraders and uses thereof |
Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US3929880A (en) * | 1971-08-21 | 1975-12-30 | Merck Patent Gmbh | Process for the preparation of oxocarboxylic acid amides and oxocarboxylic acids |
| WO2019038717A1 (en) * | 2017-08-23 | 2019-02-28 | Novartis Ag | 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof |
| US20190076542A1 (en) * | 2016-05-10 | 2019-03-14 | C4 Theraprutics, Inc. | C3-carbon linked glutarimide degronimers for target protein degradation |
| US11548870B2 (en) * | 2019-11-19 | 2023-01-10 | Bristol-Myers Squibb Company | Compounds useful as inhibitors of helios protein |
-
2024
- 2024-10-04 WO PCT/US2024/049873 patent/WO2025076284A1/en active Pending
- 2024-10-04 TW TW113137974A patent/TW202530195A/en unknown
Patent Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US3929880A (en) * | 1971-08-21 | 1975-12-30 | Merck Patent Gmbh | Process for the preparation of oxocarboxylic acid amides and oxocarboxylic acids |
| US20190076542A1 (en) * | 2016-05-10 | 2019-03-14 | C4 Theraprutics, Inc. | C3-carbon linked glutarimide degronimers for target protein degradation |
| WO2019038717A1 (en) * | 2017-08-23 | 2019-02-28 | Novartis Ag | 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof |
| US11548870B2 (en) * | 2019-11-19 | 2023-01-10 | Bristol-Myers Squibb Company | Compounds useful as inhibitors of helios protein |
Non-Patent Citations (2)
| Title |
|---|
| DATABASE PUBCHEM COMPOUND 20 October 2014 (2014-10-20), ANONYMOUS: "4-(Cyclohexylmethyl)piperidine-2,6-dione", XP093303093, Database accession no. 80401247 * |
| DATABASE PUBCHEM COMPOUND 23 October 2012 (2012-10-23), ANONYMOUS: "3-(Cyclopentylmethyl)piperidine-2,6-dione", XP093303099, Database accession no. 64159481 * |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2025232888A1 (en) * | 2024-05-10 | 2025-11-13 | Shanghai Degron Biomedical Technology Co., Ltd. | Wee1 degraders and uses thereof |
Also Published As
| Publication number | Publication date |
|---|---|
| TW202530195A (en) | 2025-08-01 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| AU2022221514B2 (en) | PYRAZOLO[3,4-b]PYRIDINE COMPOUNDS AS INHIBITORS OF TAM AND MET KINASES | |
| JP7079303B2 (en) | Substituted pyrazolo [1,5-A] pyridine compound as a RET kinase inhibitor | |
| CN114867726B (en) | Small molecule inhibitors of KRAS G12C mutants | |
| KR20240004960A (en) | RAS inhibitors | |
| KR20230067635A (en) | Indole derivatives as RAS inhibitors in the treatment of cancer | |
| US20230339902A1 (en) | Tricyclic ligands for degradation of ikzf2 or ikzf4 | |
| CN113498342A (en) | Compounds involved in synergistic binding and uses thereof | |
| CN118922423A (en) | RAS inhibitors | |
| CN120247911A (en) | PI3K-α inhibitors and methods of use thereof | |
| KR20200133747A (en) | N-(phenyl)-2-(phenyl)pyrimidine-4-carboxamide derivatives and related compounds as HPK1 inhibitors for treating cancer | |
| KR20220151160A (en) | SMARCA disintegrant and its uses | |
| TW201109327A (en) | Heterocyclic compound | |
| CA3161339A1 (en) | Cyclic compounds and methods of using same | |
| WO2025076284A1 (en) | Compounds, pharmaceutical compositions thereof, and methods of using the same | |
| CN112912142B (en) | 5-Azaindazole derivatives as adenosine receptor antagonists | |
| WO2025076285A1 (en) | Compounds, pharmaceutical compositions thereof, and methods of using the same | |
| US20240150365A1 (en) | Pyridinylsulfonamide compounds and their use in therapy | |
| WO2024044731A1 (en) | Diazepino-thieno-quinoxaline compounds and their use in therapy | |
| US20240034734A1 (en) | Tetracyclic compounds as dgk inhibitors | |
| TW202300150A (en) | Cyclic compounds and methods of using same | |
| TW202517247A (en) | Bicyclic dgk inhibitors | |
| WO2024229401A2 (en) | Diazepino-thieno-quinoxaline compounds and their use in therapy |
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: 24875420 Country of ref document: EP Kind code of ref document: A1 |